Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Prostate ; 84(6): 584-598, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38311868

RESUMO

BACKGROUND: Teverelix drug product (DP) is a novel injectable gonadotropin-releasing hormone antagonist. METHODS: An adaptive phase 2, open-label, multicenter trial was conducted in patients with advanced prostate cancer to evaluate the efficacy and safety of a combined subcutaneous (SC) and intramuscular (IM) loading dose regimen of teverelix DP of 120 mg SC + 120 mg IM (Group 1; N = 9) or 180 mg SC + 180 mg IM (Group 2; N = 41) administered at a single visit, followed by 6-weekly SC maintenance doses of 120 mg (Group 1) or 180 mg (Group 2), up to Day 168. The primary endpoint was the proportion of patients achieving castration levels with serum testosterone <0.5 ng/mL at Day 28 with a target castration rate of 90%. Injection sites were inspected by the investigator at every visit and reactions (ISRs) were proactively recorded. RESULTS: The target castration rate was reached in Group 2 (97.5%) but not in Group 1 (62.5%). The castration rates were not maintained to Day 42 (Group 2: 82.5%; Group 1: 50.0%). Suppression of testosterone to castrate levels occurred rapidly (median time: 2 days for both groups). Suppression of testosterone, prostate-specific antigen, follicle-stimulating hormone, and luteinizing hormone was sustained throughout the treatment period, being more prominent with the higher dose. The adverse event (AE) profile was similar between groups. The most common AEs were injection-site induration (n = 40: 80.0%), injection-site erythema (n = 35: 70.0%), and hot flush (n = 21: 42.0%). Most ISRs were Grade 1. CONCLUSION: Overall, the teverelix DP doses were generally well-tolerated but did not adequately maintain castration levels.


Assuntos
Neoplasias da Próstata , Humanos , Masculino , Hormônio Liberador de Gonadotropina , Oligopeptídeos , Antígeno Prostático Específico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/cirurgia , Testosterona/sangue
2.
Biometrics ; 80(1)2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38364811

RESUMO

A generalized phase 1-2-3 design, Gen 1-2-3, that includes all phases of clinical treatment evaluation is proposed. The design extends and modifies the design of Chapple and Thall (2019), denoted by CT. Both designs begin with a phase 1-2 trial including dose acceptability and optimality criteria, and both select an optimal dose for phase 3. The Gen 1-2-3 design has the following key differences. In stage 1, it uses phase 1-2 criteria to identify a set of candidate doses rather than 1 dose. In stage 2, which is intermediate between phase 1-2 and phase 3, it randomizes additional patients fairly among the candidate doses and an active control treatment arm and uses survival time data from both stage 1 and stage 2 patients to select an optimal dose. It then makes a Go/No Go decision of whether or not to conduct phase 3 based on the predictive probability that the selected optimal dose will provide a specified substantive improvement in survival time over the control. A simulation study shows that the Gen 1-2-3 design has desirable operating characteristics compared to the CT design and 2 conventional designs.


Assuntos
Projetos de Pesquisa , Humanos , Protocolos Clínicos , Simulação por Computador , Relação Dose-Resposta a Droga , Probabilidade , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto
3.
Stat Med ; 43(12): 2472-2485, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38605556

RESUMO

The statistical methodology for model-based dose finding under model uncertainty has attracted increasing attention in recent years. While the underlying principles are simple and easy to understand, developing and implementing an efficient approach for binary responses can be a formidable task in practice. Motivated by the statistical challenges encountered in a phase II dose finding study, we explore several key design and analysis issues related to the hybrid testing-modeling approaches for binary responses. The issues include candidate model selection and specifications, optimal design and efficient sample size allocations, and, notably, the methods for dose-response testing and estimation. Specifically, we consider a class of generalized linear models suited for the candidate set and establish D-optimal designs for these models. Additionally, we propose using permutation-based tests for dose-response testing to avoid asymptotic normality assumptions typically required for contrast-based tests. We perform trial simulations to enhance our understanding of these issues.


Assuntos
Simulação por Computador , Relação Dose-Resposta a Droga , Modelos Estatísticos , Humanos , Incerteza , Modelos Lineares , Ensaios Clínicos Fase II como Assunto/métodos , Ensaios Clínicos Fase II como Assunto/estatística & dados numéricos , Tamanho da Amostra , Projetos de Pesquisa , Interpretação Estatística de Dados
4.
Stat Med ; 43(15): 2972-2986, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38747472

RESUMO

The U.S. Food and Drug Administration (FDA) has launched Project Optimus to shift dose selection from the maximum tolerated dose (MTD) to the dose that produces the optimal risk-benefit tradeoff. One approach highlighted in the FDA's guidance involves conducting a randomized phase II trial following the completion of a phase I trial, where multiple doses (typically including the MTD and one or two doses lower than the MTD) are compared to identify the optimal dose that maximizes the benefit-risk tradeoff. This article focuses on the design of such a multiple-dose randomized trial, specifically the determination of the sample size. We generalized the standard definitions of type I error and power to accommodate the unique characteristics of dose optimization and derived a decision rule along with an algorithm to determine the optimal sample size. The resulting design is referred to as MERIT (Multiple-dosE RandomIzed Trial design for dose optimization based on toxicity and efficacy). Simulation studies demonstrate that MERIT has desirable operating characteristics, and a sample size between 20 and 40 per dosage arm often offers reasonable power and type I errors to ensure patient safety and benefit. To facilitate the implementation of the MERIT design, we provide software, available at https://www.trialdesign.org.


Assuntos
Algoritmos , Ensaios Clínicos Fase II como Assunto , Simulação por Computador , Dose Máxima Tolerável , Ensaios Clínicos Controlados Aleatórios como Assunto , Projetos de Pesquisa , Tamanho da Amostra , Humanos , Ensaios Clínicos Fase II como Assunto/métodos , Relação Dose-Resposta a Droga , Estados Unidos , United States Food and Drug Administration
5.
Stat Med ; 43(4): 689-705, 2024 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-38110304

RESUMO

We propose a model-based, semi-mechanistic dose-finding (SDF) design for phase I oncology trials that incorporates pharmacokinetic/pharmacodynamic (PK/PD) information when modeling the dose-toxicity relationship. This design is motivated by a phase Ib/II clinical trial of anti-CD20/CD3 T cell therapy in non-Hodgkin lymphoma patients; it extends a recently proposed SDF model framework by incorporating measurements of a PD biomarker relevant to the primary dose-limiting toxicity (DLT). We propose joint Bayesian modeling of the PK, PD, and DLT outcomes. Our extensive simulation studies show that on average the proposed design outperforms some common phase I trial designs, including modified toxicity probability interval (mTPI) and Bayesian optimal interval (BOIN) designs, the continual reassessment method (CRM), as well as an SDF design assuming a latent PD biomarker (SDF-woPD), in terms of the percentage of correct selection of maximum tolerated dose (MTD) and average number of patients allocated to MTD, under a variety of dose-toxicity scenarios. When the working PK model and the class of link function between the cumulative PD effect and DLT probability is correctly specified, the proposed design also yields better estimated dose-toxicity curves than CRM and SDF-woPD. Our sensitivity analyses suggest that the design's performance is reasonably robust to prior specification for the parameter in the link function, as well as misspecification of the PK model and class of the link function.


Assuntos
Neoplasias , Humanos , Teorema de Bayes , Neoplasias/tratamento farmacológico , Simulação por Computador , Biomarcadores , Dose Máxima Tolerável , Relação Dose-Resposta a Droga , Projetos de Pesquisa
6.
Stat Med ; 43(17): 3210-3226, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816959

RESUMO

The calibration-free odds (CFO) design has been demonstrated to be robust, model-free, and practically useful but faces challenges when dealing with late-onset toxicity. The emergence of the time-to-event (TITE) method and fractional method leads to the development of TITE-CFO and fractional CFO (fCFO) designs to accumulate delayed toxicity. Nevertheless, existing CFO-type designs have untapped potential because they primarily consider dose information from the current position and its two neighboring positions. To incorporate information from all doses, we propose the accumulative CFO (aCFO) design by utilizing data at all dose levels similar to a tug-of-war game where players distant from the center also contribute their strength. This approach enhances full information utilization while still preserving the model-free and calibration-free characteristics. Extensive simulation studies demonstrate performance improvement over the original CFO design, emphasizing the advantages of incorporating information from a broader range of dose levels. Furthermore, we propose to incorporate late-onset outcomes into the TITE-aCFO and f-aCFO designs, with f-aCFO displaying superior performance over existing methods in both fixed and random simulation scenarios. In conclusion, the aCFO and f-aCFO designs can be considered robust, efficient, and user-friendly approaches for conducting phase I trials without or with late-onsite toxicity.


Assuntos
Ensaios Clínicos Fase I como Assunto , Simulação por Computador , Humanos , Ensaios Clínicos Fase I como Assunto/métodos , Projetos de Pesquisa , Relação Dose-Resposta a Droga , Calibragem , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Modelos Estatísticos , Fatores de Tempo
7.
Stat Med ; 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39075332

RESUMO

Dual-agent treatment has become more and more popular in clinical trials. We have developed an approach called rapid enrollment dual-agent design (REDD) for dose-finding in Phase I clinical trials. This approach aims to administer treatment to patients using a dose combination that is highly probable to be the target dose combination. Unlike other non-model-based designs, rapid enrollment designs (RED and REDD) do not require waiting for all patients to complete an assessment before the assignment of the next participant. Simulations showed that across several scenarios, the average performance of REDD is comparable to that of the Bayesian optimal interval (BOIN) design and the partial order continual reassessment method (POCRM). The simulation results of REDD for late-onset toxicity assessments demonstrated that assigning patients to a dose combination as they are being enrolled, without waiting for the most recent cohort of patients to complete their follow-up, does not significantly compromise the quality of the maximum tolerated dose (MTD) estimation. Instead, it saves a considerable amount of time in clinical trial enrollment. User-friendly online applications have also been created to further facilitate the adoption of rapid enrollment designs in Phase I trials. In summary, being similar to BOIN and POCRM in performance, REDD is an approach that is easily comprehensible, straightforward to implement and offers an advantage of enrolling patients without having to wait for all current patients to complete their follow-ups for toxicity.

8.
BMC Med Res Methodol ; 24(1): 11, 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-38218799

RESUMO

BACKGROUND: In this article we describe the methodology of the time-to-event continual reassessment method in the presence of partial orders (PO-TITE-CRM) and the process of implementing this trial design into a phase I trial in head and neck cancer called ADePT-DDR. The ADePT-DDR trial aims to find the maximum tolerated dose of an ATR inhibitor given in conjunction with radiotherapy in patients with head and neck squamous cell carcinoma. METHODS: The PO-TITE-CRM is a phase I trial design that builds upon the time-to-event continual reassessment method (TITE-CRM) to allow for the presence of partial ordering of doses. Partial orders occur in the case where the monotonicity assumption does not hold and the ordering of doses in terms of toxicity is not fully known. RESULTS: We arrived at a parameterisation of the design which performed well over a range of scenarios. Results from simulations were used iteratively to determine the best parameterisation of the design and we present the final set of simulations. We provide details on the methodology as well as insight into how it is applied to the trial. CONCLUSIONS: Whilst being a very efficient design we highlight some of the difficulties and challenges that come with implementing such a design. As the issue of partial ordering may become more frequent due to the increasing investigations of combination therapies we believe this account will be beneficial to those wishing to implement a design with partial orders. TRIAL REGISTRATION: ADePT-DDR was added to the European Clinical Trials Database (EudraCT number: 2020-001034-35) on 2020-08-07.


Assuntos
Neoplasias de Cabeça e Pescoço , Projetos de Pesquisa , Humanos , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Terapia Combinada , Dose Máxima Tolerável , Relação Dose-Resposta a Droga , Simulação por Computador
9.
Clin Trials ; 21(3): 331-339, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38554038

RESUMO

Combination therapy is increasingly being explored as a promising approach for improving cancer treatment outcomes. However, identifying effective dose combinations in early oncology drug development is challenging due to limited sample sizes in early-phase clinical trials. This task becomes even more complex when multiple agents are being escalated simultaneously, potentially leading to a loss of monotonic toxicity order with respect to the dose. Traditional single-agent trial designs are insufficient for this multi-dimensional problem, necessitating the development and implementation of dose-finding methods specifically designed for drug combinations. While, in practice, approaches to this problem have focused on preselecting combinations with a known toxicity order and applying single-agent designs, this limits the number of combinations considered and may miss promising dose combinations. In recent years, several novel designs have been proposed for exploring partially ordered drug combination spaces with the goal of identifying a maximum tolerated dose combination, based on safety, or an optimal dose combination, based on toxicity and efficacy. However, their implementation in clinical practice remains limited. In this article, we describe the application of the partial order continual reassessment method and its extensions for combination therapies in early-phase clinical trials. We present completed trials that use safety endpoints to identify maximum tolerated dose combinations and adaptively use both safety and efficacy endpoints to determine optimal treatment strategies. We discuss the effectiveness of the partial-order continual reassessment method and its extensions in identifying optimal treatment strategies and provide our experience with executing these novel adaptive designs in practice. By utilizing innovative dose-finding methods, researchers and clinicians can more effectively navigate the challenges of combination therapy development, ultimately improving patient outcomes in the treatment of cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Dose Máxima Tolerável , Neoplasias , Projetos de Pesquisa , Humanos , Neoplasias/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Desenvolvimento de Medicamentos/métodos , Relação Dose-Resposta a Droga , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Ensaios Clínicos como Assunto/métodos
10.
Clin Trials ; 21(3): 267-272, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38570906

RESUMO

With the advent of targeted agents and immunological therapies, the medical research community has become increasingly aware that conventional methods for determining the best dose or schedule of a new agent are inadequate. It has been well established that conventional phase I designs cannot reliably identify safe and effective doses. This problem applies, generally, for cytotoxic agents, radiation therapy, targeted agents, and immunotherapies. To address this, the US Food and Drug Administration's Oncology Center of Excellence initiated Project Optimus, with the goal "to reform the dose optimization and dose selection paradigm in oncology drug development." As a response to Project Optimus, the articles in this special issue of Clinical Trials review recent advances in methods for choosing the dose or schedule of a new agent with an overall objective of informing clinical trialists of these innovative designs. This introductory article briefly reviews problems with conventional methods, the regulatory changes that encourage better dose optimization designs, and provides brief summaries of the articles that follow in this special issue.


Assuntos
Antineoplásicos , Relação Dose-Resposta a Droga , Projetos de Pesquisa , United States Food and Drug Administration , Humanos , Estados Unidos , Antineoplásicos/administração & dosagem , Neoplasias/tratamento farmacológico , Oncologia/métodos , Dose Máxima Tolerável , Ensaios Clínicos Fase I como Assunto/métodos , Desenvolvimento de Medicamentos/métodos
11.
Clin Trials ; 21(3): 322-330, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38591582

RESUMO

Given that novel anticancer therapies have different toxicity profiles and mechanisms of action, it is important to reconsider the current approaches for dose selection. In an effort to move away from considering the maximum tolerated dose as the optimal dose, the Food and Drug Administration Project Optimus points to the need of incorporating long-term toxicity evaluation, given that many of these novel agents lead to late-onset or cumulative toxicities and there are no guidelines on how to handle them. Numerous methods have been proposed to handle late-onset toxicities in dose-finding clinical trials. A summary and comparison of these methods are provided. Moreover, using PI3K inhibitors as a case study, we show how late-onset toxicity can be integrated into the dose-optimization strategy using current available approaches. We illustrate a re-design of this trial to compare the approach to those that only consider early toxicity outcomes and disregard late-onset toxicities. We also provide proposals going forward for dose optimization in early development of novel anticancer agents with considerations for late-onset toxicities.


Assuntos
Antineoplásicos , Relação Dose-Resposta a Droga , Dose Máxima Tolerável , Neoplasias , Humanos , Antineoplásicos/efeitos adversos , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Projetos de Pesquisa , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase/administração & dosagem
12.
Clin Trials ; 21(3): 350-357, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38618916

RESUMO

In the last few years, numerous novel designs have been proposed to improve the efficiency and accuracy of phase I trials to identify the maximum-tolerated dose (MTD) or the optimal biological dose (OBD) for noncytotoxic agents. However, the conventional 3+3 approach, known for its and poor performance, continues to be an attractive choice for many trials despite these alternative suggestions. The article seeks to underscore the importance of moving beyond the 3+3 design by highlighting a different key element in trial design: the estimation of sample size and its crucial role in predicting toxicity and determining the MTD. We use simulation studies to compare the performance of the most used phase I approaches: 3+3, Continual Reassessment Method (CRM), Keyboard and Bayesian Optimal Interval (BOIN) designs regarding three key operating characteristics: the percentage of correct selection of the true MTD, the average number of patients allocated per dose level, and the average total sample size. The simulation results consistently show that the 3+3 algorithm underperforms in comparison to model-based and model-assisted designs across all scenarios and metrics. The 3+3 method yields significantly lower (up to three times) probabilities in identifying the correct MTD, often selecting doses one or even two levels below the actual MTD. The 3+3 design allocates significantly fewer patients at the true MTD, assigns higher numbers to lower dose levels, and rarely explores doses above the target dose-limiting toxicity (DLT) rate. The overall performance of the 3+3 method is suboptimal, with a high level of unexplained uncertainty and significant implications for accurately determining the MTD. While the primary focus of the article is to demonstrate the limitations of the 3+3 algorithm, the question remains about the preferred alternative approach. The intention is not to definitively recommend one model-based or model-assisted method over others, as their performance can vary based on parameters and model specifications. However, the presented results indicate that the CRM, Keyboard, and BOIN designs consistently outperform the 3+3 and offer improved efficiency and precision in determining the MTD, which is crucial in early-phase clinical trials.


Assuntos
Algoritmos , Teorema de Bayes , Ensaios Clínicos Fase I como Assunto , Simulação por Computador , Relação Dose-Resposta a Droga , Dose Máxima Tolerável , Projetos de Pesquisa , Humanos , Tamanho da Amostra , Ensaios Clínicos Fase I como Assunto/métodos , Modelos Estatísticos
13.
J Biopharm Stat ; : 1-17, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38562014

RESUMO

Bayesian logistic regression model (BLRM) is widely used to guide dose escalation decisions in phase 1 oncology trials. An important feature of BLRM design is the appealing safety performance due to its escalation with overdose control (EWOC). However, some recent literature indicates that BLRM with EWOC may have a relatively low probability to find the maximum tolerated dose (MTD) compared to some other dose escalation designs. This work discusses this design problem and proposes a practical solution to improve the performance of BLRM design. Specifically, we suggest increasing the EWOC cutoff from routine value 0.25 to a value between 0.3 and 0.4, which will increase the chance of finding the correct MTD with minimal compromise to overdosing risk. Our comparative simulation studies indicate that BLRM with an increased EWOC cutoff has comparable operating characteristics on the correct MTD selection and over-toxicity control as other dose escalation designs (BOIN, mTPI, keyboard, etc.). Moreover, we compare the methodology and operating characteristics of BLRM designs with various decision rules that allow more flexible overdosing control. A case study of dose escalation in a recent phase 1 oncology trial is provided to show how BLRM with optimal EWOC cutoff operates well in practice.

14.
J Biopharm Stat ; : 1-20, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38888933

RESUMO

We consider a dose-optimization design for a first-in-human oncology trial that aims to identify a suitable dose for late-phase drug development. The proposed approach, called the Pharmacometrics-Enabled DOse OPtimization (PEDOOP) design, incorporates observed patient-level pharmacokinetics (PK) measurements and latent pharmacodynamics (PD) information for trial decision-making and dose optimization. PEDOOP consists of two seamless phases. In phase I, patient-level time-course drug concentrations, derived PD effects, and the toxicity outcomes from patients are integrated into a statistical model to estimate the dose-toxicity response. A simple dose-finding design guides dose escalation in phase I. At the end of the phase I dose finding, a graduation rule is used to assess the safety and efficacy of all the doses and select those with promising efficacy and acceptable safety for a randomized comparison against a control arm in phase II. In phase II, patients are randomized to the selected doses based on a fixed or adaptive randomization ratio. At the end of phase II, an optimal biological dose (OBD) is selected for late-phase development. We conduct simulation studies to assess the PEDOOP design in comparison to an existing seamless design that also combines phases I and II in a single trial.

15.
J Biopharm Stat ; : 1-14, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38335371

RESUMO

Combination therapies with multiple mechanisms of action can offer improved efficacy and/or safety profiles when compared to a single therapy with one mechanism of action. Consequently, the number of combination therapy studies have increased multi-fold, both in oncology and non-oncology indications. However, identifying the optimal doses of each drug in a combination therapy can require a large sample size and prolong study timelines, especially when full factorial designs are used. In this paper, we extend the MCP-Mod design of Bretz, Pinheiro, and Branson to a three-dimensional space to model the dose-response surface of a two-drug combination under the framework of Combination (Comb) MCP-Mod. The resulting model yields a set of dosages for each drug in the combination that elicits the target response so that an optimal dose for the combination can be selected for pivotal studies. We construct three-dimensional dose-response models for the combination and formulate the contrast test statistic to select the best model, which can then be used to select the optimal dose. Guidance to calculate power and sample size calculations are provided to assist study design. Simulation studies show that Comb MCP-Mod performs as well as the conventional multiple comparisons approach in controlling the family-wise error rate at the desired alpha level. However, Comb MCP-Mod is more powerful than the classical multiple comparisons approach in detecting dose-response relationships when treatment is non-null. The probability of correctly identifying the underlying dose-response relationship is generally higher when using Comb MCP-Mod than when using the multiple comparisons approach.

16.
J Biopharm Stat ; : 1-15, 2024 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-39127994

RESUMO

Dose optimization is a critical challenge in drug development. Historically, dose determination in oncology has followed a divergent path from other non-oncology therapeutic areas due to the unique characteristics and requirements in Oncology. However, with the emergence of new drug modalities and mechanisms of drugs in oncology, such as immune therapies, radiopharmaceuticals, targeted therapies, cytostatic agents, and others, the dose-response relationship for efficacy and toxicity could be vastly varied compared to the cytotoxic chemotherapies. The doses below the MTD may demonstrate similar efficacy to the MTD with an improved tolerability profile, resembling what is commonly observed in non-oncology treatments. Hence, alternate strategies for dose optimization are required for new modalities in oncology drug development. This paper delves into the historical evolution of dose finding methods from non-oncology to oncology, highlighting examples and summarizing the underlying drivers of change. Subsequently, a practical framework and guidance are provided to illustrate how dose optimization can be incorporated into various stages of the development program. We provide the following general recommendations: 1) The objective for phase I is to identify a dose range rather than a single MTD dose for subsequent development to better characterize the safety and tolerability profile within the dose range. 2) At least two doses separable by PK are recommended for dose optimization in phase II. 3) Ideally, dose optimization should be performed before launching the confirmatory study. Nevertheless, innovative designs such as seamless II/III design can be implemented for dose selection and may accelerate the drug development program.

17.
Pharm Stat ; 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769904

RESUMO

This paper proposes a trial design for locating group-specific doses when groups are partially or completely ordered by dose sensitivity. Previous trial designs for partially ordered groups are model-based, whereas the proposed method is model-assisted, providing clinicians with a design that is simpler. The proposed method performs similarly to model-based methods, providing simplicity without losing accuracy. Additionally, to the best of our knowledge, the proposed method is the first paper on dose-finding for partially ordered groups with convergence results. To generalize the proposed method, a framework is introduced that allows partial orders to be transferred to a grid format with a known ordering across rows but an unknown ordering within rows.

18.
Pharm Stat ; 23(4): 585-594, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38317370

RESUMO

The Bayesian logistic regression method (BLRM) is a widely adopted and flexible design for finding the maximum tolerated dose in oncology phase I studies. However, the BLRM design has been criticized in the literature for being overly conservative due to the use of the overdose control rule. Recently, a discussion paper titled "Improving the performance of Bayesian logistic regression model with overall control in oncology dose-finding studies" in Statistics in Medicine has proposed an overall control rule to address the "excessive conservativeness" of the standard BLRM design. In this short communication, we discuss the relative conservativeness of the standard BLRM design and also suggest a dose-switching rule to further enhance its performance.


Assuntos
Antineoplásicos , Teorema de Bayes , Ensaios Clínicos Fase I como Assunto , Relação Dose-Resposta a Droga , Dose Máxima Tolerável , Humanos , Modelos Logísticos , Ensaios Clínicos Fase I como Assunto/métodos , Ensaios Clínicos Fase I como Assunto/estatística & dados numéricos , Antineoplásicos/administração & dosagem , Neoplasias/tratamento farmacológico , Projetos de Pesquisa
19.
Pharm Stat ; 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39119879

RESUMO

Dose-finding studies play a crucial role in drug development by identifying the optimal dose(s) for later studies while considering tolerability. This not only saves time and effort in proceeding with Phase III trials but also improves efficacy. In an era of precision medicine, it is not ideal to assume patient homogeneity in dose-finding studies as patients may respond differently to the drug. To address this, we propose a personalized dose-finding algorithm that assigns patients to individualized optimal biological doses. Our design follows a two-stage approach. Initially, patients are enrolled under broad eligibility criteria. Based on the Stage 1 data, we fit a regression model of toxicity and efficacy outcomes on dose and biomarkers to characterize treatment-sensitive patients. In the second stage, we restrict the trial population to sensitive patients, apply a personalized dose allocation algorithm, and choose the recommended dose at the end of the trial. Simulation study shows that the proposed design reliably enriches the trial population, minimizes the number of failures, and yields superior operating characteristics compared to several existing dose-finding designs in terms of both the percentage of correct selection and the number of patients treated at target dose(s).

20.
Pharm Stat ; 23(4): 480-494, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38295856

RESUMO

For novel immuno-oncology therapies, the primary purpose of a dose-finding trial is to identify an optimal dose (OD), defined as the tolerable dose having adequate efficacy and immune response under the unpredictable dose-outcome (toxicity, efficacy, and immune response) relationships. In addition, the multiple low or moderate-grade toxicities rather than dose-limiting toxicities (DLTs) and multiple levels of efficacy should be evaluated differently in dose-finding to determine true OD for developing novel immuno-oncology therapies. We proposed a generalized Bayesian optimal interval design for immunotherapy, simultaneously considering efficacy and toxicity grades and immune response outcomes. The proposed design, named gBOIN-ETI design, is model-assisted and easy to implement to develop immunotherapy efficiently. The operating characteristics of the gBOIN-ETI are compared with other dose-finding trial designs in oncology by simulation across various realistic settings. Our simulations show that the gBOIN-ETI design could outperform the other available approaches in terms of both the percentage of correct OD selection and the average number of patients allocated to the OD across various realistic trial settings.


Assuntos
Teorema de Bayes , Simulação por Computador , Relação Dose-Resposta a Droga , Imunoterapia , Projetos de Pesquisa , Humanos , Imunoterapia/métodos , Imunoterapia/efeitos adversos , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Dose Máxima Tolerável , Modelos Estatísticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA