Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioorg Med Chem Lett ; 30(7): 126987, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32029324

RESUMEN

Pyrrolobenzodiazepines (PBDs) and their dimers (bis-PBDs) have emerged as some of the most potent chemotherapeutic compounds and are currently under development as novel payloads in antibody-drug conjugates (ADCs). However, when used as stand-alone therapeutics or as warheads for small molecule drug conjugates (SMDCs), dose-limiting toxicities are often observed. As an elegant solution to this inherent problem, we designed and synthesized a diazepine-ring-opened bis-PBD prodrug (pro-PBD-PBD) folate conjugate lacking the one of the two imine moieties found in the corresponding free bis-PBD. Upon entering a targeted cell, cleavage of the linker system, including the hydrolysis of an oxazolidine moiety, results in the formation of a reactive intermediate which possesses a newly formed aldehyde as well as an aromatic amine. A fast and spontaneous intramolecular ring-closing reaction subsequently takes place as the aromatic amine adds to the aldehyde with the loss of water to give the imine, and as a result, the diazepine ring, thereby delivering the bis-PBD to the targeted cell. The in vitro and in vivo activity of this conjugate has been evaluated on folate receptor positive KB cells. Sub-nanomolar activity with good specificity and high cure rates with minimal toxicity have been observed.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Benzodiazepinas/uso terapéutico , Receptores de Folato Anclados a GPI/metabolismo , Neoplasias/tratamiento farmacológico , Profármacos/uso terapéutico , Pirroles/uso terapéutico , Animales , Antibióticos Antineoplásicos/síntesis química , Antibióticos Antineoplásicos/farmacología , Benzodiazepinas/síntesis química , Benzodiazepinas/farmacología , Diseño de Fármacos , Femenino , Células HeLa , Humanos , Ratones Desnudos , Profármacos/síntesis química , Profármacos/farmacología , Pirroles/síntesis química , Pirroles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Bioconjug Chem ; 30(6): 1805-1813, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31075200

RESUMEN

Prostate-specific membrane antigen (PSMA) is a biomarker that is overexpressed on prostate cancer, and it is also present on the neovasculature within many non-prostate solid tumors. Herein, we report on the construction and biological testing of novel tubulysin B-containing therapeutic agents for the treatment of PSMA-expressing cancer. One of these compounds, EC1169, emerged as a lead candidate for preclinical development and phase 1 clinical testing. This water-soluble conjugate was shown to have high affinity for PSMA-positive cells. When tested in vitro, EC1169 was found to inhibit the growth of PSMA-positive cells, but it displayed no activity against PSMA-negative cells. Brief treatment of nude mice bearing PSMA-positive LNCaP human xenografts with EC1169 led to complete remissions and cures. Furthermore, this activity occurred in the absence of weight loss. In contrast, the nontargeted tubulysin B drug proved to be inactive against the LNCaP tumor model when administered at doses near to or greater than the maximum tolerated level. PSMA-negative KB tumors did not appreciably respond to EC1169 therapy, thereby confirming this compound's targeted specificity for PSMA-positive cells. Finally, treatment of LNCaP-bearing mice with docetaxel (the most active chemotherapeutic agent approved for late stage prostate cancer therapy) was found to produce only modest anti-tumor activity, and this outcome was also associated with severe weight loss. Taken together, these results strongly indicate that PSMA-positive tumors may be effectively treated using highly potent, PSMA-targeted small-molecule drug conjugates using regimens that do not cause undesirable side effects.


Asunto(s)
Antígenos de Superficie/análisis , Antineoplásicos/uso terapéutico , Glutamato Carboxipeptidasa II/análisis , Oligopéptidos/uso terapéutico , Ácidos Pipecólicos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/química , Línea Celular Tumoral , Humanos , Masculino , Ratones Desnudos , Oligopéptidos/química , Ácidos Pipecólicos/química , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Toxicol Appl Pharmacol ; 368: 49-54, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30794826

RESUMEN

INTRODUCTION: Nonalcoholic steatohepatitis (NASH) afflicts 20-36% of individuals with nonalcoholic fatty liver disease (NAFLD). A lipotoxic hepatic environment, altered innate immune signaling and inflammation are defining features of progression to NASH. Activated resident liver macrophages express folate receptor beta (FR-ß) which may be an indicator of progression from steatosis to NASH. The goals of this study were to characterize FR-ß protein expression in human NAFLD and rodent models of NASH, and demonstrate liver targeting of an FR-ß imaging agent to the liver of a rodent NASH model using FR-ß. METHODS: Rat liver lysates from methionine choline deficient (MCD) fed rats, high fat diet (HFD) and methionine choline sufficient (MC+) rat controls were analyzed for hepatic FR-ß protein. The FR-ß-targeted agent, Etarfolatide was injected into MCD and MC + -fed C57BL/6 mice for efficient FastSPECT hepatic imaging. Additionally, FR-ß expression across the stages of human NAFLD from normal to NASH was assessed. RESULTS: FastSPECT images show targeting of Etarfolatide to the liver of mice fed 8 weeks of MCD diet but not control-fed mice. The MCD rat model exhibited significantly increased protein expression of hepatic FR-ß in contrast to HFD or normal samples. Similarly human liver samples categorized as NASH Fatty or NASH Not Fatty showed elevated FR-ß protein when compared to normal liver. FR-ß transcript expression levels were elevated across both NASH Fatty and NASH Not Fatty samples. CONCLUSION: The findings in this study indicate that FR-ß expression in NASH may be harnessed to target agents directly to the liver.


Asunto(s)
Receptor 2 de Folato/metabolismo , Hígado/diagnóstico por imagen , Hígado/metabolismo , Macrófagos/metabolismo , Imagen Molecular/métodos , Enfermedad del Hígado Graso no Alcohólico/diagnóstico por imagen , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Tomografía Computarizada de Emisión de Fotón Único , Animales , Biomarcadores/metabolismo , Deficiencia de Colina/complicaciones , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Receptor 2 de Folato/genética , Ácido Fólico/administración & dosificación , Ácido Fólico/análogos & derivados , Humanos , Masculino , Metionina/deficiencia , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Compuestos de Organotecnecio/administración & dosificación , Valor Predictivo de las Pruebas , Radiofármacos/administración & dosificación , Ratas Sprague-Dawley
4.
Mol Pharm ; 16(9): 3985-3995, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31356752

RESUMEN

Folate-based small molecule drug conjugates (SMDCs) are currently under development and have shown promising preclinical and clinical results against various cancers and polycystic kidney disease. Two requisites for response to a folate-based SMDC are (i) folate receptor alpha (FRα) protein is expressed in the diseased tissues, and (ii) FRα in those tissues is accessible and functionally competent to bind systemically administered SMDCs. Here we report on the development of a small molecule reporter conjugate (SMRC), called EC2220, which is composed of a folate ligand for FRα binding, a multilysine containing linker that can cross-link to FRα in the presence of formaldehyde fixation, and a small hapten (fluorescein) used for immunohistochemical detection. Data show that EC2220 produces a far greater IHC signal in FRα-positive tissues over that produced with EC17, a folate-fluorescein SMRC that is released from the formaldehyde-denatured FRα protein. Furthermore, the extent of the EC2220 IHC signal was proportional to the level of FRα expression. This EC2220-based assay was qualified both in vitro and in vivo using normal tissue, cancer tissue, and polycystic kidneys. Overall, EC2220 is a sensitive and effective reagent for evaluating functional and accessible receptor expression in vitro and in vivo.


Asunto(s)
Receptor 1 de Folato/metabolismo , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Células A549 , Animales , Doxiciclina/farmacología , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Receptor 1 de Folato/análisis , Ácido Fólico/análogos & derivados , Ácido Fólico/química , Ácido Fólico/metabolismo , Células HeLa , Humanos , Lisina/análogos & derivados , Lisina/química , Lisina/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Neoplasias/metabolismo , Oligopéptidos/química , Oligopéptidos/metabolismo , Enfermedades Renales Poliquísticas/inducido químicamente , Enfermedades Renales Poliquísticas/metabolismo , Proteína Quinasa C/genética , Distribución Tisular , Compuestos de Tritilo/química , Compuestos de Tritilo/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Am J Physiol Renal Physiol ; 315(2): F395-F405, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717938

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is a very common genetic disease leading to renal failure. Numerous aberrantly regulated signaling pathways have been identified as promising molecular drug targets for ADPKD therapy. In rodent models, many small-molecule drugs against such targets have proven effective in reducing renal cyst growth. For example, mammalian target of rapamycin (mTOR) inhibition with rapamycin greatly ameliorates renal cystic disease in several rodent models. However, clinical trials with mTOR inhibitors were disappointing largely due to the intolerable extrarenal side effects during long-term treatment with these drugs. Most other potential drug targets in ADPKD are also widely expressed in extrarenal tissues, which makes it likely that untargeted therapies with small-molecule inhibitors against such targets will lead to systemic adverse effects during the necessary long-term treatment of years and decades in ADPKD patients. To overcome this problem, we previously demonstrated that folate-conjugated rapamycin (FC-rapa) targets polycystic kidneys due to the high expression of the folate receptor (FRα) and that treatment of a nonortholgous PKD mouse model leads to inhibition of renal cyst growth. Here we show, in a head-to-head comparison with unconjugated rapamycin, that FCrapa inhibits renal cyst growth, mTOR activation, cell cycling, and fibrosis in an orthologous Pkd1 mouse model. Both unconjugated rapamycin and FC-rapa are similarly effective on polycystic kidneys in this model. However, FC-rapa lacks the extrarenal effects of unconjugated rapamycin, in particular immunosuppressive effects. We conclude that folate-conjugation is a promising avenue for increasing the tissue specificity of small-molecule compounds to facilitate very long-term treatment in ADPKD.


Asunto(s)
Ácido Fólico/farmacología , Riñón/efectos de los fármacos , Riñón Poliquístico Autosómico Dominante/prevención & control , Inhibidores de Proteínas Quinasas/farmacología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Células A549 , Animales , Modelos Animales de Enfermedad , Composición de Medicamentos , Receptor 1 de Folato/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Humanos , Integrasas/genética , Riñón/enzimología , Ratones Noqueados , Riñón Poliquístico Autosómico Dominante/enzimología , Riñón Poliquístico Autosómico Dominante/genética , Inhibidores de Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/análogos & derivados , Sirolimus/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Canales Catiónicos TRPP/deficiencia , Canales Catiónicos TRPP/genética , Distribución Tisular
6.
Bioconjug Chem ; 29(10): 3320-3331, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30185025

RESUMEN

Use of tumor-targeted fluorescence dyes to help surgeons identify otherwise undetected tumor nodules, decrease the incidence of cancer-positive margins, and facilitate localization of malignant lymph nodes has demonstrated considerable promise for improving cancer debulking surgery. Unfortunately, the repertoire of available tumor-targeted fluorescent dyes does not permit identification of all cancer types, raising the need to develop additional tumor-specific fluorescent dyes to ensure localization of all malignant lesions during cancer surgeries. By comparing the mRNA levels of the hypoxia-induced plasma membrane protein carbonic anhydrase IX (CA IX) in 13 major human cancers with the same mRNA levels in corresponding normal tissues, we document that CA IX constitutes a nearly universal marker for the design of tumor-targeted fluorescent dyes. Motivated by this expression profile, we synthesize two new CA IX-targeted near-infrared (NIR) fluorescent imaging agents and characterize their physical and biological properties both in vitro and in vivo. We report that conjugation of either acetazolamide or 6-aminosaccharin (i.e., two CA-IX-specific ligands) to the NIR fluorescent dye, S0456, via an extended phenolic spacer creates a brightly fluorescent dye that binds CA IX with high affinity and allows rapid visualization of hypoxic regions of solid tumors at depths >1 cm beneath a tissue surface. Taken together, these data suggest that a CA IX-targeted NIR dye can constitute a useful addition to a cocktail of tumor-targeted NIR dyes designed to image all human cancers.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Anhidrasa Carbónica IX/metabolismo , Hipoxia de la Célula , Colorantes Fluorescentes/química , Neoplasias/enzimología , Animales , Antígenos de Neoplasias/genética , Anhidrasa Carbónica IX/genética , Línea Celular Tumoral , Xenoinjertos , Humanos , Rayos Infrarrojos , Ratones , Ratones Desnudos , Modelos Moleculares
7.
Bioconjug Chem ; 28(12): 2921-2931, 2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29211454

RESUMEN

Pyrrolobenzodiazepines (PBDs) and their dimers (bis-PBDs) have emerged as some of the most potent chemotherapeutic compounds, and are currently under development as novel payloads in antibody-drug conjugates (ADCs). However, when used as stand-alone therapeutics or as warheads for small molecule drug conjugates (SMDCs), dose-limiting toxicities are often observed. As an elegant solution to this inherent problem, we designed diazepine-ring-opened conjugated prodrugs lacking the imine moiety. Once the prodrug (pro-PBD) conjugate enters a targeted cell, cleavage of the linker system triggers the generation of a reactive intermediate possessing an aldehyde and aromatic amine. An intramolecular ring-closing reaction subsequently takes place as the aromatic amine adds to the aldehyde with the loss of water to give the imine and, as a result, the diazepine ring. In our pro-PBDs, we mask the aldehyde as a hydrolytically sensitive oxazolidine moiety which in turn is a part of a reductively labile self-immolative linker system. To prove the range of applications for this new class of latent DNA-alkylators, we designed and synthesized several novel latent warheads: pro-PBD dimers and hybrids of pro-PBD with other sequence-selective DNA minor groove binders. Preliminary preclinical pharmacology studies showed excellent biological activity and specificity.


Asunto(s)
Benzodiazepinas/química , Benzodiazepinas/metabolismo , Diseño de Fármacos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Profármacos/síntesis química , Profármacos/metabolismo , Pirroles/química , Pirroles/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Benzodiazepinas/farmacología , Benzodiazepinas/uso terapéutico , Técnicas de Química Sintética , Humanos , Células KB , Neoplasias/patología , Profármacos/química , Pirroles/farmacología , Pirroles/uso terapéutico
8.
Mol Med ; 21: 584-96, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26181632

RESUMEN

Folate receptor (FR)-ß has been identified as a promising target for antimacrophage and antiinflammatory therapies. In the present study, we investigated EC0565, a folic acid-derivative of everolimus, as a FR-specific inhibitor of the mammalian target of rapamycin (mTOR). Because of its amphiphilic nature, EC0565 was first evaluated for water solubility, critical micelle formation, stability in culture and FR-binding specificity. Using FR-expressing macrophages, the effect of EC0565 on mTOR signaling and cellular proliferation was studied. The pharmacokinetics, metabolism and bioavailability of EC0565 were studied in normal rats. The in vivo activity of EC0565 was assessed in rats with adjuvant arthritis, a "macrophage-rich" model with close resemblance to rheumatoid arthritis. EC0565 forms micellar aggregates in physiological buffers and demonstrates good water solubility as well as strong multivalent FR-binding capacity. EC0565 inhibited mTOR signaling in rat macrophages at nanomolar concentrations and induced G0/G1 cell cycle arrest in serum-starved RAW264.7 cells. Subcutaneously administered EC0565 in rats displayed good bioavailability and a relatively long half-life (~12 h). When given at 250 nmol/kg, EC0565 selectively inhibited proliferating cell nuclear antigen expression in thioglycollate-stimulated rat peritoneal cells. With limited dosing regimens, the antiarthritic activity of EC0565 was found superior to that of etanercept, everolimus and a nontargeted everolimus analog. The in vivo activity of EC0565 was also comparable to that of a folate-targeted aminopterin. Folate-targeted mTOR inhibition may be an effective way of suppressing activated macrophages in sites of inflammation, especially in nutrient-deprived conditions, such as in the arthritic joints. Further investigation and improvement upon the physical and biochemical properties of EC0565 are warranted.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Everolimus/análogos & derivados , Everolimus/administración & dosificación , Ácido Fólico/análogos & derivados , Ácido Fólico/administración & dosificación , Inflamación/tratamiento farmacológico , Serina-Treonina Quinasas TOR/genética , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/química , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/genética , Artritis Experimental/patología , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Proliferación Celular/efectos de los fármacos , Everolimus/química , Receptor 2 de Folato/genética , Receptor 2 de Folato/metabolismo , Ácido Fólico/química , Humanos , Inflamación/genética , Inflamación/patología , Ratas , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
9.
Drug Resist Updat ; 17(4-6): 89-95, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25457975

RESUMEN

Conventional cancer treatment modalities have several limitations including lack of sufficient efficacy, serious untoward toxicity, as well as innate and acquired drug resistance. In contrast, targeted imaging agents can identify patients with receptors overexpressed on the surface of cancer cells, thus allowing appropriate selection of patients for personalized treatment with a desirable targeted therapeutic. The folate receptor (FR) has been identified as a new molecularly targeted entity, which is highly overexpressed on the surface of a spectrum of solid tumor cells, including ovarian, kidney, lung, brain, endometrial, colorectal, pancreatic, gastric, prostate, testicular, bladder, head and neck, breast, and non-small cell lung cancer. Folic acid conjugation is a novel approach for targeting FR-expressing tissues for personalized treatment. With the development of FRα-targeted therapies comes a concomitant prerequisite for reliable methods for the quantification of FRα tissue expression. Therefore, attaching a radioactive probe to folic acid to target diseased tissue has become a novel and powerful imaging technique. Currently available diagnostic tools frequently require invasive surgical biopsy. In contrast, the noninvasive single-photon emission computed tomography-based companion imaging agent, (99m)Tc-etarfolatide ((99m)Tc-EC20), is in development for use as a companion diagnostic with the FRα-targeted folate conjugate, vintafolide (EC145), to identify patients whose tumors express FRα. Vintafolide is a folic acid conjugate of Vinca alkaloid (desacetylvinblastine hydrazide) that targets FRα-expressing tumors, thereby disrupting microtubule polymerization. (99m)Tc-etarfolatide is taken up by FR-positive tumors and allows for noninvasive, whole-body monitoring of FRα expression status throughout treatment. The combination of vintafolide plus etarfolatide has been evaluated in three Phase 2 studies for the treatment of various solid tumors, including ovarian, endometrial, peritoneal, and platinum-resistant ovarian cancer, as well as lung cancer. Patients with FR-positive tumors, as identified by etarfolatide uptake, have had better clinical outcomes than patients with FR-negative tumors, indicating the potential of etarfolatide as a companion biomarker for predicting vintafolide response. Targeted therapies combined with a reliable companion diagnostic test represent a novel approach toward efficient personalized medicine for malignant and nonmalignant disorders. Furthermore, the recent availability of the crystal structures of FRα and FRß in complex with folates and antifolates forms a realistic basis for the rational design and implementation of novel FR-targeted drugs for the treatment of cancer and inflammatory disorders.


Asunto(s)
Antagonistas del Ácido Fólico/farmacología , Antagonistas del Ácido Fólico/uso terapéutico , Transportadores de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Neoplasias/tratamiento farmacológico , Animales , Humanos , Terapia Molecular Dirigida , Medicina de Precisión/métodos
10.
Clin Immunol ; 150(1): 64-77, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24333534

RESUMEN

EC0746 is a rationally designed anti-inflammatory drug conjugate consisting of a modified folic acid-based ligand linked to a γ-hydrazide analog of aminopterin. In this report, EC0746's effectiveness was evaluated against experimental retinal S-antigen (PDSAg) induced autoimmune uveitis (EAU) and myelin-basic-protein induced autoimmune encephalomyelitis (EAE). In both models, functional FR-ß was detected on activated macrophages in local (retinal or central-nervous-system, respectively) and systemic (peritoneal cavity) sites of inflammation. In myelin-rich regions of EAE rats, an increased uptake of (99m)Tc-EC20 (etarfolatide; a FR-specific radioimaging agent) was also observed. EC0746 treatment at disease onset suppressed the clinical severity of both EAU and EAE, and it strongly attenuated progressive histopathological changes in the affected organs. In all parameters assessed, EC0746 activity was completely blocked by a benign folate competitor, suggesting that these therapeutic outcomes were specifically FR-ß mediated. EC0746 may emerge as a useful macrophage-modulating agent for treating inflammatory episodes of organ-specific autoimmunity.


Asunto(s)
Aminopterina/uso terapéutico , Antiinflamatorios/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Antagonistas del Ácido Fólico/uso terapéutico , Uveítis/tratamiento farmacológico , Aminopterina/farmacología , Animales , Antiinflamatorios/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Receptor 2 de Folato/inmunología , Antagonistas del Ácido Fólico/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratas , Ratas Endogámicas Lew , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Uveítis/inmunología , Uveítis/patología
11.
Bioconjug Chem ; 25(3): 560-8, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24564229

RESUMEN

Vintafolide is a potent folate-targeted vinca alkaloid small molecule drug conjugate (SMDC) that has shown promising results in multiple clinical oncology studies. Structurally, vintafolide consists of 4 essential modules: (1) folic acid, (2) a hydrophilic peptide spacer, (3) a disulfide-containing, self-immolative linker, and (4) the cytotoxic drug, desacetylvinblastine hydrazide (DAVLBH). Here, we report a structure-activity study evaluating the biological impact of (i) substituting DAVLBH within the vintafolide molecule with other vinca alkaloid analogues such as vincristine, vindesine, vinflunine, or vinorelbine; (ii) substituting the naturally (S)-configured Asp-Arg-Asp-Asp-Cys peptide with alternative hydrophilic spacers of varied composition; and (iii) varying the composition of the linker module. A series of vinca alkaloid-containing SMDCs were synthesized and purified by HPLC and LCMS. The SMDCs were screened in vitro against folate receptor (FR)-positive cells, and anti-tumor activity was tested against well-established subcutaneous FR-positive tumor xenografts. The cytotoxic and anti-tumor activity was directly compared to that produced by vintafolide. Among all the folate vinca alkaloid SMDCs tested, DAVLBH-containing SMDCs were active, while those constructed with vincristine, vindesine, or vinorelbine analogues failed to produce meaningful biological activity. Within the DAVLBH series, having a bioreleasable, self-immolative linker system was found to be critical for activity since multiple analogues constructed with thioether-based linkers all failed to produce meaningful activity both in vitro and in vivo. Substitutions of some or all of the natural amino acids within vintafolide's hydrophilic spacer module did not significantly change the in vitro or in vivo potency of the SMDCs. Vintafolide remains one of the most potent folate-vinca alkaloid SMDCs produced to date, and continued clinical development is warranted.


Asunto(s)
Antineoplásicos/farmacología , Ácido Fólico/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Alcaloides de la Vinca/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Ácido Fólico/química , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Conformación Molecular , Neoplasias Experimentales/patología , Estereoisomerismo , Relación Estructura-Actividad , Alcaloides de la Vinca/química
12.
Mol Pharm ; 10(3): 967-74, 2013 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-23394590

RESUMEN

Folate receptor (FR) targeting is an attractive strategy for nuclear imaging of cancer and activated macrophages through application of folic acid radioconjugates. However, significant renal accumulation of folate radioconjugates and hence exceedingly high emission of radiation from the kidneys may mask uptake of radioactivity at sites of interest such as small metastases in the abdominal region of animal models of ovarian cancer. Recently it was observed that the antifolate pemetrexed (PMX) reduces undesired renal uptake of radiofolates. A disadvantage of this strategy is the fact that pemetrexed is a chemotherapeutic agent which may have toxic side effects. The aims of this study were therefore to investigate whether the desired effect of PMX to reduce renal accumulation of folate radioconjugates would be maintained if it was applied as a cocktail together with its antidote, thymidine, and to explore whether thymidine was an effective rescue agent against PMX's related toxicity in vitro and in vivo. In vitro internalization of (67)Ga-EC0800 was investigated using FR-positive KB tumor cells and embryonic monkey MA104 kidney cells in the absence and presence of PMX alone and in combination with thymidine. Uptake of (67)Ga-EC0800 into KB cells was increased by coincubation of the cells with PMX. In contrast uptake of (67)Ga-EC0800 into MA104 cells was reduced under the same conditions. In both cell lines coincubation of thymidine did not change the results obtained with PMX. Biodistribution and SPECT/CT imaging studies of (67)Ga-EC0800 were performed with KB tumor bearing mice injected with PMX alone or with a cocktail of PMX and thymidine. The radiofolate's kidney uptake reducing effect of PMX in mice was maintained also if PMX was employed together with its antidote thymidine. The tumor uptake of (67)Ga-EC0800 remained unchanged independent of the administration of PMX or a combination of PMX and thymidine. The effect of thymidine to abrogate PMX-induced cytotoxicity was demonstrated in vitro with an MTT assay using KB and MA104 cells. Cell viability was reduced to 50% (KB cells) and 70% (MA104 cells) of untreated controls if PMX (5 µM and 15 µM, respectively) was coincubated. Addition of thymidine (10 µM or 100 µM) compensated PMX's toxic effects in a dose-dependent manner. The effect of thymidine was also investigated in non-tumor bearing mice treated with high-dosed PMX. Rescue of mice with side effects after the third and fourth cycles of PMX application (1 mg/mouse) was achieved by application of thymidine (20 mg/mouse) at five consecutive days starting the day of PMX injection. Application of PMX together with thymidine as a cocktail is effective to improve the tissue distribution of radiofolates while preventing pharmacological and potentially toxic side effects of the chemotherapeutic agent PMX. These findings open new perspectives for folate-based nuclear imaging in preclinical research potentially allowing longitudinal investigations and monitoring therapies in animal models of cancer and inflammatory diseases.


Asunto(s)
Diagnóstico por Imagen/métodos , Antagonistas del Ácido Fólico , Animales , Femenino , Glutamatos , Guanina/análogos & derivados , Humanos , Hipoxantina , Células KB , Ratones , Pemetrexed , Radiofármacos , Timidina , Tomografía Computarizada de Emisión de Fotón Único
13.
J Am Soc Nephrol ; 23(10): 1674-81, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22859856

RESUMEN

Activation of the mammalian target of rapamycin (mTOR) signaling pathway is aberrant in autosomal-dominant polycystic kidney disease (ADPKD). The mTOR inhibitors, such as rapamycin, ameliorate PKD in rodent models, but clinical trials have not shown benefit, possibly as a result of low tissue concentrations of rapamycin at clinically tolerable doses. To overcome this limitation, we synthesized a folate-conjugated form of rapamycin (FC-rapa) that is taken up by folate receptor-mediated endocytosis and cleaved intracellularly to reconstitute the active drug. We found that renal cyst-lining cells highly express the folate receptor in ADPKD and mouse models. In vitro, FC-rapa inhibited mTOR activity in a dose- and folate receptor-dependent manner. Treatment of a PKD mouse model with FC-rapa inhibited mTOR in the target tissue, strongly attenuated proliferation and growth of renal cysts and preserved renal function. Furthermore, FC-rapa inhibited mTOR activity in the kidney but not in other organs. In summary, these results suggest that targeting the kidney using FC-rapa may overcome the significant side effects and lack of renal efficacy observed in clinical trials with mTOR inhibitors in ADPKD.


Asunto(s)
Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Sirolimus/uso terapéutico , Animales , Línea Celular , Modelos Animales de Enfermedad , Endocitosis , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/uso terapéutico , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Ratones , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Transducción de Señal/efectos de los fármacos , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/metabolismo
14.
Bioconjug Chem ; 23(7): 1357-69, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22667324

RESUMEN

The folate receptor (FR) is a potentially useful biological target for the management of many human cancers. This membrane protein binds extracellular folates with very high affinity and, through an endocytic process, physically delivers them inside the cell for biological consumption. There are now many examples of how this physiological system can be exploited for the targeted delivery of biologically active molecules to cancer. In fact, strong preclinical as well as emerging clinical evidence exists showing how FR-positive cancers can be (i) anatomically identified using folate conjugates of radiodiagnostic imaging agents and (ii) effectively treated with companion folate-targeted chemotherapies. While the biological results are compelling, it is of equal importance to understand the conjugation chemistries that were developed to produce these active molecules. Therefore, this review will focus on the methods utilized to construct folate-based small-molecule drug conjugates (SMDCs), with particular attention focused on modular design, hydrophilic spacers, and self-immolative linkers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Sistemas de Liberación de Medicamentos , Ácido Fólico/química , Neoplasias/tratamiento farmacológico , Ingeniería de Proteínas , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/metabolismo , Ácido Fólico/farmacocinética , Humanos , Modelos Biológicos , Neoplasias/metabolismo
15.
J Pharmacol Exp Ther ; 336(2): 336-43, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20978169

RESUMEN

During a phase I trial of EC145 (a folate-targeted vinca alkaloid conjugate), constipation was identified as the dose-limiting toxicity, probably from a nonfolate receptor-related liver clearance process capable of releasing unconjugated vinca alkaloid from EC145 and shuttling it to the bile. Here, we report on the selective placement of novel carbohydrate segments (1-amino-1-deoxy-glucitolyl-γ-glutamate) spaced in-between the folate and vinca alkaloid moieties of EC145, which yielded a new agent (EC0489) that is equipotent but less toxic than EC145. Whereas both compounds could cure tumor-bearing mice reproducibly, EC0489 differed from EC145 with i) a shorter elimination half-life, ii) approximately 70% decrease in bile clearance, iii) a 4-fold increase in urinary excretion, and iv) improved tolerability in rodents. This combination of improvements justified the clinical evaluation of EC0489 where currently administered dose levels have exceeded the maximal tolerated dose of EC145 by approximately 70%, thereby reflecting the translational benefits to this new approach.


Asunto(s)
Antineoplásicos/farmacocinética , Ácido Fólico/análogos & derivados , Hígado/metabolismo , Alcaloides de la Vinca/farmacocinética , Animales , Antineoplásicos/toxicidad , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Femenino , Receptores de Folato Anclados a GPI/fisiología , Ácido Fólico/farmacocinética , Ácido Fólico/toxicidad , Riñón/metabolismo , Masculino , Tasa de Depuración Metabólica , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Alcaloides de la Vinca/toxicidad
16.
Bioorg Med Chem Lett ; 21(22): 6778-81, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21974954

RESUMEN

Tubuylsins are extremely potent cytotoxic agents which inhibit tubulin polymerization and lead to cell cycle arrest and apoptosis. Tubulysins have been isolated from fermentation mixtures and have been chemically synthesized; however, these efforts have been hampered by poor yields and arduous purifications. In contrast, treatment of a mixture of natural tubulysins A, B, C, G, and I, obtained from a fermentation batch with trifluoroacetic acid results in the formation of a single N-acyliminium ion. Subsequent addition of butyric, isopentyl, or acetic acid results in the formation of tubulysin B, A, or I, respectively, as a single species. New tubulysin analogs can be formed upon treatment of the acyliminium ion with other nucleophiles such as alcohols, thiols, and nitriles, resulting in corresponding N-acyl-N,O-acetals, N-acyl-N,S-thioacetals, and N,N'-diacyl-aminals. Carbon-carbon bond formation is also possible with a modification of this protocol. The cytotoxicies of the natural tubulysins and tubulysin analogs synthesized by this method were evaluated on KB cells.


Asunto(s)
Productos Biológicos/síntesis química , Técnicas de Química Sintética/métodos , Myxococcales/química , Oligopéptidos/síntesis química , Ácidos Pipecólicos/síntesis química , Moduladores de Tubulina/síntesis química , Ácido Acético/química , Productos Biológicos/química , Técnicas de Química Sintética/economía , Iminas/química , Iones/química , Oligopéptidos/química , Ácidos Pipecólicos/química , Moduladores de Tubulina/química
17.
Bioorg Med Chem Lett ; 21(4): 1202-5, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21236665

RESUMEN

Efficient syntheses of folate receptor (FR) targeting conjugates of the anti-inflammatory, aminopterin hydrazide, are described. 2-{4-Benzoylamino}-5-oxo-5-{N'-[2-(pyridin-2-yldisulfanyl)-ethoxycarbonyl]-hydrazino}-pentanoic acid is synthesized from commercially available 4-[(2-amino-4-imino-3,4-dihydro-pteridin-6-yl-methyl)-amino]-benzoic acid. Conjugation of this novel, activated aminopterin hydrazide to folic acid through cysteine-terminating (C-terminus), peptide/carbohydrate spacers results in highly water soluble conjugates which allow for the release of free aminopterin hydrazide within the endosomes of targeted cells.


Asunto(s)
Aminopterina/química , Antiinflamatorios/química , Ácido Fólico/análogos & derivados , Aminopterina/síntesis química , Aminopterina/uso terapéutico , Antiinflamatorios/síntesis química , Antiinflamatorios/uso terapéutico , Diseño de Fármacos , Ácido Fólico/síntesis química , Ácido Fólico/química , Ácido Fólico/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Estereoisomerismo
18.
Cells ; 10(8)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34440885

RESUMEN

Since activated macrophages express a functional folate receptor ß (FRß), targeting this macrophage population with folate-linked drugs could increase selectivity to treat inflammatory diseases. Using a macrophage-mediated anti-glomerular basement membrane (anti-GBM) glomerulonephritis (GN) in WKY rats, we investigated the effect of a novel folic acid-aminopterin (AMT) conjugate (EC2319) designed to intracellularly deliver AMT via the FR. We found that treatment with EC2319 significantly attenuated kidney injury and preserved renal function. Kidney protection with EC2319 was blocked by a folate competitor, indicating that its mechanism of action was specifically FRß-mediated. Notably, treatment with methotrexate (MTX), another folic acid antagonist related to AMT, did not protect from kidney damage. EC2319 reduced glomerular and interstitial macrophage infiltration and decreased M1 macrophage recruitment but not M2 macrophages. The expression of CCL2 and the pro-fibrotic cytokine TGF-ß were also reduced in nephritic glomeruli with EC2319 treatment. In EC2319-treated rats, there was a significant decrease in the deposition of collagens. In nephritic kidneys, FRß was expressed on periglomerular macrophages and macrophages present in the crescents, but its expression was not observed in normal kidneys. These data indicate that selectively targeting the activated macrophage population could represent a novel means for treating anti-GBM GN and other acute crescentic glomerulonephritis.


Asunto(s)
Receptor 2 de Folato/metabolismo , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Macrófagos/metabolismo , Aminopterina/química , Aminopterina/uso terapéutico , Animales , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Ácido Fólico/química , Ácido Fólico/uso terapéutico , Macrófagos/efectos de los fármacos , Metotrexato/uso terapéutico , Ratas
19.
Cell Rep Med ; 2(10): 100422, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34755134

RESUMEN

Provoked by sterile/nonsterile insults, prolonged monocyte mobilization and uncontrolled monocyte/macrophage activation can pose imminent or impending harm to the affected organs. Curiously, folate receptor beta (FRß), with subnanomolar affinity for the vitamin folic acid (FA), is upregulated during immune activation in hematopoietic cells of the myeloid lineage. This phenomenon has inspired a strong interest in exploring FRß-directed diagnostics/therapeutics. Previously, we have reported that FA-targeted aminopterin (AMT) therapy can modulate macrophage function and effectively treat animal models of inflammation. Our current investigation of a lead compound (EC2319) leads to discovery of a highly FR-specific mechanism of action independent of the root causes against inflammatory monocytes. We further show that EC2319 suppresses interleukin-6/interleukin-1ß release by FRß+ monocytes in a triple co-culture leukemic model of cytokine release syndrome with anti-CD19 chimeric antigen receptor T cells. Because of its chemical stability and metabolically activated linker, EC2319 demonstrates favorable pharmacokinetic characteristics and cross-species translatability to support future pre-clinical and clinical development.


Asunto(s)
Aminopterina/farmacología , Síndrome de Liberación de Citoquinas/prevención & control , Receptor 2 de Folato/genética , Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/metabolismo , Macrófagos/efectos de los fármacos , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Células CHO , Cricetulus , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Femenino , Receptor 1 de Folato/antagonistas & inhibidores , Receptor 1 de Folato/genética , Receptor 1 de Folato/inmunología , Receptor 2 de Folato/antagonistas & inhibidores , Receptor 2 de Folato/inmunología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Modelos Biológicos , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/patología , Células RAW 264.7 , Ratas , Ratas Endogámicas Lew , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
20.
Mol Pharm ; 7(2): 597-604, 2010 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-20199053

RESUMEN

Administration of certain antifolates before radiofolate application has previously proven to have a positive effect on undesired kidney uptake of radiofolates in mice bearing human tumor xenografts. The aims of this study were to (i) test the effects of the antifolates, pemetrexed and CB3717, on tissue distribution of the clinically investigated radiofolate, (99m)Tc-EC20, and (ii) to determine if pemetrexed's kidney-selective blocking effect also functions in mice bearing syngeneic tumors. Relative binding affinities of pemetrexed and CB3717 were determined in folate receptor (FR)-positive KB cells at 0 and 37 degrees C using (3)H-folic acid. In vivo studies were performed in nude mice with KB tumor xenografts (A) and in Balb/c mice bearing FR-positive M109 tumor grafts (B). (99m)Tc-EC20 was prepared via a kit formulation. The antifolates pemetrexed and CB3717 (20 mumol/kg body weight) were administered intravenously 1 h before injection of (99m)Tc-EC20 (67 nmol/kg body weight). Similar to previously published data we found that FR-binding affinities of pemetrexed and CB3717 at 0 degrees C were in the same range as that of folic acid. Interestingly, experiments performed at 37 degrees C showed that pemetrexed has a nearly approximately 700-fold lower FR-affinity than CB3717. Tissue distribution of (99m)Tc-EC20 was largely comparable in both animal models (A and B). Radiofolate accumulation was found in FR-positive tumors (A, 8.92 +/- 2.14% ID/g; B, 15.02 +/- 0.95% ID/g) and FR-positive kidneys (A, 59.10 +/- 8.03% ID/g; B, 69.44 +/- 4.66% ID/g, 4 h p.i.). Preinjection of pemetrexed resulted in a significant decrease of (99m)Tc-EC20 uptake in kidney (A, 18.80 +/- 2.73% ID/g; B, 15.27 +/- 2.64% ID/g; 4 h p.i), whereas uptake in the tumors was unaltered. However, administration of the CB3717 resulted in a reduction of (99m)Tc-EC20 uptake in both the kidney and tumor (<1% ID/g, 4 h p.i.). We have thus demonstrated that pemetrexed effectively reduces kidney uptake of radiofolates not only in xenografted mice but also in a syngeneic tumor mouse model, thereby indicating that the kidney-specific blocking effect is not based on differences between human and murine FRs that are expressed in xenografts and kidneys, respectively. This effect was not observed with the antifolate, CB3717, which targets the FR selectively in contrast to pemetrexed that is predominantly transported into cells through carrier systems.


Asunto(s)
Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/análogos & derivados , Glutamatos/farmacología , Guanina/análogos & derivados , Oligopéptidos/metabolismo , Quinazolinas/farmacología , Tecnecio/química , Animales , Línea Celular Tumoral , Femenino , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Guanina/farmacología , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Ratones Desnudos , Compuestos de Organotecnecio/química , Pemetrexed , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA