Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34508005

RESUMEN

Macromolecules such as monoclonal antibodies (mAbs) are likely to experience poor tumor penetration because of their large size, and thus low drug exposure of target cells within a tumor could contribute to suboptimal responses. Given the challenge of inadequate quantitative tools to assess mAb activity within tumors, we hypothesized that measurement of accessible target levels in tumors could elucidate the pharmacologic activity of a mAb and could be used to compare the activity of different mAbs. Using positron emission tomography (PET), we measured the pharmacodynamics of immune checkpoint protein programmed-death ligand 1 (PD-L1) to evaluate pharmacologic effects of mAbs targeting PD-L1 and its receptor programmed cell death protein 1 (PD-1). For PD-L1 quantification, we first developed a small peptide-based fluorine-18-labeled PET imaging agent, [18F]DK222, which provided high-contrast images in preclinical models. We then quantified accessible PD-L1 levels in the tumor bed during treatment with anti-PD-1 and anti-PD-L1 mAbs. Applying mixed-effects models to these data, we found subtle differences in the pharmacodynamic effects of two anti-PD-1 mAbs (nivolumab and pembrolizumab). In contrast, we observed starkly divergent target engagement with anti-PD-L1 mAbs (atezolizumab, avelumab, and durvalumab) that were administered at equivalent doses, correlating with differential effects on tumor growth. Thus, we show that measuring PD-L1 pharmacodynamics informs mechanistic understanding of therapeutic mAbs targeting PD-L1 and PD-1. These findings demonstrate the value of quantifying target pharmacodynamics to elucidate the pharmacologic activity of mAbs, independent of mAb biophysical properties and inclusive of all physiological variables, which are highly heterogeneous within and across tumors and patients.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Radioisótopos de Flúor/farmacocinética , Fragmentos de Péptidos/farmacocinética , Tomografía de Emisión de Positrones/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Apoptosis , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Radiofármacos/farmacocinética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Labelled Comp Radiopharm ; 64(6): 243-250, 2021 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-33576099

RESUMEN

[111 In]In-XYIMSR-01 is a promising single-photon emission computed tomography (SPECT) imaging agent for identification of tumors that overexpress carbonic anhydrase IX. To translate [111 In]In-XYIMSR-01 to phase I trials, we performed animal toxicity and dosimetry studies, determined the maximum dose for human use, and completed the chemistry, manufacturing, and controls component of a standard regulatory application. The production process, quality control testing, stability studies, and specifications for sterile drug product release were based on United States Pharmacopeia chapters <823> and <825>, FDA 21 CFR Part 212. Toxicity was evaluated by using nonradioactive [113/115 In]In-XYIMSR-01 according to 21 CFR Part 58 guidelines. Organ Level INternal Dose Assessment/EXponential Modeling (OLINDA/EXM) was used to calculate the maximum single dose for human studies. Three process validation runs at starting radioactivities of ~800 MBq were completed with a minimum concentration of 407 MBq/ml and radiochemical purity of ≥99% at the end of synthesis. A single intravenous dose of 55 µg/ml of [113/115 In]In-XYIMSR-01 was well tolerated in male and female Sprague-Dawley rats. The calculated maximum single dose for human injection from dosimetry studies was 390.35 MBq of [111 In]In-XYIMSR-01. We have completed toxicity and dosimetry studies as well as validated a manufacturing process to test [111 In]In-XYIMSR-01 in a phase I clinical trial.


Asunto(s)
Antígenos de Neoplasias , Anhidrasa Carbónica IX
3.
Mol Pharm ; 15(9): 3946-3952, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30037229

RESUMEN

Tumors create and maintain an immunosuppressive microenvironment that promotes cancer cell escape from immune surveillance. The immune checkpoint protein programmed death-ligand 1 (PD-L1) is expressed in many cancers and is an important contributor to the maintenance of the immunosuppressive tumor microenvironment. PD-L1 is a prominent target for cancer immunotherapy. Guidance of anti-PD-L1 therapy is currently effected through measurement of PD-L1 through biopsy and immunohistochemistry. Here, we report a peptide-based imaging agent, [68Ga]WL12, to detect PD-L1 expression in tumors noninvasively by positron emission tomography (PET). WL12, a cyclic peptide comprising 14 amino acids, binds to PD-L1 with high affinity (IC50≈ 23 nM). Synthesis of [68Ga]WL12 provided radiochemical purity >99% after purification. Biodistribution in immunocompetent mice demonstrated 11.56 ± 3.18, 4.97 ± 0.8, 1.9 ± 0.1, and 1.33 ± 0.21 percentage of injected dose per gram (%ID/g) in hPD-L1, MDAMB231, SUM149, and CHO tumors, respectively, at 1 h postinjection, with high binding specificity noted with coinjection of excess, nonradiolabeled WL12. PET imaging demonstrated high tissue contrast in all tumor models tested.


Asunto(s)
Antígeno B7-H1/metabolismo , Radioisótopos de Galio/química , Péptidos/química , Tomografía de Emisión de Positrones/métodos , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Células CHO , Cricetulus , Femenino , Citometría de Flujo , Inmunohistoquímica , Ratones
4.
Mol Imaging ; 12(1): 17-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23348788

RESUMEN

The epidermal growth factor receptor (EGFR) is overexpressed in the majority of malignancies and has been associated with poor outcomes. Panitumumab, an anti-EGFR monoclonal antibody that binds to the extracellular binding domain of EGFR, is increasingly used with radiotherapy and chemotherapy but has associated toxicities. The purpose of this study was to develop and characterize a novel targeted imaging agent for the EGFR using radiolabeled panitumumab. Flow cytometry studies were performed to evaluate EGFR expression in several cell lines. Desferrioxamine-Bz-NCS (DFO) was conjugated to panitumumab and labeled with (89)Zr. Cell uptake studies were performed in four cell lines. For biodistribution studies and micro-positron emission tomography/computed tomography (PET/CT), mouse xenograft models were generated using the same cell lines. PET was performed, and tumors and select organs were harvested for biodistribution studies. Panitumumab was radiolabeled with (89)Zr with high radiochemical purity and specific activity and was found to be stable in serum. Cell binding studies demonstrated that radiotracer uptake in cells correlated with the degree of EGFR expression. MicroPET/CT imaging studies demonstrated a high intensity of (89)Zr-panitumumab in A431 and HCT 116 tumors in comparison with the EGFR-negative tumors. Biodistribution studies confirmed the results from the imaging studies. (89)Zr-panitumumab imaging of EGFR-positive tumors demonstrated levels of radiotracer uptake associated with EGFR expression.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Receptores ErbB/metabolismo , Inmunohistoquímica/métodos , Tomografía de Emisión de Positrones/métodos , Radioisótopos/farmacocinética , Circonio/farmacocinética , Animales , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Estabilidad de Medicamentos , Receptores ErbB/análisis , Citometría de Flujo , Humanos , Ratones , Ratones Desnudos , Imagen Multimodal , Panitumumab , Radioisótopos/química , Distribución Tisular , Tomografía Computarizada por Rayos X , Trasplante Heterólogo , Circonio/química
5.
Cancer Immunol Immunother ; 61(4): 581-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22297566

RESUMEN

The tumor-associated carbohydrate antigen/hapten Thomsen-nouveau (Tn; a-D-GalpNAc-ONH2) was conjugated to a zwitterionic capsular polysaccharide, PS A1, from commensal anaerobe Bacteroides fragilis ATCC 25285/NCTC 9343 for the development of an entirely carbohydrate cancer vaccine construct and probed for immunogenicity. This communication discloses that murine anti-Tn IgG3 antibodies both bind to and recognize human tumor cells that display the Tn hapten. Furthermore, the sera from immunization of mice with Tn-PS A1 contain cytokine interleukin 17 (IL-17A), which is known to possess anti-tumor function and represents a striking difference to an IL-2, and IL-6 profile obtained with anti-PS A1 sera.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Bacteroides fragilis/inmunología , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/inmunología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Carbohidratos/química , Carbohidratos/inmunología , Línea Celular Tumoral , Femenino , Humanos , Sueros Inmunes/inmunología , Inmunidad , Inmunización , Inmunoglobulina G/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Polisacáridos Bacterianos/administración & dosificación , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/genética , Polisacáridos Bacterianos/inmunología
6.
J Am Chem Soc ; 131(28): 9622-3, 2009 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-19552408

RESUMEN

An entirely carbohydrate-based immunogen consisting of a zwitterionic polysaccharide (ZPS) PS A1 and the well-known tumor antigen Tn has been designed, synthesized, and studied for immunological effects. The PS A1 motif was included to act as an MHCII elicitor for a T-cell-dependent immune response with increased immunogenicity against tumor-associated carbohydrate antigens, providing an alternative to carrier proteins. Through the use of C57BL/6 mice, it has been shown that chemical modification of PS A1 does not alter the recognition sequence responsible for an MHCII-mediated, T-cell-dependent immune response. The Tn-PS A1 conjugate construct confers specificity toward the Tn antigen alone, and specific carbohydrate immunoglobulins, namely, IgG3, are generated from intraperitoneal immunizations with or without adjuvant. The properties of the vaccine candidate are attributed to a site-specific linking strategy that incurs significant incorporation of Tn antigen.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/inmunología , Inmunoconjugados/inmunología , Oligosacáridos/inmunología , Vacunas Sintéticas/inmunología , Animales , Antígenos de Carbohidratos Asociados a Tumores/química , Bovinos , Haptenos/inmunología , Antígenos de Histocompatibilidad/inmunología , Inmunoconjugados/química , Inmunoterapia , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Oligosacáridos/química , Vacunas Sintéticas/química
7.
Medchemcomm ; 5(8): 1143-1149, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25383162

RESUMEN

The zwitterionic polysaccharide PS A1 from anaerobe Bacteroides fragilis ATCC 25285/NCTC 9343 is known to elicit a T-cell-dependent, major histocompatibility complex class II (MHCII) immune response through a correspondingly similar protein-antigen-based mechanism/pathway. The biological activity of PS A1 is known to arise from alternating charged motifs on adjacent monosaccharides comprising a tetrameric repeating oligomeric unit creating an alpha-helical secondary structure. However, we have learned that this alpha-helical structural characteristic may not play a role in immune activation. Paradoxically, our current knowledge of structure - activity relationships (SARs) with electrostatically charged polysaccharides has become more clearly defined, yet a lack of tools/probes for measuring dynamic structural changes hinders progress in carbohydrate-based vaccine development. Site- and region-specific structural modifications of PS A1, followed by conjugation with a known carbohydrate cancer antigen, the Thomsen-nouveau (Tn = alpha-D-GalNAc-OSer/Thr) antigen, does not alter antibody isotype switching ability and leads to specific IgG3 antibodies in C57BL/6 mice. Circular dichroism (CD) and studies using fluorescently labeled PS A1, described herein, reveal information pertaining to structure - activity relationships and the nature of Tn conjugation to chemically modified PS A1. The CD spectra of a Tn-PS A1 construct at 8.5 ≥ pH ≤ 3.5 illustrates complete loss of alpha-helical character while spectra obtained in the 3.6 ≤ pH ≥ 8.4 range denotes minimal alpha-helicity in comparison to naturally occurring PS A1. Temperatures exceeding 60 °C reveal complete loss of helical character. Two methods for Alexa Fluor488® fluorescent labeling studies of chemically oxidized PS A1 have given rise to percent conjugation values (% loading) calculated to be on average 35 Tn molecules bound. Combined, our results argue that altering the structure of PS A1, without chemically modifying the electrostatic charge character, does not alter immune response/recognition in mice. These findings have important implications for the design of entirely carbohydrate-based vaccine constructs.

8.
Nucl Med Biol ; 41(7): 552-61, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25038987

RESUMEN

Over-expression of chemokine receptor 4 (CXCR4) is present in a majority of cancers, has been linked to an aggressive phenotype, and may indicate the metastatic potential of primary tumor. Several CXCR4 targeted therapeutics are in clinical trials and the development of the corresponding imaging agents is an area of active interest. Previously, (64)Cu-labeled imaging agents for CXCR4 have provided clear images of CXCR4-bearing tissues in relevant experimental models but demonstrated fast washout from tissues harboring receptor. Addition of stabilizing bridges is known to provide more robust chelator-Cu(II) complexes. In addition, bridged cyclam-based CXCR4 binding agents demonstrated increased receptor residence times relative to existing agents. Based on that knowledge we synthesized several bridged cyclam analogs of AMD3465, a monocyclam-based CXCR4 imaging agent, to increase the retention time of the tracer bound to the receptor to allow for protracted imaging and improved target-to-non-target ratios. Specific accumulation of two radiolabeled, cross-bridged analogs ([(64)Cu] RAD1-24 and [(64)Cu]RAD1-52) was observed in U87-stb-CXCR4 tumors in both PET/CT imaging and biodistribution studies. At 90min post-injection of radiotracer, tumor-to-muscle and tumor-to-blood ratios reached 106.05±17.19 and 28.08±4.78, respectively, for cross-bridged pyrimidine analog [(64)Cu]RAD1-52. Receptor blockade performed in vivo denoted target binding specificity. The biodistribution and PET/CT imaging studies with the radiolabeled bridged cyclams demonstrated longer tumor retention and comparable uptake to [(64)Cu]AMD3465, though [(64)Cu]AMD3465 demonstrated superior overall pharmacokinetics.


Asunto(s)
Lactamas Macrocíclicas/química , Tomografía de Emisión de Positrones/métodos , Receptores CXCR4/metabolismo , Tomografía Computarizada por Rayos X/métodos , Animales , Unión Competitiva , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lactamas Macrocíclicas/metabolismo , Lactamas Macrocíclicas/farmacocinética , Ratones , Distribución Tisular
9.
Nucl Med Biol ; 39(5): 609-16, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22261146

RESUMEN

INTRODUCTION: Bombesin (BN) is an amphibian peptide that binds to the gastrin-releasing peptide receptor (GRPR). It has been demonstrated that BN analogues can be radiolabeled for potential diagnosis and treatment of GRPR-expressing malignancies. Previous studies have conjugated various chelators to the eight C-terminal amino acids of BN [BN(7-14)] for radiolabeling with 64Cu. Recently, (1,4,7-triazacyclononane-1,4,7-triacetic acid) (NOTA) has been evaluated as the five-coordinate 64Cu complex, with results indicating GRPR-specific tumor uptake. This study aimed to conjugate S-2-(4-isothiocyanatobenzyl)-NOTA (p-SCN-Bn-NOTA) to BN(7-14) such that it could form a six-coordinate complex with 64Cu and to evaluate the resulting peptide. METHODS: p-SCN-NOTA was conjugated to 8-aminooctanoic acid (Aoc)-BN(7-14) in solution to yield NOTA-Bn-SCN-Aoc-BN(7-14). The unlabeled peptide was evaluated in a cell binding assay using PC-3 prostate cancer cells and 125I-Tyr4-BN to determine the IC50 value. The peptide was radiolabeled with 64Cu and evaluated for internalization into PC-3 cells and for tumor uptake in mice bearing PC-3 xenografts using biodistribution and micro-positron emission tomography imaging studies. RESULTS: The binding assay demonstrated that NOTA-Bn-SCN-Aoc-BN(7-14) bound with high affinity to GRPR with an IC50 of 1.4 nM. The radiolabeled peptide demonstrated time-dependent internalization into PC-3 cells. In vivo, the peptide demonstrated tumor-specific uptake and imaging that were comparable to those of previously reported 64Cu-labeled BN analogues. CONCLUSIONS: These studies demonstrate that 64Cu-NOTA-Bn-SCN-Aoc-BN(7-14) binds to GRPR-expressing cells and that it can be used for imaging of GRPR-expressing prostate cancer.


Asunto(s)
Bombesina/metabolismo , Caprilatos/química , Radioisótopos de Cobre , Regulación Neoplásica de la Expresión Génica , Compuestos Heterocíclicos/química , Isotiocianatos/química , Neoplasias de la Próstata/patología , Receptores de Bombesina/metabolismo , Animales , Unión Competitiva , Transporte Biológico , Bombesina/análogos & derivados , Bombesina/farmacocinética , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Humanos , Marcaje Isotópico , Masculino , Ratones , Imagen Multimodal , Tomografía de Emisión de Positrones , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/metabolismo , Tomografía Computarizada por Rayos X
10.
Nucl Med Biol ; 39(8): 1099-104, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22743158

RESUMEN

INTRODUCTION: The development of novel bifunctional chelates for attaching copper-64 to biomolecules has been an active area of research for several years. However, many of these (64)Cu-chelates have poor in vivo stability or harsh radiolabeling conditions. METHODS: In this study, two triazacyclononane analogs; C-NE3TA (4-carboxymethyl-7-[2-(carboxymethyl-amino)-3-(4-nitro-phenyl)-propyl]-[1,4,7]triazo-nan-1-yl-acetic acid) and N-NE3TA (4-carboxymethyl-7-[2-[carboxymethyl-(4-nitro-benzyl)-amino]-ethyl]-[1,4,7]triazonan-1-yl-acetic acid) were evaluated for their labeling efficiency with (64)Cu at room temperature and evaluated in vitro and in vivo. In vitro studies included complexation kinetics with Cu(II) using a spectrophotometric method and rat serum stability, while the in vivo biodistribution was evaluated using SCID mice. RESULTS: C-NE3TA and N-NE3TA were labeled at >95% efficiency up to ~3.4Ci/µmol. Both C-NE3TA and N-NE3TA formed complexes with Cu(II) almost immediately, with the Cu(II) complexation by C-NE3TA being faster than the formation of Cu(II)-N-NE3TA. Both (64)Cu-N-NE3TA and (64)Cu-C-NE3TA were 96.1% and 90.5% intact after 48h incubation in rat serum, respectively. This is compared to (64)Cu complexes of the control chelators, p-NH(2)-Bn-DOTA and p-NH(2)-Bn-NOTA, with 93.9% and 97.9% retention of (64)Cu in the complex, respectively. In vivo evaluation of (64)Cu-N-NE3TA and (64)Cu-C-NE3TA demonstrates good clearance from normal tissues except for the liver, where 59% and 51% of the radioactivity is retained at 24h compared to 1h for (64)Cu-N-NE3TA and (64)Cu-C-NE3TA, respectively. This compares to 78% and 3% retention for (64)Cu-p-NH(2)-Bn-DOTA and (64)Cu-p-NH(2)-Bn-NOTA. CONCLUSIONS: These studies demonstrate that while N-NE3TA and C-NE3TA appear to be superior chelators for (64)Cu than p-NH(2)-Bn-DOTA, they are not better than p-NH(2)-Bn-NOTA. Nevertheless, it may still be interesting to evaluate these chelators after conjugation to biomolecules.


Asunto(s)
Quelantes/química , Quelantes/farmacocinética , Radioisótopos de Cobre/química , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/farmacocinética , Radiofármacos/química , Radiofármacos/farmacocinética , Animales , Descubrimiento de Drogas , Estabilidad de Medicamentos , Femenino , Compuestos Heterocíclicos/sangre , Marcaje Isotópico , Cinética , Ratones , Radiofármacos/sangre , Ratas , Temperatura
11.
J Nucl Med ; 52(6): 986-93, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21622896

RESUMEN

The chemokine receptor 4 (CXCR4) is overexpressed in several cancers and metastases and as such presents an enticing target for molecular imaging of metastases and metastatic potential of the primary tumor. CXCR4-based imaging agents could also be useful for diagnosis, staging, and therapeutic monitoring. Here we evaluated a positron-emitting monocyclam analog, (64)Cu-{N-[1,4,8,11-tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine} ((64)Cu-AMD3465), in subcutaneous U87 brain tumors and U87 tumors stably expressing CXCR4 (U87-stb-CXCR4) and in colon tumors (HT-29) using dynamic and whole-body PET supported by ex vivo biodistribution studies. Both dynamic and whole-body PET/CT studies show specific accumulation of radioactivity in U87-stb-CXCR4 tumors, with the percentage injected dose per gram reaching a maximum of 102.70 ± 20.80 at 60 min and tumor-to-muscle ratios reaching a maximum of 362.56 ± 153.51 at 90 min after injection of the radiotracer. Similar specificity was also observed in the HT-29 colon tumor model. Treatment with AMD3465 inhibited uptake of radioactivity by the tumors in both models. Our results show that (64)Cu-AMD3465 is capable of detecting lesions in a CXCR4-dependent fashion, with high target selectivity, and may offer a scaffold for the synthesis of clinically translatable agents.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Trasplante de Neoplasias/diagnóstico por imagen , Piridinas , Radiofármacos , Receptores CXCR4/biosíntesis , Animales , Línea Celular Tumoral , Neoplasias del Colon/diagnóstico por imagen , Radioisótopos de Cobre , Femenino , Citometría de Flujo , Células HT29 , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos NOD , Tomografía de Emisión de Positrones , Piridinas/síntesis química , Piridinas/química , Ensayo de Unión Radioligante , Radiofármacos/síntesis química , Radiofármacos/química , Solubilidad , Distribución Tisular , Recuento Corporal Total
12.
Org Lett ; 10(20): 4541-4, 2008 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-18811177

RESUMEN

A one-pot, two-step synthesis of the title compounds employs a multicomponent Ugi condensation reaction, microwave irradiation, and Fe(0) as a reductant. Two pathways are accessible; both routes utilize bifunctional, o-nitro-substituted arenes leading to either C2, N4, C5 substitution (A) or C2, N4 substitution (B).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA