Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
JCO Oncol Pract ; : OP2400217, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38986031

RESUMEN

In a randomized clinical trial, instead of allocating patients equally between the treatment arms, some trials in oncology assign a higher proportion of patients to receive the experimental treatment arm (eg, a two-to-one randomization). In this commentary, we first briefly review the common reasons given for the use of a two-to-one randomization and provide some examples of trials using these designs. We then explain why the risk-benefit ratio of this approach may not be favorable as is commonly assumed.

2.
Stat Med ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38780538

RESUMEN

When designing a randomized clinical trial to compare two treatments, the sample size required to have desired power with a specified type 1 error depends on the hypothesis testing procedure. With a binary endpoint (e.g., response), the trial results can be displayed in a 2 × 2 table. If one does the analysis conditional on the number of positive responses, then using Fisher's exact test has an actual type 1 error less than or equal to the specified nominal type 1 error. Alternatively, one can use one of many unconditional "exact" tests that also preserve the type 1 error and are less conservative than Fisher's exact test. In particular, the unconditional test of Boschloo is always at least as powerful as Fisher's exact test, leading to smaller required sample sizes for clinical trials. However, many statisticians have argued over the years that the conditional analysis with Fisher's exact test is the only appropriate procedure. Since having smaller clinical trials is an extremely important consideration, we review the general arguments given for the conditional analysis of a 2 × 2 table in the context of a randomized clinical trial. We find the arguments not relevant in this context, or, if relevant, not completely convincing, suggesting the sample-size advantage of the unconditional tests should lead to their recommended use. We also briefly suggest that since designers of clinical trials practically always have target null and alternative response rates, there is the possibility of using this information to improve the power of the unconditional tests.

3.
J Clin Oncol ; : JCO2400025, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38759123

RESUMEN

New oncology therapies that extend patients' lives beyond initial expectations and improving later-line treatments can lead to complications in clinical trial design and conduct. In particular, for trials with event-based analyses, the time to observe all the protocol-specified events can exceed the finite follow-up of a clinical trial or can lead to much delayed release of outcome data. With the advent of multiple classes of oncology therapies leading to much longer survival than in the past, this issue in clinical trial design and conduct has become increasingly important in recent years. We propose a straightforward prespecified backstop rule for trials with a time-to-event analysis and evaluate the impact of the rule with both simulated and real-world trial data. We then provide recommendations for implementing the rule across a range of oncology clinical trial settings.

5.
Clin Cancer Res ; 30(4): 673-679, 2024 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-38048044

RESUMEN

In recent years, there has been increased interest in incorporation of backfilling into dose-escalation clinical trials, which involves concurrently assigning patients to doses that have been previously cleared for safety by the dose-escalation design. Backfilling generates additional information on safety, tolerability, and preliminary activity on a range of doses below the maximum tolerated dose (MTD), which is relevant for selection of the recommended phase II dose and dose optimization. However, in practice, backfilling may not be rigorously defined in trial protocols and implemented consistently. Furthermore, backfilling designs require careful planning to minimize the probability of treating additional patients with potentially inactive agents (and/or subtherapeutic doses). In this paper, we propose a simple and principled approach to incorporate backfilling into the Bayesian optimal interval design (BOIN). The design integrates data from the dose-escalation and backfilling components of the design and ensures that the additional patients are treated at doses where some activity has been seen. Simulation studies demonstrated that the proposed backfilling BOIN design (BF-BOIN) generates additional data for future dose optimization, maintains the accuracy of the MTD identification, and improves patient safety without prolonging the trial duration.


Asunto(s)
Neoplasias , Proyectos de Investigación , Humanos , Teorema de Bayes , Simulación por Computador , Dosis Máxima Tolerada , Relación Dosis-Respuesta a Droga , Neoplasias/tratamiento farmacológico
7.
J Clin Oncol ; 41(29): 4616-4620, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37471685

RESUMEN

Recent therapeutic advances have led to improved patient survival in many cancer settings. Although prolongation of survival remains the ultimate goal of cancer treatment, the availability of effective salvage therapies could make definitive phase III trials with primary overall survival (OS) end points difficult to complete in a timely manner. Therefore, to accelerate development of new therapies, many phase III trials of new cancer therapies are now designed with intermediate primary end points (eg, progression-free survival in the metastatic setting) with OS designated as a secondary end point. We review recently published phase III trials and assess contemporary practices for designing and reporting OS as a secondary end point. We then provide design and reporting recommendations for trials with OS as a secondary end point to safeguard OS data integrity and optimize access to the OS data for patient, clinician, and public-health stakeholders.

8.
Clin Cancer Res ; 29(8): 1412-1422, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-36662819

RESUMEN

Over the past decade, multiple trials, including the precision medicine trial National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH, EAY131, NCT02465060) have sought to determine if treating cancer based on specific genomic alterations is effective, irrespective of the cancer histology. Although many therapies are now approved for the treatment of cancers harboring specific genomic alterations, most patients do not respond to therapies targeting a single alteration. Further, when antitumor responses do occur, they are often not durable due to the development of drug resistance. Therefore, there is a great need to identify rational combination therapies that may be more effective. To address this need, the NCI and National Clinical Trials Network have developed NCI-ComboMATCH, the successor to NCI-MATCH. Like the original trial, NCI-ComboMATCH is a signal-seeking study. The goal of ComboMATCH is to overcome drug resistance to single-agent therapy and/or utilize novel synergies to increase efficacy by developing genomically-directed combination therapies, supported by strong preclinical in vivo evidence. Although NCI-MATCH was mainly comprised of multiple single-arm studies, NCI-ComboMATCH tests combination therapy, evaluating both combination of targeted agents as well as combinations of targeted therapy with chemotherapy. Although NCI-MATCH was histology agnostic with selected tumor exclusions, ComboMATCH has histology-specific and histology-agnostic arms. Although NCI-MATCH consisted of single-arm studies, ComboMATCH utilizes single-arm as well as randomized designs. NCI-MATCH had a separate, parallel Pediatric MATCH trial, whereas ComboMATCH will include children within the same trial. We present rationale, scientific principles, study design, and logistics supporting the ComboMATCH study.


Asunto(s)
Antineoplásicos , Neoplasias , Niño , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Terapia Combinada , National Cancer Institute (U.S.) , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Medicina de Precisión , Estados Unidos
9.
J Natl Cancer Inst ; 115(1): 14-20, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36161487

RESUMEN

As precision medicine becomes more precise, the sizes of the molecularly targeted subpopulations become increasingly smaller. This can make it challenging to conduct randomized clinical trials of the targeted therapies in a timely manner. To help with this problem of a small patient subpopulation, a study design that is frequently proposed is to conduct a small randomized clinical trial (RCT) with the intent of augmenting the RCT control arm data with historical data from a set of patients who have received the control treatment outside the RCT (historical control data). In particular, strategies have been developed that compare the treatment outcomes across the cohorts of patients treated with the standard (control) treatment to guide the use of the historical data in the analysis; this can lessen the potential well-known biases of using historical controls without any randomization. Using some simple examples and completed studies, we demonstrate in this commentary that these strategies are unlikely to be useful in precision medicine applications.


Asunto(s)
Medicina de Precisión , Proyectos de Investigación , Humanos , Resultado del Tratamiento
11.
J Natl Cancer Inst ; 115(5): 492-497, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36534891

RESUMEN

The goal of dose optimization during drug development is to identify a dose that preserves clinical benefit with optimal tolerability. Traditionally, the maximum tolerated dose in a small phase I dose escalation study is used in the phase II trial assessing clinical activity of the agent. Although it is possible that this dose level could be altered in the phase II trial if an unexpected level of toxicity is seen, no formal dose optimization has routinely been incorporated into later stages of drug development. Recently it has been suggested that formal dose optimization (involving randomly assigning patients between 2 or more dose levels) be routinely performed early in drug development, even before it is known that the experimental therapy has any clinical activity at any dose level. We consider the relative merits of performing dose optimization earlier vs later in the drug development process and demonstrate that a considerable number of patients may be exposed to ineffective therapies unless dose optimization is delayed until after clinical activity or benefit of the new agent has been established. We conclude that patient and public health interests may be better served by conducting dose optimization after (or during) phase III evaluation, with some exceptions when dose optimization should be performed after activity shown in phase II evaluation.


Asunto(s)
Desarrollo de Medicamentos , Proyectos de Investigación , Humanos , Dosis Máxima Tolerada , Relación Dosis-Respuesta a Droga
12.
Blood Cancer J ; 12(6): 98, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768410

RESUMEN

A wide variety of new therapeutic options for Multiple Myeloma (MM) have recently become available, extending progression-free and overall survival for patients in meaningful ways. However, these treatments are not curative, and patients eventually relapse, necessitating decisions on the appropriate choice of treatment(s) for the next phase of the disease. Additionally, an important subset of MM patients will prove to be refractory to the majority of the available treatments, requiring selection of effective therapies from the remaining options. Immunomodulatory agents (IMiDs), proteasome inhibitors, monoclonal antibodies, and alkylating agents are the major classes of MM therapies, with several options in each class. Patients who are refractory to one agent in a class may be responsive to a related compound or to a drug from a different class. However, rules for selection of alternative treatments in these situations are somewhat empirical and later phase clinical trials to inform those choices are ongoing. To address these issues the NCI Multiple Myeloma Steering Committee formed a relapsed/refractory working group to review optimal treatment choices, timing, and sequencing and provide recommendations. Additional issues considered include the role of salvage autologous stem cell transplantation, risk stratification, targeted approaches for genetic subsets of MM, appropriate clinical trial endpoints, and promising investigational agents. This report summarizes the deliberations of the working group and suggests potential avenues of research to improve the precision, timing, and durability of treatments for Myeloma.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple , Consenso , Humanos , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia , Trasplante Autólogo
13.
J Natl Cancer Inst ; 114(9): 1222-1227, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-35583264

RESUMEN

Recently developed clinical-benefit outcome scales by the European Society for Medical Oncology and the American Society of Clinical Oncology allow standardized objective evaluation of outcomes of randomized clinical trials. However, incorporation of clinical-benefit outcome scales into trial designs highlights a number of statistical issues: the relationship between minimal clinical benefit and the target treatment-effect alternative used in the trial design, designing trials to assess long-term benefit, potential problems with using a trial endpoint that is not overall survival, and how to incorporate subgroup analyses into the trial design. Using the European Society for Medical Oncology Magnitude of Clinical Benefit Scale as a basis for discussion, we review what these issues are and how they can guide the choice of trial-design target effects, appropriate endpoints, and prespecified subgroup analyses to increase the chances that the resulting trial outcomes can be appropriately evaluated for clinical benefit.


Asunto(s)
Neoplasias , Humanos , Oncología Médica/métodos , Neoplasias/tratamiento farmacológico
14.
Clin Trials ; 19(2): 158-161, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34991348

RESUMEN

Response-adaptive randomization, which changes the randomization ratio as a randomized clinical trial progresses, is inefficient as compared to a fixed 1:1 randomization ratio in terms of increased required sample size. It is also known that response-adaptive randomization leads to biased treatment effects if there are time trends in the accruing outcome data, for example, due to changes in the patient population being accrued, evaluation methods, or concomitant treatments. Response-adaptive-randomization analysis methods that account for potential time trends, such as time-block stratification or re-randomization, can eliminate this bias. However, as shown in this Commentary, these analysis methods cause a large additional inefficiency of response-adaptive randomization, regardless of whether a time trend actually exists.


Asunto(s)
Proyectos de Investigación , Sesgo , Humanos , Distribución Aleatoria , Tamaño de la Muestra
15.
J Natl Cancer Inst ; 114(2): 187-190, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-34289052

RESUMEN

Efficient biomarker-driven randomized clinical trials are a key tool for implementing precision oncology. A commonly used biomarker phase III design is focused on testing the treatment effect in biomarker-positive and overall study populations. This approach may result in recommending new treatments to biomarker-negative patients when these treatments have no benefit for these patients.


Asunto(s)
Neoplasias , Proyectos de Investigación , Biomarcadores , Humanos , Oncología Médica , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Medicina de Precisión , Ensayos Clínicos Controlados Aleatorios como Asunto
16.
Clin Trials ; 18(6): 746, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34524050
18.
Clin Trials ; 18(2): 188-196, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33626896

RESUMEN

BACKGROUND: Restricted mean survival time methods compare the areas under the Kaplan-Meier curves up to a time τ for the control and experimental treatments. Extraordinary claims have been made about the benefits (in terms of dramatically smaller required sample sizes) when using restricted mean survival time methods as compared to proportional hazards methods for analyzing noninferiority trials, even when the true survival distributions satisfy proportional hazardss. METHODS: Through some limited simulations and asymptotic power calculations, the authors compare the operating characteristics of restricted mean survival time and proportional hazards methods for analyzing both noninferiority and superiority trials under proportional hazardss to understand what relative power benefits there are when using restricted mean survival time methods for noninferiority testing. RESULTS: In the setting of low-event rates, very large targeted noninferiority margins, and limited follow-up past τ, restricted mean survival time methods have more power than proportional hazards methods. For superiority testing, proportional hazards methods have more power. This is not a small-sample phenomenon but requires a low-event rate and a large noninferiority margin. CONCLUSION: Although there are special settings where restricted mean survival time methods have a power advantage over proportional hazards methods for testing noninferiority, the larger issue in these settings is defining appropriate noninferiority margins. We find the restricted mean survival time methods lacking in these regards.


Asunto(s)
Estudios de Equivalencia como Asunto , Proyectos de Investigación , Tasa de Supervivencia , Humanos , Modelos de Riesgos Proporcionales , Tamaño de la Muestra , Análisis de Supervivencia
19.
ESMO Open ; 5(5): e000871, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33122353

RESUMEN

BACKGROUND: With the development of targeted agents, the approach to combination cancer therapy has evolved to focus on identifying ways in which pathway inhibition by one agent may enhance the activity of other agents. In theory, this implies that under this new paradigm, agents are no longer required to show single-agent activity, as the pathway inhibited by the targeted agent may only have a therapeutic effect when given with other agents. This raises the question of the extent to which anticancer agents without single-agent activity can contribute to effective combination regimens. PATIENTS AND METHODS: We reviewed outcomes of randomised phase 2 combination trials sponsored by the National Cancer Institute Cancer Therapy Evaluation Program that were activated in 2008 to 2017 and noted the single-agent activity of the experimental agents. RESULTS: Fifty-three trials were identified, and 50 had available results: 7 (14%), 15 (30%) and 28 (56%) had experimental agents with single-agent activity classified as active, inactive and indeterminate, respectively. Thirteen per cent (95% CI=1.7% to 40.5%) of trials evaluating inactive agents and 11.6% (95% CI=3.9% to 25.1%) of trials evaluating agents without known single-agent activity (pooled inactive and indeterminate) were positive, compared with 42.9% (95% CI=9.9% to 81.6%) for agents with single-agent activity. CONCLUSIONS: Incorporating agents without documented single-agent activity into treatment regimens is unlikely to produce meaningful improvements in activity unless there is compelling biological rationale. This finding has important implications for the prioritisation of anticancer agents for combination testing, and for the allocation of clinical trial resources.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico
20.
J Clin Oncol ; 38(17): 2003-2004, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32315276
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...