Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Cancer Ther ; 21(9): 1462-1472, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35793468

RESUMEN

Extra domain B splice variant of fibronectin (EDB+FN) is an extracellular matrix protein (ECM) deposited by tumor-associated fibroblasts, and is associated with tumor growth, angiogenesis, and invasion. We hypothesized that EDB+FN is a safe and abundant target for therapeutic intervention with an antibody-drug conjugate (ADC). We describe the generation, pharmacology, mechanism of action, and safety profile of an ADC specific for EDB+FN (EDB-ADC). EDB+FN is broadly expressed in the stroma of pancreatic, non-small cell lung (NSCLC), breast, ovarian, head and neck cancers, whereas restricted in normal tissues. In patient-derived xenograft (PDX), cell-line xenograft (CLX), and mouse syngeneic tumor models, EDB-ADC, conjugated to auristatin Aur0101 through site-specific technology, demonstrated potent antitumor growth inhibition. Increased phospho-histone H3, a pharmacodynamic biomarker of response, was observed in tumor cells distal to the target site of tumor ECM after EDB-ADC treatment. EDB-ADC potentiated infiltration of immune cells, including CD3+ T lymphocytes into the tumor, providing rationale for the combination of EDB-ADC with immune checkpoint therapy. EDB-ADC and anti-PD-L1 combination in a syngeneic breast tumor model led to enhanced antitumor activity with sustained tumor regressions. In nonclinical safety studies in nonhuman primates, EDB-ADC had a well-tolerated safety profile without signs of either on-target toxicity or the off-target effects typically observed with ADCs that are conjugated through conventional conjugation methods. These data highlight the potential for EDB-ADC to specifically target the tumor microenvironment, provide robust therapeutic benefits against multiple tumor types, and enhance activity antitumor in combination with checkpoint blockade.


Asunto(s)
Neoplasias de la Mama , Inmunoconjugados , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Fibronectinas/metabolismo , Humanos , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Ratones , Neovascularización Patológica/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
2.
MAbs ; 13(1): 1958662, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34347577

RESUMEN

IL13Rα2 is a cell surface tumor antigen that is overexpressed in multiple tumor types. Here, we studied biodistribution and targeting potential of an anti-IL13Rα2 antibody (Ab) and anti-tumor activity of anti-IL13Rα2-antibody-drug conjugate (ADC). The anti-IL13Rα2 Ab was labeled with fluorophore AF680 or radioisotope 89Zr for in vivo tracking using fluorescence molecular tomography (FMT) or positron emission tomography (PET) imaging, respectively. Both imaging modalities showed that the tumor was the major uptake site for anti-IL13Rα2-Ab, with peak uptake of 5-8% ID and 10% ID/g as quantified from FMT and PET, respectively. Pharmacological in vivo competition with excess of unlabeled anti-IL13Rα2-Ab significantly reduced the tumor uptake, indicative of antigen-specific tumor accumulation. Further, FMT imaging demonstrated similar biodistribution and pharmacokinetic profiles of an auristatin-conjugated anti-IL13Rα2-ADC as compared to the parental Ab. Finally, the anti-IL13Rα2-ADC exhibited a dose-dependent anti-tumor effect on A375 xenografts, with 90% complete responders at a dose of 3 mg/kg. Taken together, both FMT and PET showed a favorable biodistribution profile for anti-IL13Rα2-Ab/ADC, along with antigen-specific tumor targeting and excellent therapeutic efficacy in the A375 xenograft model. This work shows the great potential of this anti-IL13Rα2-ADC as a targeted anti-cancer agent.


Asunto(s)
Aminobenzoatos , Antineoplásicos Inmunológicos , Inmunoconjugados , Subunidad alfa2 del Receptor de Interleucina-13 , Melanoma Experimental , Proteínas de Neoplasias , Oligopéptidos , Aminobenzoatos/inmunología , Aminobenzoatos/farmacocinética , Aminobenzoatos/farmacología , Animales , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Humanos , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Inmunoconjugados/farmacología , Subunidad alfa2 del Receptor de Interleucina-13/antagonistas & inhibidores , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Oligopéptidos/inmunología , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33148666

RESUMEN

PURPOSE: Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN: We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS: Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS: We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.


Asunto(s)
Gemtuzumab/uso terapéutico , Inmunoconjugados/uso terapéutico , Subunidad alfa del Receptor de Interleucina-3/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Lectina 3 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Animales , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/uso terapéutico , Área Bajo la Curva , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Gemtuzumab/inmunología , Gemtuzumab/farmacocinética , Células HL-60 , Humanos , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Subunidad alfa del Receptor de Interleucina-3/inmunología , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Macaca fascicularis , Ratones , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Oncotarget ; 8(34): 57231-57245, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28915667

RESUMEN

Non-invasive imaging using radiolabels is a common technique used to study the biodistribution of biologics. Due to the limited shelf-life of radiolabels and the requirements of specialized labs, non-invasive optical imaging is an attractive alternative for preclinical studies. Previously, we demonstrated the utility of fluorescence molecular tomography (FMT) an optical imaging modality in evaluating the biodistribution of antibody-drug conjugates. As FMT is a relatively new technology, few fluorophores have been validated for in vivo imaging. The goal of this study was to characterize and determine the utility of near-infrared (NIR) fluorophores for biodistribution studies using interleukin-13 receptor subunit alpha-2 antibody (IL13Rα2-Ab). Eight fluorophores (ex/em: 630/800 nm) with an N-hydroxysuccinimide (NHS) linker were evaluated for Ab conjugation. The resulting antibody-fluorophore (Ab-F) conjugates were evaluated in vitro for degree of conjugation, stability and target-binding, followed by in vivo/ex vivo FMT imaging to determine biodistribution in a xenograft model. The Ab-F conjugates (except Ab-DyLight800) showed good in vitro stability and antigen binding. All Ab-F conjugates (except for Ab-BOD630) resulted in a quantifiable signal in vivo and had similar biodistribution profiles, with peak tumor accumulation between 6 and 24 h post-injection. In vivo/ex vivo FMT imaging showed 17-34% ID/g Ab uptake by the tumor at 96 h. Overall, this is the first study to characterize the biodistribution of an Ab using eight NIR fluorophores. Our results show that 3-dimensional optical imaging is a valuable technology to understand biodistribution and targeting, but a careful selection of the fluorophore for each Ab is warranted.

5.
Rev. colomb. cardiol ; 24(1): 60-65, ene.-feb. 2017. tab, graf
Artículo en Español | LILACS, COLNAL | ID: biblio-900494

RESUMEN

Resumen Objetivo: El manejo de la isquemia crítica de los miembros inferiores representa un reto para el cirujano vascular debido a la alta tasa de amputaciones y mortalidad. Las opciones de manejo actuales: puente femorodistal, angioplastia con o sin la colocación de Stents y la resección de la placa con láser o de manera mecánica, presentan a largo plazo una tasa de éxito muy baja y un número de amputaciones supracondíleas que continúa siendo elevado. Métodos: Para este estudio prospectivo se reclutaron 173 pacientes con diagnóstico de estadio avanzado, con isquemia crítica de miembro inferior quienes fueron tratados con alprostadil (60- 120 mcgr/día) por vía intravenosa sistémica por 28 días. La respuesta se midió clínicamente por mejoría del llenado capilar y con el uso de la escala análoga visual del dolor. Resultados: Al momento del alta hospitalaria el 94.3% de los pacientes mejoró el puntaje en la escala análoga visual del dolor (p < 0.0001). El seguimiento a más de un año del tratamiento con alprostadil mostró que el 97% de los pacientes mejoró significativamente su estadio de isquemia, evitándose así una amputación mayor. No se observó respuesta al tratamiento en pacientes previamente intervenidos por vía endovascular (5 pacientes). Conclusiones: El tratamiento de pacientes con isquemia crítica de miembro inferior con alpostadil por infusión intravenosa, con bolos diarios de entre 60 y 120 mcg durante 28 días, este medicamento es seguro y presenta mínimos efectos secundarios. Esta terapia mejora sustancialmente el estadio funcional de Rutherford en estos pacientes y evita amputaciones mayores.


Abstract Objetive: Management of critical lower limb ischemia represents a challenge for the vascular surgeon due to the high rate of amputations and mortality. Current management options include femorodistal bypass, angioplasty with or without stent and laser or mechanical resection of the plaque. They present a low success rate in the long run and a number of supracondylar amputations that still remains high. Methods: This prospective study included 173 patients diagnosed with advanced stage critical lower limb ischemia who were treated with systemic intravenous alprostadil (60 - 120 mcg/day) during 8 days. Response was measured clinically with improvement of capillary refill and using the visual analog scale for pain. Results: Upon discharge 94.3% of patients improved their visual analogue scale score for pain (p < 0.0001). Follow-up for more than a year of alprostadil treatment revealed that 97% of patients significantly improved their ischemia status, thus avoiding further amputation. No response to treatment was observed in patients who had previously undergone endovascular surgery (5 patients). Conclusions: Treating patients with critical lower limb ischemia with intravenous alprostadil, administering daily doses of between 60 and 120 mcg during 28 days shows that this drug is safe and causes minimal secondary effects. This therapy significantly improves Rutherford's function state in these patients and avoids further amputations.


Asunto(s)
Humanos , Masculino , Anciano , Enfermedad Arterial Periférica , Isquemia , Reperfusión , Prostaglandinas
6.
Bioanalysis ; 8(21): 2205-2217, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27682846

RESUMEN

AIM: Complex nature of bioconjugates require multiple bioanalytical approaches to support PK and absorption, distribution, metabolism and excretion characterization. For antibody-drug conjugate (ADC) bioanalysis both LC-MS and ligand-binding assays (LBAs) are employed. RESULTS: A method consisting of immunocapture extraction of ADC from biomatrices followed by LC-MS analysis of light and heavy chain is described. Drug antibody ratio (DAR) profiles of ADC Tras-mcVC-PF06380101 dosed at 0.3, 1 and 3 mg/kg in Sprague Dawley rats were obtained. Combined with total antibody (monoclonal antibody) measurement by LBA, conjugated payload concentration was calculated. CONCLUSION: PK profiles from LBA, ADC and calculated conjugated payload (DAR × monoclonal antibody) were in good agreement. We present a new tool for PK assessment of ADCs while also exploring ADC metabolism and DAR sensitivity of LBA ADC assay.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoconjugados/sangre , Espectrometría de Masas , Preparaciones Farmacéuticas/química , Animales , Cromatografía Líquida de Alta Presión , Semivida , Inmunoensayo , Inmunoconjugados/farmacocinética , Ratas , Ratas Sprague-Dawley , Trastuzumab/química
7.
Mol Cancer Ther ; 15(10): 2530-2540, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27466353

RESUMEN

Understanding a drug's whole-body biodistribution and tumor targeting can provide important information regarding efficacy, safety, and dosing parameters. Current methods to evaluate biodistribution include in vivo imaging technologies like positron electron tomography and single-photon emission computed tomography or ex vivo quantitation of drug concentrations in tissues using autoradiography and standard biochemical assays. These methods use radioactive compounds or are cumbersome and do not give whole-body information. Here, for the first time, we show the utility of fluorescence molecular tomography (FMT) imaging to determine the biodistribution and targeting of an antibody-drug conjugate (ADC). An anti-5T4-antibody (5T4-Ab) and 5T4-ADC were conjugated with a near-infrared (NIR) fluorophore VivoTag 680XL (VT680). Both conjugated compounds were stable as determined by SEC-HPLC and plasma stability studies. Flow cytometry and fluorescence microscopy studies showed that VT680-conjugated 5T4-ADC specifically bound 5T4-expressing cells in vitro and also exhibited a similar cytotoxicity profile as the unconjugated 5T4-ADC. In vivo biodistribution and tumor targeting in an H1975 subcutaneous xenograft model demonstrated no significant differences between accumulation of VT680-conjugated 5T4-Ab or 5T4-ADC in either normal tissues or tumor. In addition, quantitation of heart signal from FMT imaging showed good correlation with the plasma pharmacokinetic profile suggesting that it (heart FMT imaging) may be a surrogate for plasma drug clearance. These results demonstrate that conjugation of VT680 to 5T4-Ab or 5T4-ADC does not change the behavior of native biologic, and FMT imaging can be a useful tool to understand biodistribution and tumor-targeting kinetics of antibodies, ADCs, and other biologics. Mol Cancer Ther; 15(10); 2530-40. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacocinética , Inmunoconjugados/farmacocinética , Glicoproteínas de Membrana/antagonistas & inhibidores , Neoplasias/diagnóstico por imagen , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Fluorescencia , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Imagen Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Distribución Tisular , Tomografía Computarizada por Rayos X , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Bioconjug Chem ; 26(11): 2223-32, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26180901

RESUMEN

The pharmacokinetics of an antibody (huA1)-drug (auristatin microtubule disrupting MMAF) conjugate, targeting 5T4-expressing cells, were characterized during the discovery and development phases in female nu/nu mice and cynomolgus monkeys after a single dose and in S-D rats and cynomolgus monkeys from multidose toxicity studies. Plasma/serum samples were analyzed using an ELISA-based method for antibody and conjugate (ADC) as well as for the released payload using an LC-MS/MS method. In addition, the distribution of the Ab, ADC, and released payload (cys-mcMMAF) was determined in a number of tissues (tumor, lung, liver, kidney, and heart) in two tumor mouse models (H1975 and MDA-MB-361-DYT2 models) using similar LBA and LC-MS/MS methods. Tissue distribution studies revealed preferential tumor distribution of cys-mcMMAF and its relative specificity to the 5T4 target containing tissue (tumor). Single dose studies suggests lower CL values at the higher doses in mice, although a linear relationship was seen in cynomolgus monkeys at doses from 0.3 to 10 mg/kg with no evidence of TMDD. Evaluation of DAR (drug-antibody ratio) in cynomolgus monkeys (at 3 mg/kg) indicated that at least half of the payload was still on the ADC 1 to 2 weeks after IV dosing. After multiple doses, the huA1 and conjugate data in rats and monkeys indicate that exposure (AUC) increases with increasing dose in a linear fashion. Systemic exposure (as assessed by Cmax and AUC) of the released payload increased with increasing dose, although exposure was very low and its pharmacokinetics appeared to be formation rate limited. The incidence of ADA was generally low in rats and monkeys. We will discuss cross species comparison, relationships between the Ab, ADC, and released payload exposure after multiple dosing, and insights into the distribution of this ADC with a focus on experimental design as a way to address or bypass apparent obstacles and its integration into predictive models.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Inmunoconjugados/farmacocinética , Glicoproteínas de Membrana/inmunología , Oligopéptidos/farmacocinética , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/inmunología , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Macaca fascicularis , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Oligopéptidos/química , Oligopéptidos/inmunología , Ratas , Ratas Sprague-Dawley , Distribución Tisular
9.
Mol Cancer Ther ; 14(8): 1868-76, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26089370

RESUMEN

Antibody-drug conjugates (ADC) represent a promising therapeutic modality for managing cancer. Here, we report a novel humanized ADC that targets the tetraspanin-like protein TM4SF1. TM4SF1 is highly expressed on the plasma membranes of many human cancer cells and also on the endothelial cells lining tumor blood vessels. TM4SF1 is internalized upon interaction with antibodies. We hypothesized that an ADC against TM4SF1 would inhibit cancer growth directly by killing cancer cells and indirectly by attacking the tumor vasculature. We generated a humanized anti-human TM4SF1 monoclonal antibody, v1.10, and armed it with an auristatin cytotoxic agent LP2 (chemical name mc-3377). v1.10-LP2 selectively killed cultured human tumor cell lines and human endothelial cells that express TM4SF1. Acting as a single agent, v1.10-LP2 induced complete regression of several TM4SF1-expressing tumor xenografts in nude mice, including non-small cell lung cancer and pancreas, prostate, and colon cancers. As v1.10 did not react with mouse TM4SF1, it could not target the mouse tumor vasculature. Therefore, we generated a surrogate anti-mouse TM4SF1 antibody, 2A7A, and conjugated it to LP2. At 3 mpk, 2A7A-LP2 regressed several tumor xenografts without noticeable toxicity. Combination therapy with v1.10-LP2 and 2A7A-LP2 together was more effective than either ADC alone. These data provide proof-of-concept that TM4SF1-targeting ADCs have potential as anticancer agents with dual action against tumor cells and the tumor vasculature. Such agents could offer exceptional therapeutic value and warrant further investigation. Mol Cancer Ther; 14(8); 1868-76. ©2015 AACR.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de la Angiogénesis/toxicidad , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Antineoplásicos/toxicidad , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Expresión Génica , Humanos , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica , Conejos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Pharm ; 12(6): 1730-7, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25423493

RESUMEN

The use of predictive preclinical models in drug discovery is critical for compound selection, optimization, preclinical to clinical translation, and strategic decision-making. Trophoblast glycoprotein (TPBG), also known as 5T4, is the therapeutic target of several anticancer agents currently in clinical development, largely due to its high expression in tumors and low expression in normal adult tissues. In this study, mice were engineered to express human TPBG under endogenous regulatory sequences by replacement of the murine Tpbg coding sequence. The gene replacement was considered functional since the hTPBG knockin (hTPBG-KI) mice did not exhibit clinical observations or histopathological phenotypes that are associated with Tpbg gene deletion, except in rare instances. The expression of hTPBG in certain epithelial cell types and in different microregions of the brain and spinal cord was consistent with previously reported phenotypes and expression patterns. In pharmacokinetic studies, the exposure of a clinical-stage anti-TPBG antibody-drug conjugate (ADC), A1mcMMAF, was lower in hTPBG-KI versus wild-type animals, which was evidence of target-related increased clearance in hTPBG-KI mice. Thus, the hTPBG-KI mice constitute an improved system for pharmacology studies with current and future TPBG-targeted therapies and can generate more precise pharmacokinetic and pharmacodynamic data. In general the strategy of employing gene replacement to improve pharmacokinetic assessments should be broadly applicable to the discovery and development of ADCs and other biotherapeutics.


Asunto(s)
Inmunoconjugados/farmacocinética , Glicoproteínas de Membrana/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Humanos , Inmunohistoquímica , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos , Fenotipo
11.
Ann N Y Acad Sci ; 1321: 41-54, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25123209

RESUMEN

Antibody-drug conjugates (ADCs) offer promise as a therapeutic modality that can potentially reduce the toxicities and poor therapeutic indices caused by the lack of specificity of conventional anticancer therapies. ADCs combine the potency of cytotoxic agents with the target selectivity of antibodies by chemically linking a cytotoxic payload to an antibody, potentially creating a synthetic molecule that will deliver targeted antitumor therapy that is both safe and efficacious. The ADC repertoire contains a range of payload molecules, antibodies, and linkers. Two ADC molecules, Kadcyla® and Adcetris®, have been approved by the FDA, and many more are currently in clinical development.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Terapia Molecular Dirigida/tendencias , Neoplasias/tratamiento farmacológico , Ado-Trastuzumab Emtansina , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/química , Brentuximab Vedotina , Aprobación de Drogas , Diseño de Fármacos , Humanos , Inmunoconjugados/química , Maitansina/análogos & derivados , Maitansina/uso terapéutico , Trastuzumab , Estados Unidos , United States Food and Drug Administration
12.
AAPS J ; 16(3): 452-63, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24578215

RESUMEN

The objectives of this investigation were as follows: (a) to validate a mechanism-based pharmacokinetic (PK) model of ADC for its ability to a priori predict tumor concentrations of ADC and released payload, using anti-5T4 ADC A1mcMMAF, and (b) to analyze the PK model to find out main pathways and parameters model outputs are most sensitive to. Experiential data containing biomeasures, and plasma and tumor concentrations of ADC and payload, following A1mcMMAF administration in two different xenografts, were used to build and validate the model. The model performed reasonably well in terms of a priori predicting tumor exposure of total antibody, ADC, and released payload, and the exposure of released payload in plasma. Model predictions were within two fold of the observed exposures. Pathway analysis and local sensitivity analysis were conducted to investigate main pathways and set of parameters the model outputs are most sensitive to. It was discovered that payload dissociation from ADC and tumor size were important determinants of plasma and tumor payload exposure. It was also found that the sensitivity of the model output to certain parameters is dose-dependent, suggesting caution before generalizing the results from the sensitivity analysis. Model analysis also revealed the importance of understanding and quantifying the processes responsible for ADC and payload disposition within tumor cell, as tumor concentrations were sensitive to these parameters. Proposed ADC PK model provides a useful tool for a priori predicting tumor payload concentrations of novel ADCs preclinically, and possibly translating them to the clinic.


Asunto(s)
Aminobenzoatos/química , Aminobenzoatos/farmacocinética , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/farmacocinética , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Glicoproteínas de Membrana/metabolismo , Oligopéptidos/química , Oligopéptidos/farmacocinética , Animales , Anticuerpos Monoclonales/farmacocinética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Modelos Biológicos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Proc Natl Acad Sci U S A ; 111(5): 1766-71, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24443552

RESUMEN

Using an expanded genetic code, antibodies with site-specifically incorporated nonnative amino acids were produced in stable cell lines derived from a CHO cell line with titers over 1 g/L. Using anti-5T4 and anti-Her2 antibodies as model systems, site-specific antibody drug conjugates (NDCs) were produced, via oxime bond formation between ketones on the side chain of the incorporated nonnative amino acid and hydroxylamine functionalized monomethyl auristatin D with either protease-cleavable or noncleavable linkers. When noncleavable linkers were used, these conjugates were highly stable and displayed improved in vitro efficacy as well as in vivo efficacy and pharmacokinetic stability in rodent models relative to conventional antibody drug conjugates conjugated through either engineered surface-exposed or reduced interchain disulfide bond cysteine residues. The advantages of the oxime-bonded, site-specific NDCs were even more apparent when low-antigen-expressing (2+) target cell lines were used in the comparative studies. NDCs generated with protease-cleavable linkers demonstrated that the site of conjugation had a significant impact on the stability of these rationally designed prodrug linkers. In a single-dose rat toxicology study, a site-specific anti-Her2 NDC was well tolerated at dose levels up to 90 mg/kg. These experiments support the notion that chemically defined antibody conjugates can be synthesized in commercially relevant yields and can lead to antibody drug conjugates with improved properties relative to the heterogeneous conjugates formed by nonspecific chemical modification.


Asunto(s)
Anticuerpos/metabolismo , Inmunoconjugados/metabolismo , Preparaciones Farmacéuticas/síntesis química , Ingeniería de Proteínas/métodos , Animales , Anticuerpos/sangre , Anticuerpos/química , Anticuerpos/toxicidad , Técnicas de Cultivo Celular por Lotes , Células CHO , Muerte Celular/efectos de los fármacos , Línea Celular , Cricetinae , Cricetulus , Cisteína/metabolismo , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Inmunoconjugados/toxicidad , Preparaciones Farmacéuticas/sangre , Preparaciones Farmacéuticas/química , Estabilidad Proteica/efectos de los fármacos , Ratas
14.
Bioanalysis ; 6(1): 21-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24341492

RESUMEN

BACKGROUND: Antibody-drug conjugates (ADCs) are a new generation of anticancer therapeutics. The objective of this manuscript is to propose a methodology that can be used to assess the stability of the ADCs by using the PK data obtained by ligand-binding assays that measure various components of ADCs. RESULTS: The ligand-binding assays format of different components of ADCs provided unique valuable PK information. The mathematical manipulation of the bioanalytical data provided an insight into the in vivo integrity, indicating that the loading of the calicheamicin on the G193 antibody declines in an apparent slow first-order process. CONCLUSION: This report demonstrates the value of analyzing various components of the ADC and their PK profiles to better understand the disposition and in vivo stability of ADCs.


Asunto(s)
Aminoglicósidos/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/farmacocinética , Enediinos/farmacocinética , Inmunoconjugados/farmacocinética , Aminoglicósidos/sangre , Aminoglicósidos/química , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/química , Antineoplásicos/sangre , Antineoplásicos/química , Área Bajo la Curva , Bioensayo , Estabilidad de Medicamentos , Enediinos/sangre , Enediinos/química , Femenino , Semivida , Inmunoconjugados/sangre , Inmunoconjugados/química , Inyecciones Intravenosas , Masculino , Modelos Estadísticos , Ratas , Proteínas Recombinantes/sangre , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética
15.
J Pharmacokinet Pharmacodyn ; 40(5): 557-71, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23933716

RESUMEN

Objectives of the present investigation were: (1) to compare three literature reported tumor growth inhibition (TGI) pharmacodynamic (PD) models and propose an optimal new model that best describes the xenograft TGI data for antibody drug conjugates (ADC), (2) to translate efficacy of the ADC Trastuzumab-emtansine (T-DM1) from mice to patients using the optimized PD model, and (3) to apply the translational strategy to predict clinically efficacious concentrations of a novel in-house anti-5T4 ADC, A1mcMMAF. First, the performance of all four of the PD models (i.e. 3 literature reported + 1 proposed) was evaluated using TGI data of T-DM1 obtained from four different xenografts. Based on the estimates of the pharmacodynamic/pharmacokinetic (PK/PD) modeling, a secondary parameter representing the efficacy index of the drug was calculated, which is termed as the tumor static concentration (TSC). TSC values derived from all four of the models were compared with each other, and with literature reported values, to assess the performance of these models. Subsequently, using the optimized PK/PD model, PD parameters obtained from different cell lines, human PK, and the proposed translational strategy, clinically efficacious doses of T-DM1 were projected. The accuracy of projected efficacious dose range for T-DM1 was verified by comparison with the clinical doses. Aforementioned strategy was then applied to A1mcMMAF for projecting its efficacious concentrations in clinic. TSC values for A1mcMMAF, obtained by fitting TGI data from 4 different xenografts with the proposed PK/PD model, were estimated to range from 0.6 to 11.5 µg mL⁻¹. Accordingly, the clinically efficacious doses for A1mcMMAF were projected retrospectively. All in all, the improved PD model and proposed translational strategy presented here suggest that appropriate correction for the clinical exposure and employing the TSC criterion can help translate mouse TGI data to predict first in human doses of ADCs.


Asunto(s)
Anticuerpos/farmacología , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Inmunoconjugados/farmacología , Inmunoconjugados/farmacocinética , Neoplasias Experimentales/tratamiento farmacológico , Ado-Trastuzumab Emtansina , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticuerpos Monoclonales Humanizados/farmacología , Línea Celular Tumoral , Femenino , Humanos , Maitansina/análogos & derivados , Maitansina/farmacocinética , Maitansina/farmacología , Ratones , Ratones Desnudos , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
16.
Mol Cancer Ther ; 12(1): 38-47, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23223830

RESUMEN

Antibody-drug conjugates (ADC) represent a promising therapeutic modality for the clinical management of cancer. We sought to develop a novel ADC that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells (TIC), which comprise the most aggressive cell population in the tumor. We optimized an anti-5T4 ADC (A1mcMMAF) by sulfydryl-based conjugation of the humanized A1 antibody to the tubulin inhibitor monomethylauristatin F (MMAF) via a maleimidocaproyl linker. A1mcMMAF exhibited potent in vivo antitumor activity in a variety of tumor models and induced long-term regressions for up to 100 days after the last dose. Strikingly, animals showed pathologic complete response in each model with doses as low as 3 mg antibody/kg dosed every 4 days. In a non-small cell lung cancer patient-derived xenograft model, in which 5T4 is preferentially expressed on the less differentiated tumor cells, A1mcMMAF treatment resulted in sustained tumor regressions and reduced TIC frequency. These results highlight the potential of ADCs that target the most aggressive cell populations within tumors, such as TICs. In exploratory safety studies, A1mcMMAF exhibited no overt toxicities when administered to cynomolgus monkeys at doses up to 10 mg antibody/kg/cycle × 2 and displayed a half-life of 5 days. The preclinical efficacy and safety data established a promising therapeutic index that supports clinical testing of A1mcMMAF.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Glicoproteínas de Membrana/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Monoclonales Humanizados/toxicidad , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Macaca fascicularis , Masculino , Dosis Máxima Tolerada , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Inducción de Remisión , Distribución Tisular , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Pharm Sci ; 100(2): 402-10, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20862773

RESUMEN

This commentary is a reply to a recent article by Mahmood commenting on the authors' article on the use of fixed-exponent allometry in predicting human clearance. The commentary discusses eight issues that are related to criticisms made in Mahmood's article and examines the controversies (fixed-exponent vs. varying-exponent allometry) from the perspective of statistics and mathematics. The key conclusion is that any allometric method, which is to establish a power function based on a limited number of animal species and to extrapolate the resulting power function to human values (varying-exponent allometry), is infused with fundamental statistical errors.


Asunto(s)
Biometría , Farmacocinética , Animales , Disponibilidad Biológica , Humanos , Tasa de Depuración Metabólica , Modelos Biológicos , Especificidad de la Especie
18.
Rev. baiana saúde pública ; 34(Supl 1)jul.-dez. 2010.
Artículo en Portugués | LILACS | ID: lil-598697

RESUMEN

Introdução: Necrose avascular óssea NAV é a maior e mais debilitante complicação músculo-esquelética pós-transplante renal. A alta incidência de NAV no pós transplante renal tem sido historicamente associada a dosagens elevadas de corticosteroide. A introdução da terapia imunossupressora com ciclosporina A e tacrolimus permitiu a redução da dosagem de corticosteroide no pós-transplante renal e a incidência de NAV. Diversos estudos mostraram diferenças ainda não totalmente esclarecidas entre os fármacos imunossupressores a respeito do risco de desenvolvimento de NAV como efeito adverso. Objetivo Comparar o risco de NAV em pacientes transplantados renais que fizeram uso de tacrolimus ou ciclosporina A. Metodologia Realizou-se meta análise e, para a busca bibliográfica, foram utilizadas as bases de dados MedLine e Scielo. Resultados Os estudos incluídos foram dois ensaios clínicos randomizados controlados e um estudo de coorte retrospectivo. A análise dos três estudos incluídos na meta análise evidenciou que os pacientes tratados com ciclosporina A apresentavam maior frequência de NAV. A análise isolada do risco relativo de cada estudo mostrou uma redução do risco de desenvolvimento de necrose avascular no grupo tacrolimus, no entanto em apenas um dos estudos essa redução foi significativa. Conclusão A meta análise evidenciou que o uso de tacrolimus diminui o risco de desenvolvimento de osteonecrose avascular na terapia imunossupressora em transplantados renais.


Introduction: Bone avascular necrosis (BAN) is the largest and most debilitating musculoskeletal complication after renal transplantation. The high incidence of BAN in the post-kidney transplantation has been historically associated with high doses of corticosteroids. The introduction of immunosuppressive therapy with cyclosporin A and tacrolimus allowed a reduction in the dosage of corticosteroids after renal transplantation and in the incidence of BAN. Several studies have shown differences not yet fully understood among immunosuppressive drugs regarding the risk of BAN as adverse effects. Objective: To compare the risk of BAN in renal transplant patients who used tacrolimus or cyclosporine A. Methods: We conducted a meta-analysis, and for the literature search, we used the Medline and Scielo databases. Results: The two included studies were randomized controlled trials and a retrospective cohort study. The analysis of the three studies included in the meta-analysis showed that patients treated with cyclosporine A had higher frequency of BAN. A separate analysis of the relative risk of each study showed a reduced risk of developing avascular necrosis in the tacrolimus group, however in only one study this reduction was significant. Conclusion: The meta-analysis showed that the use of tacrolimus decreases the risk of avascular osteonecrosis in immunosuppressive therapy in kidney transplant patients.


Introducción: La Necrosis Avascular Ósea (NAV) es la mayor y más debilitante complicación músculo-esquelética post-transplante renal. La alta incidencia de NAV en el post-transplante renal ha sido historicamente asociada a dosajes elevados de corticosteroide. La introducción de la terapia imunosupresora con ciclosporina A y tacrolimus permitió la reducción del dosaje de corticosteroide en el post-transplante renal y la incidencia de NAV. Diversos estudios mostraron diferencias aún no totalmente esclarecidas entre los fármacos imunosupresores a respecto del riesgo de desarrollo de NAV como efecto adverso. Objetivo: Comparar el riesgo de NAV en pacientes transplantados renales que hicieron uso de tacrolimus o ciclosporina A. Metodología: Se realizó meta-análisis y, para la busca bibliográfica, fueron utilizadas las bases de datos MedLine y Scielo. Resultados: Los estudios incluidos fueron dos ensayos clínicos randomizados controlados y un estudio de corte retrospectivo. El análisis de los tres estudios incluidos en el meta-análisis evidenció que los pacientes tratados con ciclosporina A presentaban mayor frecuencia de NAV. El análisis aislado del riesgo relativo de cada estudio mostró una reducción del riesgo de desarrollo de necrosis avascular en el grupo tacrolimus, sin embargo, en apenas uno de los estudios esa reducción fue significativa. Conclusión: El meta-análisis evidenció que el uso de tacrolimus disminuyó el riesgo de desarrollo de osteonecrosis avascular en la terapia imunosupresora en transplantados renales.


Asunto(s)
Osteonecrosis , Tacrolimus/administración & dosificación , Insuficiencia Renal
19.
Bioanalysis ; 2(1): 81-94, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21083122

RESUMEN

BACKGROUND: To develop and validate a sensitive and novel bioanalytical method for measuring tigecycline concentrations in human skin using LC-MS/MS. RESULTS: The method utilizes addition of a stabilizing agent to the human skin or surrogate (human liver or rat skin), homogenization of human skin in a strong acidic-methanol extraction solvent, centrifugation of the skin suspension, filtration of the skin suspension supernatant, separation by LC (Polaris™ C18-A 50 × 2.0 mm), and detection of tigecycline by MS/MS. Linearity was 50-20,000 ng/g, using a sample size of 100 mg. The intra-and inter-day accuracy and precision of the assay met acceptance criteria. CONCLUSION: This method has been successfully applied to 17 incurred human skin samples from volunteers with surgical infections who received intravenous doses of tigecycline (100 mg initial loading dose and 50 mg every 12 h for at least 2 days). Tigecycline concentrations in these samples ranged from 185 to 2853 ng/g.


Asunto(s)
Antibacterianos/análisis , Cromatografía Liquida/métodos , Minociclina/análogos & derivados , Piel/química , Espectrometría de Masas en Tándem/métodos , Animales , Humanos , Hígado/química , Minociclina/análisis , Ratas , Ratas Sprague-Dawley , Tigeciclina
20.
J Pharm Biomed Anal ; 48(3): 866-75, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18692977

RESUMEN

Tigecycline (Tygacil,Wyeth) is a first-in-class, broad spectrum antibiotic with activity against multiple-resistant organisms. In order to address the unexpectedly low tigecycline concentrations in human bone samples analyzed using a LC/MS/MS method developed elsewhere, we have developed and validated a new and sensitive human bone assay for the quantitation of tigecycline using LC/MS/MS. The new method utilizes the addition of a stabilizing agent to the human bone sample, homogenization of human bone in a strong acidic-methanol extraction solvent, centrifugation of the bone suspension, separation by liquid chromatography, and detection of tigecycline by mass spectrometry. Linearity was demonstrated over the concentration range from 50 to 20,000 ng/g using a 0.1g human bone sample. The intra- and inter-day accuracy of the assay was within 100+/-15%, and the corresponding precision (CV) was <15%. The stability of tigecycline was evaluated and shown to be acceptable under the assay conditions. The extraction recovery of tigecycline with the current method was 79% when using radio-labeled rat bone samples as a substitute for human bone samples. Twenty-four human bone samples collected previously from volunteers without infections who had elective orthopedic surgery after receiving a single dose of tigecycline were re-analyzed using the current validated method. Tigecycline concentrations in these samples ranged from 238 to 794 ng/g with a mean value 9 times higher than the mean concentration previously reported. The data demonstrated that the current method has significantly higher extraction efficiency than the previously reported method.


Asunto(s)
Antibacterianos/análisis , Huesos/química , Cromatografía Liquida/métodos , Minociclina/análogos & derivados , Espectrometría de Masas en Tándem/métodos , Animales , Antibacterianos/química , Bioensayo , Calibración , Cromatografía Líquida de Alta Presión/instrumentación , Cromatografía Líquida de Alta Presión/métodos , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Minociclina/análisis , Minociclina/química , Estructura Molecular , Control de Calidad , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Solventes/química , Tigeciclina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...