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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biopharm Stat ; 34(2): 205-221, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36988397

RESUMEN

For multiple rare diseases as defined by a common biomarker signature, or a disease with multiple disease subtypes of low frequency, it is often possible to provide confirmatory evidence for these disease or subtypes (baskets) as a combined group. A novel drug, as a second generation, may have marginal improvement in efficacy overall but superior efficacy in some baskets. In this situation, it is appealing to test hypotheses of both non-inferiority overall and superiority on certain baskets. The challenge is designing a confirmatory study efficient to address multiple questions in one trial. A two-stage adaptive design is proposed to test the non-inferiority hypothesis at the interim stage, followed by pruning and pooling before testing a superiority hypothesis at the final stage. Such a design enables an efficient and novel registration pathway, including an early claim of non-inferiority followed by a potential label extension with superiority on certain baskets and an improved benefit-risk profile demonstrated by longer term efficacy and safety data. Operating characteristics of this design are examined by simulation studies, and its appealing features make it ready for use in a confirmatory setting, especially in emerging markets, where both the need and the possibility for efficient use of resources may be the greatest.


Asunto(s)
Proyectos de Investigación , Humanos , Simulación por Computador
2.
J Immunol ; 205(7): 1962-1977, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32878910

RESUMEN

The reliable prediction of the affinity of candidate peptides for the MHC is important for predicting their potential antigenicity and thus influences medical applications, such as decisions on their inclusion in T cell-based vaccines. In this study, we present a rapid, predictive computational approach that combines a popular, sequence-based artificial neural network method, NetMHCpan 4.0, with three-dimensional structural modeling. We find that the ensembles of bound peptide conformations generated by the programs MODELLER and Rosetta FlexPepDock are less variable in geometry for strong binders than for low-affinity peptides. In tests on 1271 peptide sequences for which the experimental dissociation constants of binding to the well-characterized murine MHC allele H-2Db are known, by applying thresholds for geometric fluctuations the structure-based approach in a standalone manner drastically improves the statistical specificity, reducing the number of false positives. Furthermore, filtering candidates generated with NetMHCpan 4.0 with the structure-based predictor led to an increase in the positive predictive value (PPV) of the peptides correctly predicted to bind very strongly (i.e., K d < 100 nM) from 40 to 52% (p = 0.027). The combined method also significantly improved the PPV when tested on five human alleles, including some with limited data for training. Overall, an average increase of 10% in the PPV was found over the standalone sequence-based method. The combined method should be useful in the rapid design of effective T cell-based vaccines.


Asunto(s)
Antígenos/metabolismo , Antígeno de Histocompatibilidad H-2D/metabolismo , Péptidos/metabolismo , Algoritmos , Animales , Antígenos/química , Antígenos/inmunología , Inteligencia Artificial , Biología Computacional , Cristalografía por Rayos X , Antígeno de Histocompatibilidad H-2D/química , Humanos , Ratones , Modelos Moleculares , Conformación Molecular , Péptidos/química , Péptidos/inmunología , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
3.
Proc Natl Acad Sci U S A ; 116(52): 26863-26872, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31806761

RESUMEN

Human colorectal cancers (CRCs) contain both clonal and subclonal mutations. Clonal driver mutations are positively selected, present in most cells, and drive malignant progression. Subclonal mutations are randomly dispersed throughout the genome, providing a vast reservoir of mutant cells that can expand, repopulate the tumor, and result in the rapid emergence of resistance, as well as being a major contributor to tumor heterogeneity. Here, we apply duplex sequencing (DS) methodology to quantify subclonal mutations in CRC tumor with unprecedented depth (104) and accuracy (<10-7). We measured mutation frequencies in genes encoding replicative DNA polymerases and in genes frequently mutated in CRC, and found an unexpectedly high effective mutation rate, 7.1 × 10-7. The curve of subclonal mutation accumulation as a function of sequencing depth, using DNA obtained from 5 different tumors, is in accord with a neutral model of tumor evolution. We present a theoretical approach to model neutral evolution independent of the infinite-sites assumption (which states that a particular mutation arises only in one tumor cell at any given time). Our analysis indicates that the infinite-sites assumption is not applicable once the number of tumor cells exceeds the reciprocal of the mutation rate, a circumstance relevant to even the smallest clinically diagnosable tumor. Our methods allow accurate estimation of the total mutation burden in clinical cancers. Our results indicate that no DNA locus is wild type in every malignant cell within a tumor at the time of diagnosis (probability of all cells being wild type, 10-308).

