Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Commun Biol ; 7(1): 497, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658677

RESUMEN

Most lung cancer patients with metastatic cancer eventually relapse with drug-resistant disease following treatment and EGFR mutant lung cancer is no exception. Genome-wide CRISPR screens, to either knock out or overexpress all protein-coding genes in cancer cell lines, revealed the landscape of pathways that cause resistance to the EGFR inhibitors osimertinib or gefitinib in EGFR mutant lung cancer. Among the most recurrent resistance genes were those that regulate the Hippo pathway. Following osimertinib treatment a subpopulation of cancer cells are able to survive and over time develop stable resistance. These 'persister' cells can exploit non-genetic (transcriptional) programs that enable cancer cells to survive drug treatment. Using genetic and pharmacologic tools we identified Hippo signalling as an important non-genetic mechanism of cell survival following osimertinib treatment. Further, we show that combinatorial targeting of the Hippo pathway and EGFR is highly effective in EGFR mutant lung cancer cells and patient-derived organoids, suggesting a new therapeutic strategy for EGFR mutant lung cancer patients.


Asunto(s)
Acrilamidas , Resistencia a Antineoplásicos , Receptores ErbB , Indoles , Neoplasias Pulmonares , Mutación , Pirimidinas , Factores de Transcripción , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Resistencia a Antineoplásicos/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Línea Celular Tumoral , Acrilamidas/farmacología , Acrilamidas/uso terapéutico , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Compuestos de Anilina/farmacología , Compuestos de Anilina/uso terapéutico , Gefitinib/farmacología , Vía de Señalización Hippo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transducción de Señal , Factores de Transcripción de Dominio TEA , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/farmacología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Sistemas CRISPR-Cas
2.
Cancer Discov ; 14(5): 846-865, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38456804

RESUMEN

Oncology drug combinations can improve therapeutic responses and increase treatment options for patients. The number of possible combinations is vast and responses can be context-specific. Systematic screens can identify clinically relevant, actionable combinations in defined patient subtypes. We present data for 109 anticancer drug combinations from AstraZeneca's oncology small molecule portfolio screened in 755 pan-cancer cell lines. Combinations were screened in a 7 × 7 concentration matrix, with more than 4 million measurements of sensitivity, producing an exceptionally data-rich resource. We implement a new approach using combination Emax (viability effect) and highest single agent (HSA) to assess combination benefit. We designed a clinical translatability workflow to identify combinations with clearly defined patient populations, rationale for tolerability based on tumor type and combination-specific "emergent" biomarkers, and exposures relevant to clinical doses. We describe three actionable combinations in defined cancer types, confirmed in vitro and in vivo, with a focus on hematologic cancers and apoptotic targets. SIGNIFICANCE: We present the largest cancer drug combination screen published to date with 7 × 7 concentration response matrices for 109 combinations in more than 750 cell lines, complemented by multi-omics predictors of response and identification of "emergent" combination biomarkers. We prioritize hits to optimize clinical translatability, and experimentally validate novel combination hypotheses. This article is featured in Selected Articles from This Issue, p. 695.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias , Humanos , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales/métodos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
3.
Artículo en Inglés | MEDLINE | ID: mdl-38532525

RESUMEN

Trastuzumab deruxtecan (T-DXd; DS-8201; ENHERTU®) is a human epithelial growth factor receptor 2 (HER2)-directed antibody drug conjugate (ADC) with demonstrated antitumor activity against a range of tumor types. Aiming to understand the relationship between antigen expression and downstream efficacy outcomes, T-DXd was administered in tumor-bearing mice carrying NCI-N87, Capan-1, JIMT-1, and MDA-MB-468 xenografts, characterized by varying HER2 levels. Plasma pharmacokinetics (PK) of total antibody, T-DXd, and released DXd and tumor concentrations of released DXd were evaluated, in addition to monitoring γΗ2AX and pRAD50 pharmacodynamic (PD) response. A positive relationship was observed between released DXd concentrations in tumor and HER2 expression, with NCI-N87 xenografts characterized by the highest exposures compared to the remaining cell lines. γΗ2AX and pRAD50 demonstrated a sustained increase over several days occurring with a time delay relative to tumoral-released DXd concentrations. In vitro investigations of cell-based DXd disposition facilitated the characterization of DXd kinetics across tumor cells. These outputs were incorporated into a mechanistic mathematical model, utilized to describe PK/PD trends. The model captured plasma PK across dosing arms as well as tumor PK in NCI-N87, Capan-1, and MDA-MB-468 models; tumor concentrations in JIMT-1 xenografts required additional parameter adjustments reflective of complex receptor dynamics. γΗ2AX longitudinal trends were well characterized via a unified PD model implemented across xenografts demonstrating the robustness of measured PD trends. This work supports the application of a mechanistic model as a quantitative tool, reliably projecting tumor payload concentrations upon T-DXd administration, as the first step towards preclinical-to-clinical translation.

4.
Blood Cancer Discov ; 5(2): 95-105, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38232314

RESUMEN

Combination therapy is an important part of cancer treatment and is often employed to overcome or prevent drug resistance. Preclinical screening strategies often prioritize synergistic drug combinations; however, studies of antibiotic combinations show that synergistic drug interactions can accelerate the emergence of resistance because resistance to one drug depletes the effect of both. In this study, we aimed to determine whether synergy drives the development of resistance in cancer cell lines using live-cell imaging. Consistent with prior models of tumor evolution, we found that when controlling for activity, drug synergy is associated with increased probability of developing drug resistance. We demonstrate that these observations are an expected consequence of synergy: the fitness benefit of resisting a drug in a combination is greater in synergistic combinations than in nonsynergistic combinations. These data have important implications for preclinical strategies aiming to develop novel combinations of cancer therapies with robust and durable efficacy. SIGNIFICANCE: Preclinical strategies to identify combinations for cancer treatment often focus on identifying synergistic combinations. This study shows that in AML cells combinations that rely on synergy can increase the likelihood of developing resistance, suggesting that combination screening strategies may benefit from a more holistic approach rather than focusing on drug synergy. See related commentary by Bhola and Letai, p. 81. This article is featured in Selected Articles from This Issue, p. 80.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Antibacterianos , Línea Celular , Terapia Combinada , Combinación de Medicamentos
5.
Sci Transl Med ; 12(541)2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32350132

RESUMEN

Gastrointestinal stromal tumor (GIST) is the most common human sarcoma driven by mutations in KIT or platelet-derived growth factor α (PDGFRα). Although first-line treatment, imatinib, has revolutionized GIST treatment, drug resistance due to acquisition of secondary KIT/PDGFRα mutations develops in a majority of patients. Second- and third-line treatments, sunitinib and regorafenib, lack activity against a plethora of mutations in KIT/PDGFRα in GIST, with median time to disease progression of 4 to 6 months and inhibition of vascular endothelial growth factor receptor 2 (VEGFR2) causing high-grade hypertension. Patients with GIST have an unmet need for a well-tolerated drug that robustly inhibits a range of KIT/PDGFRα mutations. Here, we report the discovery and pharmacological characterization of AZD3229, a potent and selective small-molecule inhibitor of KIT and PDGFRα designed to inhibit a broad range of primary and imatinib-resistant secondary mutations seen in GIST. In engineered and GIST-derived cell lines, AZD3229 is 15 to 60 times more potent than imatinib in inhibiting KIT primary mutations and has low nanomolar activity against a wide spectrum of secondary mutations. AZD3229 causes durable inhibition of KIT signaling in patient-derived xenograft (PDX) models of GIST, leading to tumor regressions at doses that showed no changes in arterial blood pressure (BP) in rat telemetry studies. AZD3229 has a superior potency and selectivity profile to standard of care (SoC) agents-imatinib, sunitinib, and regorafenib, as well as investigational agents, avapritinib (BLU-285) and ripretinib (DCC-2618). AZD3229 has the potential to be a best-in-class inhibitor for clinically relevant KIT/PDGFRα mutations in GIST.


Asunto(s)
Antineoplásicos , Tumores del Estroma Gastrointestinal , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Humanos , Mutación , Naftiridinas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/genética , Pirazoles , Pirroles , Ratas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Triazinas , Urea/análogos & derivados , Factor A de Crecimiento Endotelial Vascular
6.
CPT Pharmacometrics Syst Pharmacol ; 8(11): 858-868, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31508894

RESUMEN

Haematological toxicity associated with cancer therapeutics is monitored by changes in blood cell count, and their primary effect is on proliferative progenitors in the bone marrow. Using observations in rat bone marrow and blood, we characterize a mathematical model that comprises cell proliferation and differentiation of the full haematopoietic phylogeny, with interacting feedback loops between lineages in homeostasis as well as following carboplatin exposure. We accurately predicted the temporal dynamics of several mature cell types related to carboplatin-induced bone marrow toxicity and identified novel insights into haematopoiesis. Our model confirms a significant degree of plasticity within bone marrow cells, with the number and type of both early progenitors and circulating cells affecting cell balance, via feedback mechanisms, through fate decisions of the multipotent progenitors. We also demonstrated cross-species translation of our predictions to patients, applying the same core model structure and considering differences in drug-dependent and physiology-dependent parameters.


Asunto(s)
Médula Ósea/efectos de los fármacos , Carboplatino/toxicidad , Biología de Sistemas/métodos , Animales , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Homeostasis , Humanos , Modelos Teóricos , Ratas
7.
Toxicol Sci ; 166(1): 123-130, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30060248

RESUMEN

CKA, a chemokine receptor antagonist intended for treating inflammatory conditions, produced dose-dependent hepatotoxicity in rats but advanced into the clinic where single doses of CKA up to 600 mg appeared safe in humans. Because existing toxicological platforms used during drug development are not perfectly predictive, a quantitative systems toxicology model investigated the hepatotoxic potential of CKA in humans and rats through in vitro assessments of CKA on mitochondrial respiration, oxidative stress, and bile acid transporters. DILIsym predicted that single doses of CKA caused serum ALT >3xULN in a subset of the simulated rat population, while single doses in a simulated human population did not produce serum ALT elevations. Species differences were largely attributed to differences in liver exposure, but increased sensitivity to inhibition of mitochondrial respiration in the rat also contributed. We conclude that mechanistic modeling can elucidate species differences in the hepatotoxic potential of drug candidates.


Asunto(s)
Ácidos Carboxílicos/toxicidad , Proteínas Portadoras/antagonistas & inhibidores , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Indoles/toxicidad , Glicoproteínas de Membrana/antagonistas & inhibidores , Modelos Biológicos , Estrés Oxidativo/efectos de los fármacos , Receptores de Quimiocina/antagonistas & inhibidores , Adulto , Animales , Ácidos Carboxílicos/administración & dosificación , Ácidos Carboxílicos/farmacocinética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Células Hep G2 , Humanos , Indoles/administración & dosificación , Indoles/farmacocinética , Pruebas de Función Hepática , Masculino , Persona de Mediana Edad , Ratas Wistar , Especificidad de la Especie , Distribución Tisular
8.
Clin Pharmacol Ther ; 104(4): 644-654, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29604045

RESUMEN

Balancing antitumor efficacy with toxicity is a significant challenge, and drug-induced myelosuppression is a common dose-limiting toxicity of cancer treatments. Mathematical modeling has proven to be a powerful ally in this field, scaling results from animal models to humans, and designing optimized treatment regimens. Here we outline existing mathematical approaches for studying bone marrow toxicity, identify gaps in current understanding, and make future recommendations to advance this vital field of safety research further.


Asunto(s)
Antineoplásicos/efectos adversos , Médula Ósea/efectos de los fármacos , Enfermedades Hematológicas/inducido químicamente , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Modelos Biológicos , Pruebas de Toxicidad/métodos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Médula Ósea/patología , Médula Ósea/fisiopatología , Linaje de la Célula , Relación Dosis-Respuesta a Droga , Enfermedades Hematológicas/patología , Enfermedades Hematológicas/fisiopatología , Células Madre Hematopoyéticas/patología , Humanos , Medición de Riesgo
9.
CPT Pharmacometrics Syst Pharmacol ; 7(1): 26-33, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28941225

RESUMEN

Gastrointestinal (GI) adverse events (AEs) are frequently dose limiting for oncology agents, requiring extensive clinical testing of alternative schedules to identify optimal dosing regimens. Here, we develop a translational mathematical model to predict these clinical AEs starting from preclinical GI toxicity data. The model structure incorporates known biology and includes stem cells, daughter cells, and enterocytes. Published data, including cellular numbers and division times, informed the system parameters for humans and rats. The drug-specific parameters were informed with preclinical histopathology data from rats treated with irinotecan. The model fit the rodent irinotecan-induced pathology changes well. The predicted time course of enterocyte loss in patients treated with weekly doses matched observed AE profiles. The model also correctly predicts a lower level of AEs for every 3 weeks (Q3W), as compared to the weekly schedule.


Asunto(s)
Antineoplásicos/administración & dosificación , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Tracto Gastrointestinal/efectos de los fármacos , Irinotecán/administración & dosificación , Modelos Biológicos , Biología de Sistemas/métodos , Animales , Antineoplásicos/toxicidad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Tracto Gastrointestinal/fisiología , Humanos , Irinotecán/toxicidad , Valor Predictivo de las Pruebas , Especificidad de la Especie , Investigación Biomédica Traslacional
10.
Br J Pharmacol ; 175(4): 618-630, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29161763

RESUMEN

BACKGROUND AND PURPOSE: Several anti-angiogenic cancer drugs that inhibit VEGF receptor (VEGFR) signalling for efficacy are associated with a 15-60% incidence of hypertension. Tyrosine kinase inhibitors (TKIs) that have off-target activity at VEGFR-2 may also cause blood pressure elevation as an undesirable side effect. Therefore, the ability to translate VEGFR-2 off-target potency into blood pressure elevation would be useful in development of novel TKIs. Here, we have sought to quantify the relationship between VEGFR-2 inhibition and blood pressure elevation for a range of kinase inhibitors. EXPERIMENTAL APPROACH: Porcine aortic endothelial cells overexpressing VEGFR-2 (PAE) were used to determine IC50 for VEGFR-2 phosphorylation. These IC50 values were compared with published reports of exposure attained during clinical use and the corresponding incidence of all-grade hypertension. Unbound average plasma concentration (Cav,u ) was selected to be the most appropriate pharmacokinetic parameter. The pharmacokinetic-pharmacodynamic (PKPD) relationship for blood pressure elevation was investigated for selected kinase inhibitors, using data derived either from clinical papers or from rat telemetry experiments. KEY RESULTS: All-grade hypertension was predominantly observed when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . Furthermore, based on the PKPD analysis, an exposure-dependent blood pressure elevation >1 mmHg was observed only when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . CONCLUSIONS AND IMPLICATIONS: Taken together, these data show that the risk of blood pressure elevation is proportional to the amount of VEGFR-2 inhibition, and a margin of >10-fold between VEGFR-2 IC50 and Cav,u appears to confer a minimal risk of hypertension.


Asunto(s)
Inhibidores de la Angiogénesis/toxicidad , Presión Sanguínea/fisiología , Hipertensión/inducido químicamente , Inhibidores de Proteínas Quinasas/toxicidad , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Axitinib , Presión Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hipertensión/metabolismo , Imidazoles/toxicidad , Indazoles/toxicidad , Ratas , Porcinos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Eur J Pharm Sci ; 94: 93-95, 2016 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-27251780

RESUMEN

Ensuring that drugs are safe and effective is a very high priority for drug development and the US Food and Drug Administration review process. This is especially true today because of faster approval times and smaller clinical trials, especially in oncology and rare diseases. In light of these trends, systems pharmacology is seen as an essential strategy to understand and predict adverse drug events during drug development by analyzing interactions between drugs and multiple targets rather than the traditional "one-drug-one-target" approach. This commentary offers an overview of the current trends and challenges of using systems pharmacology to reduce the risks of unintended adverse events.


Asunto(s)
Descubrimiento de Drogas/tendencias , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Farmacología Clínica/tendencias , Biología de Sistemas/tendencias , Animales , Descubrimiento de Drogas/métodos , Predicción , Humanos , Farmacología Clínica/métodos , Biología de Sistemas/métodos
12.
AAPS J ; 17(2): 462-73, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25630504

RESUMEN

The application of modeling and simulation techniques is increasingly common in preclinical stages of the drug discovery and development process. A survey focusing on preclinical pharmacokinetic/pharmacodynamics (PK/PD) analysis was conducted across pharmaceutical companies that are members of the International Consortium for Quality and Innovation in Pharmaceutical Development. Based on survey responses, ~68% of companies use preclinical PK/PD analysis in all therapeutic areas indicating its broad application. An important goal of preclinical PK/PD analysis in all pharmaceutical companies is for the selection/optimization of doses and/or dose regimens, including prediction of human efficacious doses. Oncology was the therapeutic area with the most PK/PD analysis support and where it showed the most impact. Consistent use of more complex systems pharmacology models and hybrid physiologically based pharmacokinetic models with PK/PD components was less common compared to traditional PK/PD models. Preclinical PK/PD analysis is increasingly being included in regulatory submissions with ~73% of companies including these data to some degree. Most companies (~86%) have seen impact of preclinical PK/PD analyses in drug development. Finally, ~59% of pharmaceutical companies have plans to expand their PK/PD modeling groups over the next 2 years indicating continued growth. The growth of preclinical PK/PD modeling groups in pharmaceutical industry is necessary to establish required resources and skills to further expand use of preclinical PK/PD modeling in a meaningful and impactful manner.


Asunto(s)
Simulación por Computador , Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/métodos , Modelos Biológicos , Recolección de Datos , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Descubrimiento de Drogas/métodos , Industria Farmacéutica/estadística & datos numéricos , Humanos
13.
PLoS One ; 9(10): e109892, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25360756

RESUMEN

Toxicity often limits the utility of oncology drugs, and optimization of dose schedule represents one option for mitigation of this toxicity. Here we explore the schedule-dependency of neutropenia, a common dose-limiting toxicity. To this end, we analyze previously published mathematical models of neutropenia to identify a pharmacokinetic (PK) predictor of the neutrophil nadir, and confirm this PK predictor in an in vivo experimental system. Specifically, we find total AUC and Cmax are poor predictors of the neutrophil nadir, while a PK measure based on the moving average of the drug concentration correlates highly with neutropenia. Further, we confirm this PK parameter for its ability to predict neutropenia in vivo following treatment with different doses and schedules. This work represents an attempt at mechanistically deriving a fundamental understanding of the underlying pharmacokinetic drivers of neutropenia, and provides insights that can be leveraged in a translational setting during schedule selection.


Asunto(s)
Antineoplásicos/efectos adversos , Esquema de Medicación , Modelos Biológicos , Neutropenia/inducido químicamente , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Docetaxel , Etopósido/administración & dosificación , Etopósido/efectos adversos , Etopósido/farmacocinética , Humanos , Masculino , Modelos Teóricos , Ratas Sprague-Dawley , Taxoides/administración & dosificación , Taxoides/efectos adversos , Taxoides/farmacocinética , Topotecan/administración & dosificación , Topotecan/efectos adversos , Topotecan/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA