Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Pharm ; 18(11): 4058-4066, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34579528

RESUMEN

To overcome a lack of selectivity during the chemical modification of native non-engineered antibodies, we have developed a technology platform termed "AJICAP" for the site-specific chemical conjugation of antibodies through the use of a class of IgG Fc-affinity reagents. To date, a limited number of antibody-drug conjugates (ADCs) have been synthesized via this approach, and no toxicological study was reported. Herein, we describe the compatibility and robustness of AJICAP technology, which enabled the synthesis of a wide variety of ADCs. A stability assessment of a thiol-modified antibody synthesized by AJICAP technology indicated no appreciable increase in aggregation or decomposition upon prolonged storage, indicating that the unexpectedly stable thiol intermediate has a great potential intermediate for payload or linker screening or large-scale manufacturing. Payload conjugation with this stable thiol intermediate generated several AJICAP-ADCs. In vivo xenograft studies indicated that the AJICAP-ADCs displayed significant tumor inhibition comparable to benchmark ADC Kadcyla. Furthermore, a rat pharmacokinetic analysis and toxicology study indicated an increase in the maximum tolerated dose, demonstrating an expansion of the AJICAP-ADC therapeutic index, compared with stochastic conjugation technology. This is the first report of the therapeutic index estimation of site-specific ADCs produced by utilizing Fc affinity reagent conjugation. The described site-specific conjugation technology is a powerful platform to enable next-generation ADCs through reduced heterogeneity and enhanced therapeutic index.


Asunto(s)
Antineoplásicos/farmacocinética , Composición de Medicamentos/métodos , Inmunoconjugados/farmacocinética , Neoplasias/tratamiento farmacológico , Ado-Trastuzumab Emtansina/administración & dosificación , Ado-Trastuzumab Emtansina/farmacocinética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/toxicidad , Química Farmacéutica , Estabilidad de Medicamentos , Femenino , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Inmunoconjugados/toxicidad , Dosis Máxima Tolerada , Ratones , Neoplasias/patología , Ratas , Índice Terapéutico , Pruebas de Toxicidad Aguda , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cancer Treat Res Commun ; 27: 100314, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33545569

RESUMEN

HER2-positive metastatic breast cancer is an aggressive disease with a limited number of treatment options. In the last 15 years, new drugs such as trastuzumab, pertuzumab, lapatinib or trastuzumab emtansine (TDM-1) have sprouted for these patients. There is a huge lack of evidence on the use of some of these drugs in patients with chronic renal failure, who need hemodialysis. We have reviewed the use of TDM-1 in these type of patients in the literature with unsuccessful results. In this article we want to present a case report to illustrate the safety and efficacy of TDM-1 in a patient on hemodialysis.


Asunto(s)
Ado-Trastuzumab Emtansina/administración & dosificación , Anemia/inducido químicamente , Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Fallo Renal Crónico/terapia , Ado-Trastuzumab Emtansina/efectos adversos , Ado-Trastuzumab Emtansina/farmacocinética , Anciano , Anemia/sangre , Anemia/diagnóstico , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/farmacocinética , Neoplasias de la Mama/sangre , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/patología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Fallo Renal Crónico/sangre , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/fisiopatología , Receptor ErbB-2/análisis , Receptor ErbB-2/metabolismo , Diálisis Renal/efectos adversos , Eliminación Renal/fisiología , Resultado del Tratamiento
3.
Clin Transl Sci ; 14(1): 395-404, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33073529

RESUMEN

Antibody drug conjugates (ADCs) provide targeted delivery of cytotoxic agents directly inside tumor cells. However, many ADCs targeting solid tumors have exhibited limited clinical efficacy, in part, due to insufficient penetration within tumors. To better understand the relationship between ADC tumor penetration and efficacy, previously applied Krogh cylinder models that explore tumor growth dynamics following ADC administration in preclinical species were expanded to a clinical framework by integrating clinical pharmacokinetics, tumor penetration, and tumor growth inhibition. The objective of this framework is to link ADC tumor penetration and distribution to clinical efficacy. The model was validated by comparing virtual patient population simulations to observed overall response rates from trastuzumab-DM1 treated patients with metastatic breast cancer. To capture clinical outcomes, we expanded upon previous Krogh cylinder models to include the additional mechanism of heterogeneous tumor growth inhibition spatially across the tumor. This expansion mechanistically captures clinical response rates by describing heterogeneous ADC binding and tumor cell killing; high binding and tumor cell death close to capillaries vs. low binding, and high tumor cell proliferation far from capillaries. Sensitivity analyses suggest that clinical efficacy could be optimized through dose fractionation, and that clinical efficacy is primarily dependent on the ADC-target affinity, payload potency, and tumor growth rate. This work offers a mechanistic basis to predict and optimize ADC clinical efficacy for solid tumors, allowing dosing strategy optimization to improve patient outcomes.


Asunto(s)
Ado-Trastuzumab Emtansina/farmacocinética , Antineoplásicos/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Inmunoconjugados/farmacocinética , Modelos Biológicos , Ado-Trastuzumab Emtansina/administración & dosificación , Antineoplásicos/administración & dosificación , Área Bajo la Curva , Mama/patología , Neoplasias de la Mama/patología , Femenino , Humanos , Inmunoconjugados/administración & dosificación , Distribución Tisular , Carga Tumoral
4.
Mol Cancer Ther ; 19(9): 1866-1874, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32651200

RESUMEN

Trastuzumab and the related ADC, ado-trastuzumab emtansine (T-DM1), both target HER2-overexpressing cells. Together, these drugs have treatment indications in both early-stage and metastatic settings for HER2+ breast cancer. T-DM1 retains the antibody functionalities of trastuzumab and adds the potency of a cytotoxic maytansine payload. Interestingly, in the clinic, T-DM1 cannot always replace the use of trastuzumab plus chemotherapy administered together as single agents. We hypothesize that this failure may be due, in part, to the limited systemic exposure achieved by T-DM1 relative to trastuzumab because of toxicity-related dosing constraints on the ADC. We have developed a trastuzumab-based ADC site specifically conjugated to maytansine through a noncleavable linker. This construct, termed CAT-01-106, has a drug-to-antibody ratio (DAR) of 1.8, approximately half the average DAR of T-DM1, which comprises a mixture of antibodies variously conjugated with DARs ranging from 0 to 8. The high DAR species present in T-DM1 contribute to its toxicity and limit its clinical dose. CAT-01-106 showed superior in vivo efficacy compared with T-DM1 at equal payload dosing and was equally or better tolerated compared with T-DM1 at equal payload dosing up to 120 mg/kg in Sprague-Dawley rats and 60 mg/kg in cynomolgus monkeys. CAT-01-106 also showed improved pharmacokinetics in rats relative to T-DM1, with 40% higher ADC exposure levels. Together, the data suggest that CAT-01-106 may be sufficiently tolerable to enable clinical dosing at trastuzumab-equivalent exposure levels, combining the functions of both the antibody and the payload in one drug and potentially improving patient outcomes.


Asunto(s)
Ado-Trastuzumab Emtansina/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Inmunoconjugados/administración & dosificación , Maitansina/química , Trastuzumab/química , Ado-Trastuzumab Emtansina/efectos adversos , Ado-Trastuzumab Emtansina/farmacocinética , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/efectos adversos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Macaca fascicularis , Dosis Máxima Tolerada , Ratas , Ratas Sprague-Dawley , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Pharmacokinet Pharmacodyn ; 47(5): 513-526, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32710210

RESUMEN

A modeling and simulation approach was used for quantitative comparison of a new generation HER2 antibody drug conjugate (ADC, PF-06804103) with trastuzumab-DM1 (T-DM1). To compare preclinical efficacy, the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of PF-06804103 and T-DM1 was determined across a range of mouse tumor xenograft models, using a tumor growth inhibition model. The tumor static concentration was assigned as the minimal efficacious concentration. PF-06804103 was concluded to be more potent than T-DM1 across cell lines studied. TSCs ranged from 1.0 to 9.8 µg/mL (n = 7) for PF-06804103 and from 4.7 to 29 µg/mL (n = 5) for T-DM1. Two experimental models which were resistant to T-DM1, responded to PF-06804103 treatment. A mechanism-based target mediated drug disposition (TMDD) model was used to predict the human PK of PF-06804103. This model was constructed and validated based on T-DM1 which has non-linear PK at doses administered in the clinic, driven by binding to shed HER2. Non-linear PK is predicted for PF-06804103 in the clinic and is dependent upon circulating HER2 extracellular domain (ECD) concentrations. The models were translated to human and suggested greater efficacy for PF-06804103 compared to T-DM1. In conclusion, a fit-for-purpose translational PK/PD strategy for ADCs is presented and used to compare a new generation HER2 ADC with T-DM1.


Asunto(s)
Ado-Trastuzumab Emtansina/farmacocinética , Antineoplásicos Inmunológicos/farmacocinética , Inmunoconjugados/farmacocinética , Neoplasias/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Administración Intravenosa , Ado-Trastuzumab Emtansina/administración & dosificación , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Línea Celular Tumoral , Simulación por Computador , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunoconjugados/administración & dosificación , Macaca fascicularis , Masculino , Ratones , Modelos Biológicos , Neoplasias/patología , Receptor ErbB-2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Br J Cancer ; 123(6): 1000-1011, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32572172

RESUMEN

BACKGROUND: Trastuzumab-emtansine (T-DM1), one of the most potent HER2-targeted drugs, shows impressive efficacy in patients with HER2-positive breast cancers. However, resistance inevitably occurs and becomes a critical clinical problem. METHODS: We modelled the development of acquired resistance by exposing HER2-positive cells to escalating concentrations of T-DM1. Signalling pathways activation was detected by western blotting, gene expression was analysed by qRT-PCR and gene copy numbers were determined by qPCR. The role of Yes on resistance was confirmed by siRNA-mediated knockdown and stable transfection-mediated overexpression. The in vivo effects were tested in xenograft model. RESULTS: We found that Yes is overexpressed in T-DM1-resistant cells owing to amplification of chromosome region 18p11.32, where the YES1 gene resides. Yes activated multiple proliferation-related signalling pathways, including EGFR, PI3K and MAPK, and led to cross-resistance to all types of HER2-targeted drugs, including antibody-drug conjugate, antibody and small molecule inhibitor. The outcome of this cross-resistance may be a clinically incurable condition. Importantly, we found that inhibiting Yes with dasatinib sensitised resistant cells in vitro and in vivo. CONCLUSIONS: Our study revealed that YES1 amplification conferred resistance to HER2-targeted drugs and suggested the potential application of the strategy of combining HER2 and Yes inhibition in the clinic.


Asunto(s)
Ado-Trastuzumab Emtansina/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Amplificación de Genes , Proteínas Proto-Oncogénicas c-yes/genética , Receptor ErbB-2/análisis , Ado-Trastuzumab Emtansina/farmacocinética , Animales , Neoplasias de la Mama/química , Neoplasias de la Mama/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-yes/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Familia-src Quinasas/fisiología
7.
AAPS J ; 22(2): 29, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31942650

RESUMEN

The pharmaceutical industry has invested significantly in antibody-drug conjugates (ADCs) with five FDA-approved therapies and several more showing promise in late-stage clinical trials. The FDA-approved therapeutic Kadcyla (ado-trastuzumab emtansine or T-DM1) can extend the survival of patients with tumors overexpressing HER2. However, tumor histology shows that most T-DM1 localizes perivascularly, but coadministration with its unconjugated form (trastuzumab) improves penetration of the ADC into the tumor and subsequent treatment efficacy. ADC dosing schedule, e.g., dose fractionation, has also been shown to improve tolerability. However, it is still not clear how coadministration with carrier doses impacts efficacy in terms of receptor expression, dosing regimens, and payload potency. Here, we develop a hybrid agent-based model (ABM) to capture ADC and/or antibody delivery and to predict tumor killing and growth kinetics. The results indicate that a carrier dose improves efficacy when the increased number of cells targeted by the ADC outweighs the reduced fractional killing of the targeted cells. The threshold number of payloads per cell required for killing plays a pivotal role in defining this cutoff. Likewise, fractionated dosing lowers ADC efficacy due to lower tissue penetration from a reduced maximum plasma concentration. It is only beneficial when an increase in tolerability from fractionation allows a higher ADC/payload dose that more than compensates for the loss in efficacy from fractionation. Overall, the multiscale model enables detailed depictions of heterogeneous ADC delivery, cancer cell death, and tumor growth to show how carrier dosing impacts efficacy to design the most efficacious regimen.


Asunto(s)
Ado-Trastuzumab Emtansina/administración & dosificación , Ado-Trastuzumab Emtansina/farmacocinética , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/farmacocinética , Inmunoconjugados/administración & dosificación , Inmunoconjugados/farmacocinética , Modelos Biológicos , Neoplasias Gástricas/tratamiento farmacológico , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Simulación por Computador , Relación Dosis-Respuesta a Droga , Composición de Medicamentos , Femenino , Ratones Desnudos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Distribución Tisular , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
AAPS J ; 22(2): 28, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31938899

RESUMEN

It has been proposed that the binding-site barrier (BSB) for antibody-drug conjugates (ADCs) can be overcome with the help of antibody coadministration. However, broad utility of this strategy remains in question. Consequently, here, we have conducted in vivo experiments and pharmacokinetics-pharmacodynamics (PK-PD) modeling and simulation (M&S) to further evaluate the antibody coadministration hypothesis in a quantitative manner. Two different Trastuzumab-based ADCs, T-DM1 (no bystander effect) and T-vc-MMAE (with a bystander effect), were evaluated in high-HER2 (N87) and low-HER2 (MDA-MB-453) expressing tumors, with or without the coadministration of 1, 3, or 8-fold higher Trastuzumab. The tumor growth inhibition (TGI) data was quantitatively characterized using a semi-mechanistic PK-PD model to determine the nature of drug interaction for each coadministration regimen, by estimating the interaction parameter ψ. It was found that the coadministration strategy improved ADC efficacy under certain conditions and had no impact on ADC efficacy in others. The benefit was more pronounced for N87 tumors with very high antigen expression levels where the effect on treatment was synergistic (a synergistic drug interaction, ψ = 2.86 [2.6-3.12]). The benefit was diminished in tumor with lower antigen expression (MDA-MB-453) and payload with bystander effect. Under these conditions, the coadministration regimens resulted in an additive or even less than additive benefit (ψ ≤ 1). As such, our results suggest that while antibody coadministration may be helpful for ADCs in certain circumstances, one should not broadly apply this strategy to all the scenarios without first identifying the costs and benefits of this approach.


Asunto(s)
Ado-Trastuzumab Emtansina/farmacocinética , Antineoplásicos Inmunológicos/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Inmunoconjugados/farmacocinética , Modelos Biológicos , Oligopéptidos/farmacocinética , Receptor ErbB-2/antagonistas & inhibidores , Neoplasias Gástricas/tratamiento farmacológico , Trastuzumab/farmacocinética , Ado-Trastuzumab Emtansina/administración & dosificación , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Efecto Espectador , Línea Celular Tumoral , Simulación por Computador , Composición de Medicamentos , Femenino , Humanos , Inmunoconjugados/administración & dosificación , Masculino , Ratones Endogámicos NOD , Ratones SCID , Oligopéptidos/administración & dosificación , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/metabolismo , Distribución Tisular , Trastuzumab/administración & dosificación , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Drug Metab Dispos ; 47(10): 1146-1155, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31358513

RESUMEN

Antibody-drug conjugates (ADCs) contain a disease-receptor antibody and a payload drug connected via a linker. The payload delivery depends on both tumor properties and ADC characteristics. In this study, we used different linkers, attachment sites, and doses to modulate payload delivery of several ADCs bearing maytansinoids (e.g., DM1), auristatins (e.g., MMAE), and DNA alkylating agents [e.g., pyrrolo[2,1-c][1,4]benzodiazepine-dimer (PBD)] as payloads in HER2- or CD22-expressing xenograft models. The tumor growth inhibition and ADC stability and exposure data were collected and analyzed from these dosed animals. The trend analysis suggests that intratumoral payload exposures that directly related the combination of conjugate linker and dose correlate with the corresponding efficacies of three payload types in two antigen-expressing xenograft models. These preliminary correlations also suggest that a minimal threshold concentration of intratumoral payload is required to support sustained efficacy. In addition, an ADC can deliver an excessive level of payload to tumors that does not enhance efficacy ("Plateau" effect). In contrast to tumor payload concentrations, the assessments of systemic exposures of total antibody (Tab) as well as the linker, dose, site of attachment, plasma stability, and drug-to-antibody ratio changes of these ADCs did not consistently rationalize the observed ADC efficacies. The requirement of a threshold payload concentration for efficacy is further supported by dose fractionation studies with DM1-, MMAE-, and PBD-containing ADCs, which demonstrated that single-dose regimens showed better efficacies than fractionated dosing. Overall, this study demonstrates that 1) the linker and dose together determine the tissue payload concentration that correlates with the antitumor efficacy of ADCs and 2) an ADC can deliver an unnecessary level of payload to tumors in xenograft models.


Asunto(s)
Antineoplásicos Inmunológicos/farmacocinética , Inmunoconjugados/farmacocinética , Receptor ErbB-2/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Ado-Trastuzumab Emtansina/administración & dosificación , Ado-Trastuzumab Emtansina/farmacocinética , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/química , Benzodiazepinas/química , Brentuximab Vedotina/administración & dosificación , Brentuximab Vedotina/farmacocinética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunoconjugados/administración & dosificación , Ratones , Ratones Transgénicos , Pirroles/química , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Chemother Pharmacol ; 84(1): 175-185, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31102024

RESUMEN

PURPOSE: The phase III MARIANNE study investigated single-agent trastuzumab emtansine (T-DM1) and combination T-DM1 plus pertuzumab as the first-line treatment for human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC). Pharmacokinetic properties of T-DM1 and pertuzumab in these patients and the potential for drug-drug interactions (DDIs) were assessed. METHODS: Pharmacokinetic samples of T-DM1-related analytes (T-DM1 conjugate, total trastuzumab, DM1) and pertuzumab were analyzed. Observed pharmacokinetic data were summarized for all analytes. Historical population pharmacokinetic models for T-DM1 conjugate and pertuzumab in HER2-positive MBC were used to derive empirical Bayes estimates of pharmacokinetic parameters. RESULTS: In MARIANNE (N = 375), mean ± standard deviation population pharmacokinetic model-predicted Cycle 1 Cmax for T-DM1 conjugate was 74.4 ± 10.1 µg/mL, Cycle 1 Ctrough was 1.34 ± 0.802 µg/mL, and area under the concentration-time curve from time zero to infinity after first dose (AUCinf) was 338 ± 69.5 µg*day/mL. These values were similar to other T-DM1 studies. Pharmacokinetics of T-DM1 conjugate and other analytes (total trastuzumab, DM1) were similar with or without pertuzumab. In the pertuzumab plus T-DM1 arm, mean model-predicted Cycle 1 pertuzumab Cmax, Ctrough, and AUCinf were 276 ± 50.0 µg/mL, 64.8 ± 17.9 µg/mL, and 4470 ± 1360 µg*day/mL, respectively. These values were similar to other pertuzumab studies. CONCLUSIONS: Based on the population pharmacokinetic analysis of T-DM1 conjugate and pertuzumab, pharmacokinetics are similar across different lines of treatment and stages of disease including previously untreated MBC patients, and no DDIs were identified for combined use of T-DM1 and pertuzumab.


Asunto(s)
Ado-Trastuzumab Emtansina/administración & dosificación , Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Receptor ErbB-2/metabolismo , Ado-Trastuzumab Emtansina/farmacocinética , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos Inmunológicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Bajo la Curva , Neoplasias de la Mama/patología , Interacciones Farmacológicas , Femenino , Humanos , Modelos Biológicos , Recurrencia Local de Neoplasia , Estadificación de Neoplasias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...