4.
Cancer Control ; 27(1): 1073274820962008, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32991214

RESUMEN

Choosing and optimizing treatment strategies for cancer requires capturing its complex dynamics sufficiently well for understanding but without being overwhelmed. Mathematical models are essential to achieve this understanding, and we discuss the challenge of choosing the right level of complexity to address the full range of tumor complexity from growth, the generation of tumor heterogeneity, and interactions within tumors and with treatments and the tumor microenvironment. We discuss the differences between conceptual and descriptive models, and compare the use of predator-prey models, evolutionary game theory, and dynamic precision medicine approaches in the face of uncertainty about mechanisms and parameter values. Although there is of course no one-size-fits-all approach, we conclude that broad and flexible thinking about cancer, based on combined modeling approaches, will play a key role in finding creative and improved treatments.


Asunto(s)
Evolución Biológica , Teoría del Juego , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/patología , Humanos , Neoplasias/genética , Dinámica Poblacional , Microambiente Tumoral
5.
J Theor Biol ; 474: 88-102, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31077681

RESUMEN

Despite recent advances in targeted drugs and immunotherapy, cancer remains "the emperor of all maladies" due to almost inevitable emergence of resistance. Drug resistance is thought to be driven by genetic alterations and/or dynamic plasticity that deregulate pathway activities and regulatory programs of a highly heterogeneous tumour. In this study, we propose a modelling framework to simulate population dynamics of heterogeneous tumour cells with reversible drug resistance. Drug sensitivity of a tumour cell is determined by its internal states, which are demarcated by coordinated activities of multiple interconnected oncogenic pathways. Transitions between cellular states depend on the effects of targeted drugs and regulatory relations between the pathways. Under this framework, we build a simple model to capture drug resistance characteristics of BRAF-mutant melanoma, where two cell states are determined by two mutually inhibitory - main and alternative - pathways. We assume that cells with an activated main pathway are proliferative yet sensitive to the BRAF inhibitor, and cells with an activated alternative pathway are quiescent but resistant to the drug. We describe a dynamical process of tumour growth under various drug regimens using the explicit solutions of mean-field equations. Based on these solutions, we compare efficacy of three treatment strategies from simulated data: static treatments with continuous and constant dosages, periodic treatments with regular intermittent active phases and drug holidays, and treatments derived from optimal control theory (OCT). Periodic treatments outperform static treatments with a considerable margin, while treatments based on OCT outperform the best periodic treatment. Our results provide insights regarding optimal cancer treatment modalities for heterogeneous tumours, and may guide the development of optimal therapeutic strategies to circumvent plastic drug resistance. They can also be used to evaluate the efficacy of suboptimal treatments that may account for side effects of the treatment and the cost of its application.


Asunto(s)
Resistencia a Antineoplásicos , Melanoma , Modelos Biológicos , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf , Humanos , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Melanoma/genética , Melanoma/patología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo
6.
Stat Med ; 38(29): 5470-5485, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31621949

RESUMEN

As biomarker information from early-phase trials can be unreliable due to high variability, it is logical to take a prospective two-stage approach when designing a late-phase confirmatory trial, ie, refining the target population at the first stage and performing the hypothesis testing at the second stage. The use of a reliable intermediate endpoint at the first stage can further improve the trial efficiency from both time and cost perspectives. Nevertheless, there are needs for expanding such two-stage confirmatory designs to more stages for monitoring efficacy on the refined population. There is limited literature on this matter, particularly for two popular designs with population selection midway, ie, the biomarker enrichment design and the basket design. In this manuscript, we focus on these two popular designs and discuss how to implement the interim efficacy analyses after population refinement while controlling type I error. Power and stopping probability are also explored for the two designs.


Asunto(s)
Ensayos Clínicos como Asunto/métodos , Ensayos Clínicos Adaptativos como Asunto/métodos , Ensayos Clínicos Adaptativos como Asunto/estadística & datos numéricos , Biomarcadores/análisis , Bioestadística , Carcinoma de Pulmón de Células no Pequeñas/terapia , Ensayos Clínicos como Asunto/estadística & datos numéricos , Ensayos Clínicos Fase III como Asunto/métodos , Ensayos Clínicos Fase III como Asunto/estadística & datos numéricos , Determinación de Punto Final , Humanos , Neoplasias Pulmonares/terapia , Modelos Estadísticos , Probabilidad , Supervivencia sin Progresión , Estudios Prospectivos , Análisis de Supervivencia
7.
Stat Med ; 36(12): 1843-1861, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28303586

RESUMEN

A personalized medicine may benefit a subpopulation with certain predictive biomarker signatures or certain disease types. However, there is great uncertainty about drug activity in a subpopulation when designing a confirmatory trial in practice, and it is logical to take a two-stage approach with the study unless credible external information is available for decision-making purpose. The first stage deselects (or prunes) non-performing subpopulations at an interim analysis, and the second stage pools the remaining subpopulations in the final analysis. The endpoints used at the two stages can be different in general. A key issue of interest is the statistical property of the test statistics and point estimate at the final analysis. Previous research has focused on type I error control and power calculation for such two-stage designs. This manuscript will investigate estimation bias of the treatment effect, which is implicit in the adjustment of nominal type I error for multiplicity control in such two-stage designs. Previous work handles the treatment effect of an intermediate endpoint as a nuisance parameter to provide the most conservative type I error control. This manuscript takes the same approach to explore the bias. The methodology is applied to the two previously studied designs. In the first design, patients with different biomarker levels are enrolled in a study, and the treatment effect is assumed to be in an order. The goal of the interim analysis is to identify a biomarker cut-off point for the subpopulations. In the second design, patients with different tumour types but the same biomarker signature are included in a trial applying a basket design. The goal of the interim analysis is to identify a subset of tumour types in the absence of treatment effect ordering. Closed-form equations are provided for the estimation bias as well as the variance under the two designs. Simulations are conducted under various scenarios to validate the analytic results that demonstrated that the bias can be properly estimated in practice. Worked examples are presented. Extensions to general adaptive designs and operational considerations are discussed. Copyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Interpretación Estadística de Datos , Determinación de Punto Final/métodos , Neoplasias/tratamiento farmacológico , Medicina de Precisión/métodos , Sesgo , Biomarcadores/análisis , Humanos , Modelos Estadísticos , Neoplasias/diagnóstico , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Proyectos de Investigación , Resultado del Tratamiento
8.
J Hepatol ; 63(4): 896-904, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26071796

RESUMEN

BACKGROUND & AIMS: Tigatuzumab is a humanized monoclonal antibody that acts as a death receptor-5 agonist and exerts tumour necrosis factor-related apoptosis-inducing ligand-like activity. In this phase II study, safety and tolerability of the combination of tigatuzumab and sorafenib was evaluated in patients with advanced hepatocellular carcinoma. METHODS: Adults with advanced hepatocellular carcinoma, measurable disease, and an Eastern Cooperative Oncology Group performance score⩽1 were enrolled. Eligible subjects were randomly assigned 1:1:1 to tigatuzumab (6 mg/kg loading, 2 mg/kg/week maintenance) plus sorafenib 400 mg twice daily; tigatuzumab (6 mg/kg loading, 6 mg/kg/week maintenance) plus sorafenib 400 mg twice daily; or sorafenib 400 mg twice daily. The primary end point was time to progression. Secondary end points included overall survival and safety. RESULTS: 163 subjects were randomized to treatment. Median time to progression was 3.0 months in the tigatuzumab 6/2 mg/kg combination group (p=0.988 vs. sorafenib), 3.9 months in the tigatuzumab 6/6 mg/kg combination group (p=0.586 vs. sorafenib), and 2.8 months in the sorafenib alone group. Median overall survival was 12.2 months in the tigatuzumab 6/6 mg/kg combination group (p=0.659 vs. sorafenib), vs. 8.2 months in both other treatment groups (p=0.303, tigatuzumab 6/2 mg/kg combination vs. sorafenib). The most common treatment-emergent adverse events were palmar-plantar erythrodysesthesia syndrome, diarrhea, and decreased appetite. CONCLUSIONS: Tigatuzumab combined with sorafenib vs. sorafenib alone in adults with advanced hepatocellular carcinoma did not meet its primary efficacy end point, although tigatuzumab plus sorafenib is well tolerated in hepatocellular carcinoma.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Estadificación de Neoplasias , Niacinamida/análogos & derivados , Compuestos de Fenilurea/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Carcinoma Hepatocelular/patología , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Niacinamida/administración & dosificación , Receptores de Factores de Crecimiento Endotelial Vascular , Estudios Retrospectivos , Sorafenib , Factores de Tiempo , Resultado del Tratamiento
9.
Adv Exp Med Biol ; 867: 81-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26530361

RESUMEN

Predictive biomarkers, defined as biomarkers that can be used to identify patient populations who will optimally benefit from therapy, are an important part of the future of oncology. They have the potential to reduce the size and cost of clinical development programs for oncology therapy, while increasing their probability of success and the ultimate value of cancer medicines. But predictive biomarkers do not always work, and under these circumstances they add cost, complexity, and time to drug development. This chapter describes Phase 2 and 3 development methods which efficiently and adaptively evaluate the ability of the biomarker to predict clinical outcomes. In the end, the biomarker is emphasized to the extent that it is actually predictive. This allows clinical cancer drug developers to manage uncertainty in the validity of biomarkers, leading to maximal value for predictive biomarkers and their associated oncology therapies.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias/terapia , Humanos , Neoplasias/diagnóstico , Medicina de Precisión
10.
Proc Natl Acad Sci U S A ; 109(36): 14586-91, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22891318

RESUMEN

Cancers are heterogeneous and genetically unstable. Current practice of personalized medicine tailors therapy to heterogeneity between cancers of the same organ type. However, it does not yet systematically address heterogeneity at the single-cell level within a single individual's cancer or the dynamic nature of cancer due to genetic and epigenetic change as well as transient functional changes. We have developed a mathematical model of personalized cancer therapy incorporating genetic evolutionary dynamics and single-cell heterogeneity, and have examined simulated clinical outcomes. Analyses of an illustrative case and a virtual clinical trial of over 3 million evaluable "patients" demonstrate that augmented (and sometimes counterintuitive) nonstandard personalized medicine strategies may lead to superior patient outcomes compared with the current personalized medicine approach. Current personalized medicine matches therapy to a tumor molecular profile at diagnosis and at tumor relapse or progression, generally focusing on the average, static, and current properties of the sample. Nonstandard strategies also consider minor subclones, dynamics, and predicted future tumor states. Our methods allow systematic study and evaluation of nonstandard personalized medicine strategies. These findings may, in turn, suggest global adjustments and enhancements to translational oncology research paradigms.


Asunto(s)
Epigénesis Genética , Evolución Molecular , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Medicina de Precisión/métodos , Biología de Sistemas/métodos , Simulación por Computador , Humanos , Medicina de Precisión/tendencias
11.
Clin Cancer Res ; 30(3): 480-488, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37792436

RESUMEN

Since the first approval of a tumor-agnostic indication in 2017, a total of seven tumor-agnostic indications involving six drugs have received approval from the FDA. In this paper, the master protocol subteam of the Statistical Methods in Oncology Scientific Working Group, Biopharmaceutical Session, American Statistical Association, provides a comprehensive summary of these seven tumor-agnostic approvals, describing their mechanisms of action; biomarker prevalence; study design; companion diagnostics; regulatory aspects, including comparisons of global regulatory requirements; and health technology assessment approval. Also discussed are practical considerations relating to the regulatory approval of tumor-agnostic indications, specifically (i) recommendations for the design stage to mitigate the risk that exceptions may occur if a treatment is initially hypothesized to be effective for all tumor types and (ii) because drug development continues after approval of a tumor-agnostic indication, recommendations for further development of tumor-specific indications in first-line patients in the setting of a randomized confirmatory basket trial, acknowledging the challenges in this area. These recommendations and practical considerations may provide insights for the future development of drugs for tumor-agnostic indications.


Asunto(s)
Aprobación de Drogas , Neoplasias , Humanos , Estados Unidos , United States Food and Drug Administration , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Desarrollo de Medicamentos , Biomarcadores
12.
Chin J Cancer ; 32(5): 233-41, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23489587

RESUMEN

Predictive biomarkers are important to the future of oncology; they can be used to identify patient populations who will benefit from therapy, increase the value of cancer medicines, and decrease the size and cost of clinical trials while increasing their chance of success. But predictive biomarkers do not always work. When unsuccessful, they add cost, complexity, and time to drug development. This perspective describes phases 2 and 3 development methods that efficiently and adaptively check the ability of a biomarker to predict clinical outcomes. In the end, the biomarker is emphasized to the extent that it can actually predict.


Asunto(s)
Biomarcadores de Tumor , Ensayos Clínicos como Asunto/métodos , Medicina Basada en la Evidencia , Neoplasias/diagnóstico , Desarrollo de Programa/métodos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Técnicas de Apoyo para la Decisión , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Valor Predictivo de las Pruebas
13.
Clin Pharmacol Ther ; 114(4): 802-809, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37489911

RESUMEN

The decentralized clinical trial (DCT) approach is increasingly recognized as a means to accelerate the development of potential therapeutic interventions. DCTs have a crucial advantage over traditional clinical trials: patients are monitored in their environment using technology (e.g., wearables), that capture data as they continue in daily life. This narrative review outlines a gap analysis focused on the frameworks and guidance from expert working groups and regulatory agencies for the design and execution of DCTs. Eight DCT elements guided the analysis and summarized the frameworks and guidance: (1) suitability, (2) protocol, (3) investigational medicinal product (IMP) supply, (4) investigators and health care providers, (5) safety, (6) regulatory and ethics, (7) data and technology, and (8) engagement, communication, and advocacy. Based on the gap analysis, two key takeaways were identified: (1) a need for a comprehensive sustainability assessment of each DCT element; and (2) current frameworks and guidance provide recommendations on social sustainability and some on economic sustainability. DCTs are an essential evolution in healthcare research; however, more guidance related to a comprehensive assessment of designing and executing sustainable DCTs is needed. This is especially the case for environmental sustainability, including, for example, carbon footprint and disposal of IMPs and sensors.


Asunto(s)
Ensayos Clínicos como Asunto , Humanos
14.
Ther Innov Regul Sci ; 57(6): 1136-1147, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37615880

RESUMEN

Master protocols (MPs) are an important addition to the clinical trial repertoire. As defined by the U.S. Food and Drug Administration (FDA), this term means "a protocol designed with multiple sub-studies, which may have different objectives (goals) and involve coordinated efforts to evaluate one or more investigational drugs in one or more disease subtypes within the overall trial structure." This means we now have a unique, scientifically based MP that describes how a clinical trial will be conducted using one or more potential candidate therapies to treat patients in one or more diseases. Patient engagement (PE) is also a critical factor that has been recognized by FDA through its Patient-Focused Drug Development (PFDD) initiative, and by the European Medicines Agency (EMA), which states on its website that it has been actively interacting with patients since the creation of the Agency in 1995. We propose that utilizing these PE principles in MPs can make them more successful for sponsors, providers, and patients. Potential benefits of MPs for patients awaiting treatment can include treatments that better fit a patient's needs; availability of more treatments; and faster access to treatments. These make it possible to develop innovative therapies (especially for rare diseases and/or unique subpopulations, e.g., pediatrics), to minimize untoward side effects through careful dose escalation practices and, by sharing a control arm, to lower the probability of being assigned to a placebo arm for clinical trial participants. This paper is authored by select members of the American Statistical Association (ASA)/DahShu Master Protocol Working Group (MPWG) People and Patient Engagement (PE) Subteam. DahShu is a 501(c)(3) non-profit organization, founded to promote research and education in data science. This manuscript does not include direct feedback from US or non-US regulators, though multiple regulatory-related references are cited to confirm our observation that improving patient engagement is supported by regulators. This manuscript represents the authors' independent perspective on the Master Protocol; it does not represent the official policy or viewpoint of FDA or any other regulatory organization or the views of the authors' employers. The objective of this manuscript is to provide drug developers, contract research organizations (CROs), third party capital investors, patient advocacy groups (PAGs), and biopharmaceutical executives with a better understanding of how including the patient voice throughout MP development and conduct creates more efficient clinical trials. The PE Subteam also plans to publish a Plain Language Summary (PLS) of this publication for clinical trial participants, patients, caregivers, and the public as they seek to understand the risks and benefits of MP clinical trial participation.

15.
Endocrinology ; 164(12)2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37897495

RESUMEN

Breast tumors overexpressing human epidermal growth factor receptor (HER2) confer intrinsic resistance to endocrine therapy (ET), and patients with HER2/estrogen receptor-positive (HER2+/ER+) breast cancer (BCa) are less responsive to ET than HER2-/ER+. However, real-world evidence reveals that a large subset of patients with HER2+/ER+ receive ET as monotherapy, positioning this treatment pattern as a clinical challenge. In the present study, we developed and characterized 2 in vitro models of ET-resistant (ETR) HER2+/ER+ BCa to identify possible therapeutic vulnerabilities. To mimic ETR to aromatase inhibitors (AIs), we developed 2 long-term estrogen deprivation (LTED) cell lines from BT-474 (BT474) and MDA-MB-361 (MM361). Growth assays, PAM50 subtyping, and genomic and transcriptomic analyses, followed by validation and functional studies, were used to identify targetable differences between ET-responsive parental and ETR-LTED HER2+/ER+ cells. Compared to their parental cells, MM361 LTEDs grew faster, lost ER, and increased HER2 expression, whereas BT474 LTEDs grew slower and maintained ER and HER2 expression. Both LTED variants had reduced responsiveness to fulvestrant. Whole-genome sequencing of aggressive MM361 LTEDs identified mutations in genes encoding transcription factors and chromatin modifiers. Single-cell RNA sequencing demonstrated a shift towards non-luminal phenotypes, and revealed metabolic remodeling of MM361 LTEDs, with upregulated lipid metabolism and ferroptosis-associated antioxidant genes, including GPX4. Combining a GPX4 inhibitor with anti-HER2 agents induced significant cell death in both MM361 and BT474 LTEDs. The BT474 and MM361 AI-resistant models capture distinct phenotypes of HER2+/ER+ BCa and identify altered lipid metabolism and ferroptosis remodeling as vulnerabilities of this type of ETR BCa.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/genética , Fulvestrant/farmacología , Fulvestrant/uso terapéutico , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/uso terapéutico , Estrógenos/metabolismo , Línea Celular Tumoral , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
16.
Orphanet J Rare Dis ; 18(1): 79, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37041605

RESUMEN

BACKGROUND: Traditional clinical trials require tests and procedures that are administered in centralized clinical research sites, which are beyond the standard of care that patients receive for their rare and chronic diseases. The limited number of rare disease patients scattered around the world makes it particularly challenging to recruit participants and conduct these traditional clinical trials. MAIN BODY: Participating in clinical research can be burdensome, especially for children, the elderly, physically and cognitively impaired individuals who require transportation and caregiver assistance, or patients who live in remote locations or cannot afford transportation. In recent years, there is an increasing need to consider Decentralized Clinical Trials (DCT) as a participant-centric approach that uses new technologies and innovative procedures for interaction with participants in the comfort of their home. CONCLUSION: This paper discusses the planning and conduct of DCTs, which can increase the quality of trials with a specific focus on rare diseases.


Asunto(s)
Cuidadores , Enfermedades Raras , Anciano , Niño , Humanos , Ensayos Clínicos como Asunto
17.
bioRxiv ; 2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37662291

RESUMEN

Background: Breast tumors overexpressing human epidermal growth factor receptor (HER2) confer intrinsic resistance to endocrine therapy (ET), and patients with HER2/ estrogen receptor-positive (HER2+/HR+) breast cancer (BCa) are less responsive to ET than HER2-/ER+. However, real-world evidence reveals that a large subset of HER2+/ER+ patients receive ET as monotherapy, positioning this treatment pattern as a clinical challenge. In the present study, we developed and characterized two distinct in vitro models of ET-resistant (ETR) HER2+/ER+ BCa to identify possible therapeutic vulnerabilities. Methods: To mimic ETR to aromatase inhibitors (AI), we developed two long-term estrogen-deprived (LTED) cell lines from BT-474 (BT474) and MDA-MB-361 (MM361). Growth assays, PAM50 molecular subtyping, genomic and transcriptomic analyses, followed by validation and functional studies, were used to identify targetable differences between ET-responsive parental and ETR-LTED HER2+/ER+ cells. Results: Compared to their parental cells, MM361 LTEDs grew faster, lost ER, and increased HER2 expression, whereas BT474 LTEDs grew slower and maintained ER and HER2 expression. Both LTED variants had reduced responsiveness to fulvestrant. Whole-genome sequencing of the more aggressive MM361 LTED model system identified exonic mutations in genes encoding transcription factors and chromatin modifiers. Single-cell RNA sequencing demonstrated a shift towards non-luminal phenotypes, and revealed metabolic remodeling of MM361 LTEDs, with upregulated lipid metabolism and antioxidant genes associated with ferroptosis, including GPX4. Combining the GPX4 inhibitor RSL3 with anti-HER2 agents induced significant cell death in both the MM361 and BT474 LTEDs. Conclusions: The BT474 and MM361 AI-resistant models capture distinct phenotypes of HER2+/ER+ BCa and identify altered lipid metabolism and ferroptosis remodeling as vulnerabilities of this type of ETR BCa.

18.
Cancer ; 118(21): 5403-13, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22570147

RESUMEN

BACKGROUND: Efatutazone (CS-7017), a novel peroxisome proliferator-activated receptor gamma (PPARγ) agonist, exerts anticancer activity in preclinical models. The authors conducted a phase 1 study to determine the recommended phase 2 dose, safety, tolerability, and pharmacokinetics of efatutazone. METHODS: Patients with advanced solid malignancies and no curative therapeutic options were enrolled to receive a given dose of efatutazone, administered orally (PO) twice daily for 6 weeks, in a 3 + 3 intercohort dose-escalation trial. After the third patient, patients with diabetes mellitus were excluded. Efatutazone dosing continued until disease progression or unacceptable toxicity, with measurement of efatutazone pharmacokinetics and plasma adiponectin levels. RESULTS: Thirty-one patients received efatutazone at doses ranging from 0.10 to 1.15 mg PO twice daily. Dose escalation stopped when maximal impact on PPARγ-related biomarkers had been reached before any protocol-defined maximum-tolerated dose level. On the basis of a population pharmacokinetic/pharmacodynamic analysis, the recommended phase 2 dose was 0.5 mg PO twice daily. A majority of patients experienced peripheral edema (53.3%), often requiring diuretics. Three episodes of dose-limiting toxicities, related to fluid retention, were noted in the 0.10-, 0.25-, and 1.15-mg cohorts. Of 31 treated patients, 27 were evaluable for response. A sustained partial response (PR; 690 days on therapy) was observed in a patient with myxoid liposarcoma. Ten additional patients had stable disease (SD) for ≥60 days. Exposures were approximately dose proportional, and adiponectin levels increased after 4 weeks of treatment at all dose levels. Immunohistochemistry of archived specimens demonstrated that PPARγ and retinoid X receptor expression levels were significantly greater in patients with SD for ≥60 days or PR (P = .0079), suggesting a predictive biomarker. CONCLUSIONS: Efatutazone demonstrates acceptable tolerability with evidence of disease control in patients with advanced malignancies.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias/tratamiento farmacológico , PPAR gamma/agonistas , Tiazolidinedionas/administración & dosificación , Administración Oral , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Tiazolidinedionas/efectos adversos , Tiazolidinedionas/farmacocinética
19.
Semin Cancer Biol ; 20(5): 340-52, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20934514

RESUMEN

Cancer development requires multiple oncogenic mutations. Pathogenic mechanisms which accelerate this process may be favored carcinogenic pathways. Mutator mutations are mutations in genetic stability genes, and increase the mutation rate, speeding up the accumulation of oncogenic mutations. The mutator hypothesis states that mutator mutations play a critical role in carcinogenesis. Alternatively, tumors might arise by mutations occurring at the normal rate followed by selection and expansion of various premalignant lineages on the path to cancer. This alternative pathway is a significant argument against the mutator hypothesis. Mutator mutations may also lead to accumulation of deleterious mutations, which could lead to extinction of premalignant lineages before they become cancerous, another argument against the mutator hypothesis. Finally, the need for acquisition of a mutator mutation imposes an additional step on the carcinogenic process. Accordingly, the mutator hypothesis has been a seminal but controversial idea for several decades despite considerable experimental and theoretical work. To resolve this debate, the concept of efficiency has been introduced as a metric for comparing carcinogenic mechanisms, and a new theoretical approach of focused quantitative modeling has been applied. The results demonstrate that, given what is already known, the predominance of mutator mechanisms is likely inevitable, as they overwhelm less efficient non-mutator pathways to cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes Relacionados con las Neoplasias , Mutación , Neoplasias/genética , Inestabilidad Genómica , Humanos , Modelos Biológicos , Fenotipo
20.
Paediatr Drugs ; 24(6): 657-669, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36241954

RESUMEN

Many of the afflictions of children are rare diseases. This creates numerous drug development challenges related to small populations, including limited information about the disease state, enrollment challenges, and diminished incentives for pediatric development of novel therapies by pharmaceutical and biotechnology sponsors. We review selected innovations in clinical development that may partially mitigate some of these difficulties, starting with the concept of development efficiency for individual clinical trials, clinical programs (involving multiple trials for a single drug), and clinical portfolios of multiple drugs, and decision analysis as a tool to optimize efficiency. Development efficiency is defined as the ability to reach equally rigorous or more rigorous conclusions in less time, with fewer trial participants, or with fewer resources. We go on to discuss efficient methods for matching targeted therapies to biomarker-defined subgroups, methods for eliminating or reducing the need for natural history data to guide rare disease development, the use of basket trials to enhance efficiency by grouping multiple similar disease applications in a single clinical trial, and the use of alternative data sources including historical controls to augment or replace concurrent controls in clinical studies. Greater understanding and broader application of these methods could lead to improved therapies and/or more widespread and rapid access to novel therapies for rare diseases in both children and adults.


Asunto(s)
Desarrollo de Medicamentos , Enfermedades Raras , Adulto , Humanos , Niño , Enfermedades Raras/tratamiento farmacológico , Biomarcadores , Preparaciones Farmacéuticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA