1
|
Zhan T, Kang J. A general, flexible, and harmonious framework to construct interpretable functions in regression analysis. Biometrics 2025; 81:ujaf014. [PMID: 40037599 DOI: 10.1093/biomtc/ujaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/04/2024] [Accepted: 02/05/2025] [Indexed: 03/06/2025]
Abstract
An interpretable model or method has several appealing features, such as reliability to adversarial examples, transparency of decision-making, and communication facilitator. However, interpretability is a subjective concept, and even its definition can be diverse. The same model may be deemed as interpretable by a study team, but regarded as a black-box algorithm by another squad. Simplicity, accuracy and generalizability are some additional important aspects of evaluating interpretability. In this work, we present a general, flexible and harmonious framework to construct interpretable functions in regression analysis with a focus on continuous outcomes. We formulate a functional skeleton in light of users' expectations of interpretability. A new measure based on Mallows's $C_p$-statistic is proposed for model selection to balance approximation, generalizability, and interpretability. We apply this approach to derive a sample size formula in adaptive clinical trial designs to demonstrate the general workflow, and to explain operating characteristics in a Bayesian Go/No-Go paradigm to show the potential advantages of using meaningful intermediate variables. Generalization to categorical outcomes is illustrated in an example of hypothesis testing based on Fisher's exact test. A real data analysis of NHANES (National Health and Nutrition Examination Survey) is conducted to investigate relationships between some important laboratory measurements. We also discuss some extensions of this method.
Collapse
Affiliation(s)
- Tianyu Zhan
- Data and Statistical Sciences, AbbVie Inc., 1 Waukegan Road, North Chicago, IL 60064, United States
| | - Jian Kang
- Department of Biostatistics, University of Michigan Ann Arbor, 1415 Washington Heights, Ann Arbor, MI 48109, United States
| |
Collapse
|
2
|
Chen J, Takanami Y, Jansson J, Rossiter G. Practical considerations of promising zone design for interim sample size Re-estimation: An application to GRAPHITE for graft vs host disease. Contemp Clin Trials 2025; 148:107765. [PMID: 39603384 DOI: 10.1016/j.cct.2024.107765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/07/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Sample size calculation and power estimate are an integral part of clinical trials. With accelerated development to address the unmet medical needs, the fast-paced development may lead to uncertainties in initial planning and assumptions of clinical trials. Promising zone design presents sponsors an opportunity to re-estimate the sample size based on the interim data to mitigate risks, reduce uncertainties, and increase probability of trial success. METHODS This paper aims to use the GRAPHITE trial (NCT03657160) as a real data application to showcase the practical considerations in implementation of promising zone design for interim sample size re-estimation (SSR), in light of sample size adaptation rules, maximum sample size allowed, multiplicity adjustment, and sponsor access to interim results. GRAPHITE is a phase 3 trial with vedolizumab for prophylaxis of acute graft vs host disease (aGvHD) after allogeneic hematopoietic stem cell transplant (allo-HSCT). The primary efficacy endpoint is lower intestinal aGVHD-free survival by Day +180 after allo-HSCT. A simulation study was conducted to demonstrate the evaluation of operating characteristics by various true underlying treatment effects at the design stage. CONCLUSION The application of promising zone design for interim SSR is novel and has successfully helped the sponsor achieve the balance between minimizing the risks and maintaining scientific integrity. This work aims to highlight the necessity of empirical guidance to gain better insights for clinical researchers in practice and is expected to facilitate the understanding and implementation of promising zone design for interim SSR in phase 3 trials.
Collapse
Affiliation(s)
- Jingjing Chen
- Takeda Development Center Americas, Inc., Cambridge, MA, USA.
| | | | - Johan Jansson
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | |
Collapse
|
3
|
Abrams SA, Ernst KD, Weitkamp JH, Mascarenhas M, Anderson-Berry A, Rudolph J, Ling CY, Robinson DT, Shores D, Hair AB, Lai J, Lane B, McCallie KR, Levit O, Kim JH. Safety and Efficacy of a Composite Lipid Emulsion with Fish Oil in Hospitalized Neonates and Infants Requiring Prolonged Parenteral Nutrition - A Randomized, Double-Blind, Multicenter, Controlled Trial. J Nutr 2024; 154:3615-3625. [PMID: 39374788 DOI: 10.1016/j.tjnut.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/14/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Intravenous lipids are critical to the care of extremely premature and other high-risk infants. OBJECTIVES This study evaluated safety and efficacy of parenteral nutrition (PN) with composite intravenous lipid emulsion (CO-ILE) with fish oil compared with pure soybean oil lipid emulsion (SOLE). METHODS Randomized, controlled, double-blind, multicenter study (NCT02579265) in neonates/infants anticipated to require ≥28 d of PN due to gastrointestinal malformations or injury. Duration of the initial and extended treatment phase was 28 d and 84 d, respectively (for patients with PN indication after day 28). RESULTS Eighty-three patients (mean postnatal age 11.4 d, 54 preterm) received CO-ILE and 78 patients received SOLE (mean postnatal age 8.3 d, 59 preterm). Thirty-three patients per group completed 28 d of treatment. Risk of having conjugated bilirubin values >2 mg/dL confirmed by a second sample 7 d after the first during the initial treatment phase (primary outcome) was 2.4% (2 of 83) with CO-ILE and 3.8% (3 of 78) with SOLE (risk ratio: 0.59; 95% confidence interval [CI]: 0.09, 3.76). Between days 29 and 84, the number of patients with confirmed conjugated bilirubin values >2 mg/dL did not increase in the CO-ILE group (n = 2) and increased in the SOLE group (n = 9). At the end of the initial treatment phase, conjugated bilirubin concentrations were 45.6% lower under CO-ILE than under SOLE (P = 0.006). There was no clinical or laboratory evidence of essential fatty acid deficiency in patients in the CO-ILE group. Median time to discharge alive was 56.7 d and 66.4 d with CO-ILE and SOLE, respectively (hazard ratio: 1.16; 95% CI: 0.81, 1.68). CONCLUSIONS CO-ILE was associated with a possible lower risk of cholestasis and significantly lower conjugated bilirubin concentration at the end of the initial treatment phase in high-risk neonates and infants as compared with patients treated with SOLE. In summary, these data indicate that CO-ILE can be considered safe and may be preferable over SOLE in high-risk neonates. This trial was registered at clinicaltrials.gov as NCT02579265.
Collapse
Affiliation(s)
- Steven A Abrams
- Department of Pediatrics, Dell Medical School at the University of Texas-Austin, Austin, TX, United States.
| | - Kimberly D Ernst
- Division of Neonatal-Perinatal Medicine, The University of Oklahoma Children's Hospital, Oklahoma City, OK, United States
| | - Joern-Hendrik Weitkamp
- Department of Pediatrics, Mildred Stahlman Divison of Neonatology, Vanderbilt University Medical Center, Monroe Carrol Jr. Children's Hospital, Nashville, TN, United States
| | - Maria Mascarenhas
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, and Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ann Anderson-Berry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jeffrey Rudolph
- Division of Gastroenterology, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Con Y Ling
- Department of Pediatrics, University of Utah and Division of Neonatology at Primary Children's Hospital, Salt Lake City, UT, United States
| | - Daniel T Robinson
- Department of Pediatrics, Northwestern University Feinberg School of Medicine and Division of Neonatology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Darla Shores
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Amy B Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Joanne Lai
- Department of Pediatric Gastroenterology, Susan and Leonard Feinstein IBD Clinical Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Brian Lane
- Division of Neonatology, Departments of Pediatrics, University of California, San Diego and Rady Children's Hospital San Diego, San Diego, CA, United States
| | - Katherine R McCallie
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
| | - Orly Levit
- Neonatal-Perinatal Medicine, General Neonatology, Yale New Haven Children's Hospital, New Haven, CT, United States
| | - Jae H Kim
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
4
|
Held L. Beyond the two-trials rule. Stat Med 2024; 43:5023-5042. [PMID: 38573319 DOI: 10.1002/sim.10055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/10/2023] [Accepted: 12/10/2023] [Indexed: 04/05/2024]
Abstract
The two-trials rule for drug approval requires "at least two adequate and well-controlled studies, each convincing on its own, to establish effectiveness." This is usually implemented by requiring two significant pivotal trials and is the standard regulatory requirement to provide evidence for a new drug's efficacy. However, there is need to develop suitable alternatives to this rule for a number of reasons, including the possible availability of data from more than two trials. I consider the case of up to three studies and stress the importance to control the partial Type-I error rate, where only some studies have a true null effect, while maintaining the overall Type-I error rate of the two-trials rule, where all studies have a null effect. Some less-knownP $$ P $$ -value combination methods are useful to achieve this: Pearson's method, Edgington's method and the recently proposed harmonic meanχ 2 $$ {\chi}^2 $$ -test. I study their properties and discuss how they can be extended to a sequential assessment of success while still ensuring overall Type-I error control. I compare the different methods in terms of partial Type-I error rate, project power and the expected number of studies required. Edgington's method is eventually recommended as it is easy to implement and communicate, has only moderate partial Type-I error rate inflation but substantially increased project power.
Collapse
Affiliation(s)
- Leonhard Held
- Epidemiology, Biostatistics and Prevention Institute (EBPI) and Center for Reproducible Science (CRS), University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Berry LR, Marion J, Berry SM, Viele K. Optimal sample size division in two-stage seamless designs. Pharm Stat 2024; 23:854-863. [PMID: 38676420 DOI: 10.1002/pst.2394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 02/21/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024]
Abstract
Inferentially seamless 2/3 designs are increasingly popular in clinical trials. It is important to understand their relative advantages compared with separate phase 2 and phase 3 trials, and to understand the consequences of design choices such as the proportion of patients included in the phase 2 portion of the design. Extending previous work in this area, we perform a simulation study across multiple numbers of arms and efficacy response curves. We consider a design space crossing the choice of a separate versus seamless design with the choice of allocating 0%-100% of available patients in phase 2, with the remainder in phase 3. The seamless designs achieve greater power than their separate trial counterparts. Importantly, the optimal seamless design is more robust than the optimal separate program, meaning that one range of values for the proportion of patients used in phase 2 (30%-50% of the total phase 2/3 sample size) is nearly optimal for a wide range of response scenarios. In contrast, a percentage of patients used in phase 2 for separate trials may be optimal for some alternative scenarios but decidedly inferior for other alternative scenarios. When operationally and scientifically viable, seamless trials provide superior performance compared with separate phase 2 and phase 3 trials. The results also provide guidance for the implementation of these trials in practice.
Collapse
Affiliation(s)
| | - Joe Marion
- Berry Consultants, LLC, Austin, Texas, USA
| | - Scott M Berry
- Berry Consultants, LLC, Austin, Texas, USA
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kert Viele
- Berry Consultants, LLC, Austin, Texas, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
6
|
Li R, Wu L, Liu R, Lin J. Flexible seamless 2-in-1 design with sample size adaptation. J Biopharm Stat 2024; 34:1007-1025. [PMID: 38549502 DOI: 10.1080/10543406.2024.2330211] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 01/08/2024] [Indexed: 11/29/2024]
Abstract
The 2-in-1 design is becoming popular in oncology drug development, with the flexibility in using different endpoints at different decision time. Based on the observed interim data, sponsors can choose to seamlessly advance a small phase 2 trial to a full-scale confirmatory phase 3 trial with a pre-determined maximum sample size or remain in a phase 2 trial. While this approach may increase efficiency in drug development, it is rigid and requires a pre-specified fixed sample size. In this paper, we propose a flexible 2-in-1 design with sample size adaptation, while retaining the advantage of allowing an intermediate endpoint for interim decision-making. The proposed design reflects the needs of the recent FDA's Project FrontRunner initiative, which encourages the use of an earlier surrogate endpoint to potentially support accelerated approval with conversion to standard approval with long-term endpoints from the same randomized study. Additionally, we identify the interim decision cut-off to allow a conventional test procedure at the final analysis. Extensive simulation studies showed that the proposed design requires much a smaller sample size and shorter timeline than the simple 2-in-1 design, while achieving similar power. We present a case study in multiple myeloma to demonstrate the benefits of the proposed design.
Collapse
Affiliation(s)
- Runjia Li
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Liwen Wu
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Rachael Liu
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Jianchang Lin
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| |
Collapse
|
7
|
Dong Y, Paux G, Broglio K, Cooner F, Gao G, He W, Gao L, Xue X, He P. Use of Seamless Study Designs in Oncology Clinical Development- A Survey Conducted by IDSWG Oncology Sub-team. Ther Innov Regul Sci 2024; 58:978-986. [PMID: 38909174 DOI: 10.1007/s43441-024-00676-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/07/2024] [Indexed: 06/24/2024]
Abstract
Seamless study designs have the potential to accelerate clinical development. The use of innovative seamless designs has been increasing in the oncology area; however, while the concept of seamless designs becomes more popular and accepted, many challenges remain in both the design and conduct of these trials. This may be especially true when seamless designs are used in late phase development supporting regulatory decision-making. The Innovative Design Scientific Working Group (IDSWG) Oncology team conducted a survey to understand the current use of seamless study designs for registration purposes in oncology clinical development. The survey was designed to provide insights into the benefits and to identify the roadblocks. A total of 16 questions were included in the survey that was distributed using the ASA Biopharmaceutical Section and IDSWG email listings from August to September 2022. A total of 51 responses were received, with 39 (76%) respondents indicating that their organizations had seamless oncology studies in planning or implementation for registration purposes. Detailed survey results are presented in the manuscript. Overall, while seamless designs offer advantages in terms of timeline reduction and cost saving, they also present challenges related to additional complexity and the need for efficient surrogate clinical endpoints in oncology drug development.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei He
- AstraZeneca, Cambridge, MA, USA
| | - Lei Gao
- Moderna, Inc, Cambridge, MA, USA
| | | | - Philip He
- Daiichi Sankyo, Basking Ridge, NJ, USA
| |
Collapse
|
8
|
Greenstreet P, Jaki T, Bedding A, Harbron C, Mozgunov P. A multi-arm multi-stage platform design that allows preplanned addition of arms while still controlling the family-wise error. Stat Med 2024; 43:3613-3632. [PMID: 38880949 DOI: 10.1002/sim.10135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/18/2024]
Abstract
There is growing interest in platform trials that allow for adding of new treatment arms as the trial progresses as well as being able to stop treatments part way through the trial for either lack of benefit/futility or for superiority. In some situations, platform trials need to guarantee that error rates are controlled. This paper presents a multi-stage design, that allows additional arms to be added in a platform trial in a preplanned fashion, while still controlling the family-wise error rate, under the assumption of known number and timing of treatments to be added, and no time trends. A method is given to compute the sample size required to achieve a desired level of power and we show how the distribution of the sample size and the expected sample size can be found. We focus on power under the least favorable configuration which is the power of finding the treatment with a clinically relevant effect out of a set of treatments while the rest have an uninteresting treatment effect. A motivating trial is presented which focuses on two settings, with the first being a set number of stages per active treatment arm and the second being a set total number of stages, with treatments that are added later getting fewer stages. Compared to Bonferroni, the savings in the total maximum sample size are modest in a trial with three arms, <1% of the total sample size. However, the savings are more substantial in trials with more arms.
Collapse
Affiliation(s)
- Peter Greenstreet
- Department of Mathematics and Statistics, Lancaster University, Lancaster, UK
- Exeter Clinical Trials Unit, University of Exeter, Exeter, UK
| | - Thomas Jaki
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- University of Regensburg, Regensburg, Germany
| | | | | | - Pavel Mozgunov
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Boumendil L, Chevret S, Lévy V, Biard L. Two-stage randomized clinical trials with a right-censored endpoint: Comparison of frequentist and Bayesian adaptive designs. Stat Med 2024; 43:3364-3382. [PMID: 38844988 DOI: 10.1002/sim.10130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 07/17/2024]
Abstract
Adaptive randomized clinical trials are of major interest when dealing with a time-to-event outcome in a prolonged observation window. No consensus exists either to define stopping boundaries or to combinep $$ p $$ values or test statistics in the terminal analysis in the case of a frequentist design and sample size adaptation. In a one-sided setting, we compared three frequentist approaches using stopping boundaries relying onα $$ \alpha $$ -spending functions and a Bayesian monitoring setting with boundaries based on the posterior distribution of the log-hazard ratio. All designs comprised a single interim analysis with an efficacy stopping rule and the possibility of sample size adaptation at this interim step. Three frequentist approaches were defined based on the terminal analysis: combination of stagewise statistics (Wassmer) or ofp $$ p $$ values (Desseaux), or on patientwise splitting (Jörgens), and we compared the results with those of the Bayesian monitoring approach (Freedman). These different approaches were evaluated in a simulation study and then illustrated on a real dataset from a randomized clinical trial conducted in elderly patients with chronic lymphocytic leukemia. All approaches controlled for the type I error rate, except for the Bayesian monitoring approach, and yielded satisfactory power. It appears that the frequentist approaches are the best in underpowered trials. The power of all the approaches was affected by the violation of the proportional hazards (PH) assumption. For adaptive designs with a survival endpoint and a one-sided alternative hypothesis, the Wassmer and Jörgens approaches after sample size adaptation should be preferred, unless violation of PH is suspected.
Collapse
Affiliation(s)
- Luana Boumendil
- INSERM U1153, Team ECSTRRA, Hôpital Saint Louis, Paris, France
- Université Paris Cité, Paris, France
- AP-HP Hôpital Saint Louis, Service de Biostatistique et Information Médicale, Paris, France
| | - Sylvie Chevret
- INSERM U1153, Team ECSTRRA, Hôpital Saint Louis, Paris, France
- Université Paris Cité, Paris, France
- AP-HP Hôpital Saint Louis, Service de Biostatistique et Information Médicale, Paris, France
| | - Vincent Lévy
- INSERM U1153, Team ECSTRRA, Hôpital Saint Louis, Paris, France
- Université Paris 13, Villetaneuse, France
- AP-HP Hôpital Avicenne, Unité de Recherche Clinique Bobigny, Bobigny, France
| | - Lucie Biard
- INSERM U1153, Team ECSTRRA, Hôpital Saint Louis, Paris, France
- Université Paris Cité, Paris, France
- AP-HP Hôpital Saint Louis, Service de Biostatistique et Information Médicale, Paris, France
| |
Collapse
|
10
|
Burnett T, König F, Jaki T. Adding experimental treatment arms to multi-arm multi-stage platform trials in progress. Stat Med 2024; 43:3447-3462. [PMID: 38852991 DOI: 10.1002/sim.10090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/16/2024] [Accepted: 04/15/2024] [Indexed: 06/11/2024]
Abstract
Multi-arm multi-stage (MAMS) platform trials efficiently compare several treatments with a common control arm. Crucially MAMS designs allow for adjustment for multiplicity if required. If for example, the active treatment arms in a clinical trial relate to different dose levels or different routes of administration of a drug, the strict control of the family-wise error rate (FWER) is paramount. Suppose a further treatment becomes available, it is desirable to add this to the trial already in progress; to access both the practical and statistical benefits of the MAMS design. In any setting where control of the error rate is required, we must add corresponding hypotheses without compromising the validity of the testing procedure.To strongly control the FWER, MAMS designs use pre-planned decision rules that determine the recruitment of the next stage of the trial based on the available data. The addition of a treatment arm presents an unplanned change to the design that we must account for in the testing procedure. We demonstrate the use of the conditional error approach to add hypotheses to any testing procedure that strongly controls the FWER. We use this framework to add treatments to a MAMS trial in progress. Simulations illustrate the possible characteristics of such procedures.
Collapse
Affiliation(s)
- Thomas Burnett
- Department of Mathematical Sciences, University of Bath, Bath, UK
| | - Franz König
- Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Thomas Jaki
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Faculty of Computer Science and Data Science, University of Regensburg, Regensburg, Germany
| |
Collapse
|
11
|
Held L, Pawel S, Micheloud C. The assessment of replicability using the sum of p-values. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240149. [PMID: 39205991 PMCID: PMC11349439 DOI: 10.1098/rsos.240149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 09/04/2024]
Abstract
Statistical significance of both the original and the replication study is a commonly used criterion to assess replication attempts, also known as the two-trials rule in drug development. However, replication studies are sometimes conducted although the original study is non-significant, in which case Type-I error rate control across both studies is no longer guaranteed. We propose an alternative method to assess replicability using the sum of p -values from the two studies. The approach provides a combined p -value and can be calibrated to control the overall Type-I error rate at the same level as the two-trials rule but allows for replication success even if the original study is non-significant. The unweighted version requires a less restrictive level of significance at replication if the original study is already convincing which facilitates sample size reductions of up to 10%. Downweighting the original study accounts for possible bias and requires a more stringent significance level and larger sample sizes at replication. Data from four large-scale replication projects are used to illustrate and compare the proposed method with the two-trials rule, meta-analysis and Fisher's combination method.
Collapse
Affiliation(s)
- Leonhard Held
- Epidemiology Biostatistics and Prevention Institute (EBPI) and Center for Reproducible Science (CRS), University of Zurich, Hirschengraben 84, Zurich8001, Switzerland
| | - Samuel Pawel
- Epidemiology Biostatistics and Prevention Institute (EBPI) and Center for Reproducible Science (CRS), University of Zurich, Hirschengraben 84, Zurich8001, Switzerland
| | - Charlotte Micheloud
- Epidemiology Biostatistics and Prevention Institute (EBPI) and Center for Reproducible Science (CRS), University of Zurich, Hirschengraben 84, Zurich8001, Switzerland
| |
Collapse
|
12
|
Poli S, Grohmann C, Wenzel DA, Poli K, Tünnerhoff J, Nedelmann M, Fiehler J, Burghaus I, Lehmann M, Glauch M, Schadwinkel HM, Kalmbach P, Zeller J, Peters T, Eschenfelder C, Agostini H, Campbell BC, Fischer MD, Sykora M, Mac Grory B, Feltgen N, Kowarik M, Seiffge D, Strbian D, Albrecht M, Alzureiqi MS, Auffarth G, Bäzner H, Behnke S, Berberich A, Bode F, Bohmann FO, Cheng B, Czihal M, Danyel LA, Dimopoulos S, Pinhal Ferreira de Pinho JD, Fries FN, Gamulescu MA, Gekeler F, Gomez-Exposito A, Gumbinger C, Guthoff R, Hattenbach LO, Kellert L, Khoramnia R, Kohnen T, Kürten D, Lackner B, Laible M, Lee JI, Leithner C, Liegl R, Lochner P, Mackert M, Mbroh J, Müller S, Nagel S, Prasuhn M, Purrucker J, Reich A, Mundiyanapurath S, Royl G, Salchow DJ, Schäfer JH, Schlachetzki F, Schmack I, Thomalla G, Tieck Fernandez MP, Wakili P, Walter P, Wolf A, Wolf M, Bartz-Schmidt KU, Schultheiss M, Spitzer MS. Early REperfusion therapy with intravenous alteplase for recovery of VISION in acute central retinal artery occlusion (REVISION): Study protocol of a phase III trial. Int J Stroke 2024; 19:823-829. [PMID: 38591748 DOI: 10.1177/17474930241248516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
RATIONALE Meta-analyses of case series of non-arteritic central retinal artery occlusion (CRAO) indicate beneficial effects of intravenous thrombolysis when initiated early after symptom onset. Randomized data are lacking to address this question. AIMS The REperfusion therapy with intravenous alteplase for recovery of VISION in acute central retinal artery occlusion (REVISION) investigates intravenous alteplase within 4.5 h of monocular vision loss due to acute CRAO. METHODS This study is the randomized (1:1), double-blind, placebo-controlled, multicenter adaptive phase III trial. STUDY OUTCOMES Primary outcome is functional recovery to normal or mildly impaired vision in the affected eye defined as best-corrected visual acuity of the Logarithm of the Minimum Angle of Resolution of 0.5 or less at 30 days (intention-to-treat analysis). Secondary efficacy outcomes include modified Rankin Score at 90 days and quality of life. Safety outcomes include symptomatic intracranial hemorrhage, major bleeding (International Society on Thrombosis and Haemostasis definition) and mortality. Exploratory analyses of optical coherence tomography/angiography, ultrasound and magnetic resonance imaging (MRI) biomarkers will be conducted. SAMPLE SIZE Using an adaptive design with interim analysis at 120 patients, up to 422 participants (211 per arm) would be needed for 80% power (one-sided alpha = 0.025) to detect a difference of 15%, assuming functional recovery rates of 10% in the placebo arm and 25% in the alteplase arm. DISCUSSION By enrolling patients within 4.5 h of CRAO onset, REVISION uses insights from meta-analyses of CRAO case series and randomized thrombolysis trials in acute ischemic stroke. Increased rates of early reperfusion and good neurological outcomes in stroke may translate to CRAO with its similar pathophysiology. TRIAL REGISTRATION ClinicalTrials.gov: NCT04965038; EU Trial Number: 2023-507388-21-00.
Collapse
Affiliation(s)
- Sven Poli
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Carsten Grohmann
- Department of Ophthalmology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel A Wenzel
- Department of Ophthalmology, University of Tübingen, Germany
| | - Khouloud Poli
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Johannes Tünnerhoff
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Max Nedelmann
- Department of Neurology, Regio Kliniken GmbH, Pinneberg, Germany
| | - Jens Fiehler
- Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Eppdata GmbH, Hamburg, Germany
| | - Ina Burghaus
- Coordination Centre for Clinical Trials (KKS), Medical Faculty & Heidelberg University Hospital, Germany
| | - Monika Lehmann
- Coordination Centre for Clinical Trials (KKS), Medical Faculty & Heidelberg University Hospital, Germany
| | - Monika Glauch
- Center for Rare Diseases, University of Tübingen, Germany
| | - Hauke M Schadwinkel
- Department of Ophthalmology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pia Kalmbach
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Julia Zeller
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Tobias Peters
- Department of Ophthalmology, University of Tübingen, Germany
| | | | | | - Bruce Cv Campbell
- Department of Medicine and Neurology, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - M Dominik Fischer
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, UK
- Nuffield Laboratory of Ophthalmology, University of Oxford, UK
| | - Marek Sykora
- Department of Neurology, St. John's Hospital, Vienna, Austria
- Medical Faculty, Sigmund Freud University, Vienna, Austria
| | - Brian Mac Grory
- Duke Clinical Research Institute, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Nicolas Feltgen
- Department of Ophthalmology, Universitätsspital Basel, Switzerland
| | - Markus Kowarik
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - David Seiffge
- Department of Neurology, University of Bern, Switzerland
| | - Daniel Strbian
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Finland
| | | | - Mohammad S Alzureiqi
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gerd Auffarth
- Department of Ophthalmology, University of Heidelberg, Germany
| | | | - Stefanie Behnke
- Department of Neurology, Klinik Sulzbach, Knappschaftsklinikum Saar, Germany
| | | | - Felix Bode
- Department of Neurology, University Hospital Bonn, Germany
| | - Ferdinand O Bohmann
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Bastian Cheng
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Czihal
- Medical Clinic and Policlinic IV, Division of Vascular Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Leon A Danyel
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | | | - Fabian N Fries
- Department of Ophthalmology, Saarland University Medical Center, Homburg/Saar, Germany
| | | | | | - Alexandra Gomez-Exposito
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | | | - Rainer Guthoff
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | | | - Lars Kellert
- Department of Neurology, Ludwig Maximilian University (LMU), Munich, Germany
| | - Ramin Khoramnia
- Department of Ophthalmology, University of Heidelberg, Germany
| | - Thomas Kohnen
- Department of Ophthalmology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - David Kürten
- Department of Ophthalmology, University Hospital, RWTH Aachen University, Aachen, Germany
| | | | - Mona Laible
- Department of Neurology, University Hospital Ulm, Germany
| | - John-Ih Lee
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Christoph Leithner
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raffael Liegl
- Department of Ophthalmology, University Hospital Bonn, Germany
| | - Piergiorgio Lochner
- Department of Neurology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Marc Mackert
- Department of Ophthalmology, Ludwig Maximilian University (LMU), Munich, Germany
| | - Joshua Mbroh
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Susanne Müller
- Department of Neurology, University Hospital Ulm, Germany
| | - Simon Nagel
- Department of Neurology, Klinikum Ludwigshafen, Germany
| | - Michelle Prasuhn
- Department of Ophthalmology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany
| | - Jan Purrucker
- Department of Neurology, University of Heidelberg, Germany
| | - Arno Reich
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany
| | | | - Georg Royl
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany
| | - Daniel J Salchow
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan H Schäfer
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | | | - Ingo Schmack
- Department of Ophthalmology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria P Tieck Fernandez
- Department of Neurology & Stroke, University of Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Philip Wakili
- Department of Ophthalmology, Klinik Sulzbach, Knappschaftsklinikum Saar, Germany
| | - Peter Walter
- Department of Ophthalmology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Armin Wolf
- Department of Ophthalmology, Klinik Sulzbach, Knappschaftsklinikum Saar, Germany
| | - Marc Wolf
- Department of Neurology, Klinikum Stuttgart, Germany
| | | | - Maximilian Schultheiss
- Department of Ophthalmology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
- AugenChirurgie München, Eye Clinic Herzog Carl Theodor, Munich, Germany
| | - Martin S Spitzer
- Department of Ophthalmology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
13
|
Lee KM, Emsley R. The impact of heterogeneity on the analysis of platform trials with normally distributed outcomes. BMC Med Res Methodol 2024; 24:163. [PMID: 39080538 PMCID: PMC11290279 DOI: 10.1186/s12874-024-02293-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND A platform trial approach allows adding arms to on-going trials to speed up intervention discovery programs. A control arm remains open for recruitment in a platform trial while intervention arms may be added after the onset of the study and could be terminated early for efficacy and/or futility when early stopping is allowed. The topic of utilising non-concurrent control data in the analysis of platform trials has been explored and discussed extensively. A less familiar issue is the presence of heterogeneity, which may exist for example due to modification of enrolment criteria and recruitment strategy. METHOD We conduct a simulation study to explore the impact of heterogeneity on the analysis of a two-stage platform trial design. We consider heterogeneity in treatment effects and heteroscedasticity in outcome data across stages for a normally distributed endpoint. We examine the performance of some hypothesis testing procedures and modelling strategies. The use of non-concurrent control data is also considered accordingly. Alongside standard regression analysis, we examine the performance of a novel method that was known as the pairwise trials analysis. It is similar to a network meta-analysis approach but adjusts for treatment comparisons instead of individual studies using fixed effects. RESULTS Several testing strategies with concurrent control data seem to control the type I error rate at the required level when there is heteroscedasticity in outcome data across stages and/or a random cohort effect. The main parameter of treatment effects in some analysis models correspond to overall treatment effects weighted by stage wise sample sizes; while others correspond to the effect observed within a single stage. The characteristics of the estimates are not affected significantly by the presence of a random cohort effect and/ or heteroscedasticity. CONCLUSION In view of heterogeneity in treatment effect across stages, the specification of null hypotheses in platform trials may need to be more subtle. We suggest employing testing procedure of adaptive design as opposed to testing the statistics from regression models; comparing the estimates from the pairwise trials analysis method and the regression model with interaction terms may indicate if heterogeneity is negligible.
Collapse
Affiliation(s)
- Kim May Lee
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK.
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Richard Emsley
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
14
|
Cui L. Sample size adaptation designs and efficiency comparison with group sequential designs. Stat Med 2024; 43:2203-2215. [PMID: 38545849 DOI: 10.1002/sim.10066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/25/2024] [Accepted: 03/12/2024] [Indexed: 05/18/2024]
Abstract
This study is to give a systematic account of sample size adaptation designs (SSADs) and to provide direct proof of the efficiency advantage of general SSADs over group sequential designs (GSDs) from a different perspective. For this purpose, a class of sample size mapping functions to define SSADs is introduced. Under the two-stage adaptive clinical trial setting, theorems are developed to describe the properties of SSADs. Sufficient conditions are derived and used to prove analytically that SSADs based on the weighted combination test can be uniformly more efficient than GSDs in a range of likely values of the true treatment differenceδ $$ \delta $$ . As shown in various scenarios, given a GSD, a fully adaptive SSAD can be obtained that has sufficient statistical power similar to that of the GSD but has a smaller average sample size for allδ $$ \delta $$ in the range. The associated sample size savings can be substantial. A practical design example and suggestions on the steps to find efficient SSADs are also provided.
Collapse
Affiliation(s)
- Lu Cui
- Independent Researcher, Washington DC, USA
| |
Collapse
|
15
|
Holubovska O, Babich P, Mironenko A, Milde J, Lebed Y, Stammer H, Mueller L, te Velthuis AJW, Margitich V, Goy A. RNA Polymerase Inhibitor Enisamium for Treatment of Moderate COVID-19 Patients: A Randomized, Placebo-Controlled, Multicenter, Double-Blind Phase 3 Clinical Trial. Adv Respir Med 2024; 92:202-217. [PMID: 38804439 PMCID: PMC11130936 DOI: 10.3390/arm92030021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/14/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024]
Abstract
Enisamium is an orally available therapeutic that inhibits influenza A virus and SARS-CoV-2 replication. We evaluated the clinical efficacy of enisamium treatment combined with standard care in adult, hospitalized patients with moderate COVID-19 requiring external oxygen. Hospitalized patients with laboratory-confirmed SARS-CoV-2 infection were randomly assigned to receive either enisamium (500 mg per dose, four times a day) or a placebo. The primary outcome was an improvement of at least two points on an eight-point severity rating (SR) scale within 29 days of randomization. We initially set out to study the effect of enisamium on patients with a baseline SR of 4 or 5. However, because the study was started early in the COVID-19 pandemic, and COVID-19 had been insufficiently studied at the start of our study, an interim analysis was performed alongside a conditional power analysis in order to ensure patient safety and assess whether the treatment was likely to be beneficial for one or both groups. Following this analysis, a beneficial effect was observed for patients with an SR of 4 only, i.e., patients with moderate COVID-19 requiring supplementary oxygen. The study was continued for these COVID-19 patients. Overall, a total of 592 patients were enrolled and randomized between May 2020 and March 2021. Patients with a baseline SR of 4 were divided into two groups: 142 (49.8%) were assigned to the enisamium group and 143 (50.2%) to the placebo group. An analysis of the population showed that if patients were treated within 4 days of the onset of COVID-19 symptoms (n = 33), the median time to improvement was 8 days for the enisamium group and 13 days for the placebo group (p = 0.005). For patients treated within 10 days of the onset of COVID-19 symptoms (n = 154), the median time to improvement was 10 days for the enisamium group and 12 days for the placebo group (p = 0.002). Our findings suggest that enisamium is safe to use with COVID-19 patients, and that the observed clinical benefit of enisamium is worth reporting and studying in detail.
Collapse
Affiliation(s)
- Olga Holubovska
- Department of Infectious Diseases, O.O. Bogomolets National Medical University, T. Shevchenko Blvd. 13, 01601 Kyiv, Ukraine;
| | - Pavlo Babich
- State Expert Center, Smolenska Str. 10, 03057 Kyiv, Ukraine;
| | - Alla Mironenko
- Department of Respiratory and Other Viral Infections, L.V. Gromashevsky Institute of Epidemiology and Infectious Diseases of the NAMS of Ukraine, Amosova Str. 5a, 03083 Kyiv, Ukraine;
| | - Jens Milde
- Pharmalog Institut für Klinische Forschung GmbH, Oskar-Messter-Str. 29, 85737 Ismaning, Germany; (J.M.); (H.S.)
| | - Yuriy Lebed
- Pharmaxi LLC, Filatova Str. 10A, 01042 Kyiv, Ukraine;
| | - Holger Stammer
- Pharmalog Institut für Klinische Forschung GmbH, Oskar-Messter-Str. 29, 85737 Ismaning, Germany; (J.M.); (H.S.)
| | - Lutz Mueller
- Regenold GmbH, Zöllinplatz 4, 79410 Badenweiler, Germany;
| | - Aartjan J. W. te Velthuis
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Division of Virology, Department of Pathology, University of Cambridge Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK
| | - Victor Margitich
- Farmak Joint Stock Company, Kyrylivska Str., 04080 Kyiv, Ukraine
| | - Andrew Goy
- Farmak Joint Stock Company, Kyrylivska Str., 04080 Kyiv, Ukraine
| |
Collapse
|
16
|
Meis J, Pilz M, Bokelmann B, Herrmann C, Rauch G, Kieser M. Point estimation, confidence intervals, and P-values for optimal adaptive two-stage designs with normal endpoints. Stat Med 2024; 43:1577-1603. [PMID: 38339872 DOI: 10.1002/sim.10020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 09/25/2023] [Accepted: 12/18/2023] [Indexed: 02/12/2024]
Abstract
Due to the dependency structure in the sampling process, adaptive trial designs create challenges in point and interval estimation and in the calculation of P-values. Optimal adaptive designs, which are designs where the parameters governing the adaptivity are chosen to maximize some performance criterion, suffer from the same problem. Various analysis methods which are able to handle this dependency structure have already been developed. In this work, we aim to give a comprehensive summary of these methods and show how they can be applied to the class of designs with planned adaptivity, of which optimal adaptive designs are an important member. The defining feature of these kinds of designs is that the adaptive elements are completely prespecified. This allows for explicit descriptions of the calculations involved, which makes it possible to evaluate different methods in a fast and accurate manner. We will explain how to do so, and present an extensive comparison of the performance characteristics of various estimators between an optimal adaptive design and its group-sequential counterpart.
Collapse
Affiliation(s)
- Jan Meis
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Maximilian Pilz
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Björn Bokelmann
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carolin Herrmann
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Technische Universität Berlin, Berlin, Germany
| | - Meinhard Kieser
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Gao P, Zhang W. Adaptive sequential design for phase II single-arm oncology trials: an expansion of Simon's design. J Biopharm Stat 2024:1-15. [PMID: 38619921 DOI: 10.1080/10543406.2024.2341673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
Single-arm phase II trials are very common in oncology. A fixed sample trial may lack sufficient power if the true efficacy is less than the assumed one. Adaptive designs have been proposed in the literature. We propose a Simon's design based, adaptive sequential design. Simon's design is the most used fixed sample design for single-arm phase II oncology trials. A prominent feature of Simon's design is that it minimizes the sample size when there is no clinically meaningful efficacy. We identify Simon's trial as a special group sequential design. Established methods for sample size re-estimation (SSR) can be readily applied to Simon's design. Simulations show that simply adding SSR to Simon's design may still not provide desirable power. We propose some expansions to Simon's design. The expanded design with SSR can provide even more power.
Collapse
Affiliation(s)
- Ping Gao
- Biostatistics, Innovatio Statistics, Inc, Bridgewater, New Jersey, USA
| | - Weidong Zhang
- Biostatistics, Sana Biotechnology, Inc. Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Gass P, Thiel FC, Häberle L, Ackermann S, Theuser AK, Hummel N, Boehm S, Kimmig R, Reinthaller A, Becker S, Hilpert F, Janni W, Vergote I, Harter P, Emons J, Hein A, Beckmann MW, Fasching PA, Pöschke P. Primary results of the AGO-Zervix-1 Study: A prospective, randomized phase III study to compare the effects of paclitaxel and topotecan with those of cisplatin and topotecan in the treatment of patients with recurrent and persistent cervical cancer. Gynecol Oncol 2024; 183:25-32. [PMID: 38490057 DOI: 10.1016/j.ygyno.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Before the era of immunotherapies and antibody-drug conjugates, there were limited chemotherapeutic options for patients with recurrent and metastatic cervical cancer. Combination therapies with cisplatin have shown some superiority over monotherapy. This study examined platinum-free treatment regimens, comparing a combination of topotecan and paclitaxel (TP) with topotecan and cisplatin (TC) in patients with recurrent or metastatic cervical cancer, with or without prior platinum-based treatment. METHODS The AGO-Zervix-1 Study (NCT01405235) is a prospective, randomized phase III study in which patients were randomly assigned at a 1:1 ratio to treatment within the control arm with topotecan (0.75 mg/m2) on days 1-3 and cisplatin (50 mg/m2) on day 1 every 3 weeks and in the study arm topotecan (1.75 mg/m2) and paclitaxel (70 mg/m2) on days 1, 8, and 15 every 4 weeks or treatment. The primary study aim was overall survival; progression-free survival, toxicity, and quality of life were secondary aims. The interim and final analysis is here reported after recruitment of 173 of 312 planned patients. RESULTS Median overall survival in the TP arm was 9.6 months, compared with 12.0 months in the TC arm (log-rank test, P = 0.33). Median progression-free survival rates were 4.4 months with TP and 4.2 months with TC (log-rank test, P = 0.47). Leukopenia and nausea/vomiting were more frequent in the cisplatin-containing arm. Otherwise, toxicity profiles were comparable. There were no differences in FACT-G-assessed quality of life. CONCLUSION Platinum-based combination chemotherapy remains the standard of care chemotherapy regimen for patients with recurrent or metastatic cervical cancer.
Collapse
Affiliation(s)
- Paul Gass
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany.
| | - Falk C Thiel
- Department of Gynecology and Obstetrics, Alb Fils Clinics, Klinik am Eichert, Göppingen, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany; Biostatistics Unit, Department of Gynecology and Obstetrics, Erlangen University Hospital, Erlangen, Germany
| | - Sven Ackermann
- Department of Gynecology and Obstetrics, Klinikum Darmstadt, Darmstadt, Germany
| | | | - Nadine Hummel
- Institut für Frauengesundheit GmbH, Erlangen, Germany
| | - Sibylle Boehm
- Institut für Frauengesundheit GmbH, Erlangen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, Essen University Hospital, Essen, Germany
| | - Alexander Reinthaller
- Department of Gynecology and Gynecologic Oncology, AKH Vienna University Hospital, Vienna, Austria
| | - Sven Becker
- Department of Gynecology and Obstetrics, Frankfurt University Hospital, Frankfurt am Main, Germany
| | - Felix Hilpert
- Onkologisches Therapiezentrum, Krankenhaus Jerusalem, Hamburg, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Ignace Vergote
- Department of Gynaecology and Obstetrics, Division of Gynaecological Oncology, Leuven University Hospitals, Leuven, Belgium
| | - Phlipp Harter
- Department of Gynecology & Gynecologic Oncology, Ev. Kliniken Essen-Mitte, Essen, Germany
| | - Julius Emons
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexander Hein
- Department of Gynaecology and Obstetrics, Klinikum Esslingen, Esslingen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Patrik Pöschke
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
19
|
Lu N, Chen WC, Li H, Song C, Tiwari R, Wang C, Xu Y, Yue LQ. Propensity score-incorporated adaptive design approaches when incorporating real-world data. Pharm Stat 2024; 23:204-218. [PMID: 38014753 DOI: 10.1002/pst.2347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 09/07/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
The propensity score-integrated composite likelihood (PSCL) method is one method that can be utilized to design and analyze an application when real-world data (RWD) are leveraged to augment a prospectively designed clinical study. In the PSCL, strata are formed based on propensity scores (PS) such that similar subjects in terms of the baseline covariates from both the current study and RWD sources are placed in the same stratum, and then composite likelihood method is applied to down-weight the information from the RWD. While PSCL was originally proposed for a fixed design, it can be extended to be applied under an adaptive design framework with the purpose to either potentially claim an early success or to re-estimate the sample size. In this paper, a general strategy is proposed due to the feature of PSCL. For the possibility of claiming early success, Fisher's combination test is utilized. When the purpose is to re-estimate the sample size, the proposed procedure is based on the test proposed by Cui, Hung, and Wang. The implementation of these two procedures is demonstrated via an example.
Collapse
Affiliation(s)
- Nelson Lu
- Division of Biostatistics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Wei-Chen Chen
- Division of Biostatistics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Heng Li
- Division of Biostatistics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Changhong Song
- Division of Biostatistics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ram Tiwari
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Lawrence Township, New Jersey, USA
| | - Chenguang Wang
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yunling Xu
- Division of Biostatistics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lilly Q Yue
- Division of Biostatistics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
20
|
Zang Y, Thall PF, Yuan Y. A generalized phase 1-2-3 design integrating dose optimization with confirmatory treatment comparison. Biometrics 2024; 80:ujad022. [PMID: 38364811 PMCID: PMC10873567 DOI: 10.1093/biomtc/ujad022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/10/2023] [Accepted: 12/06/2023] [Indexed: 02/18/2024]
Abstract
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.
Collapse
Affiliation(s)
- Yong Zang
- Department of Biostatistics and Health Data Science; Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, IN 46202, United States
| | - Peter F Thall
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
21
|
Wang R, Mehta C. Power Considerations in Designing and Interpreting Adaptive Clinical Trials. NEJM EVIDENCE 2024; 3:EVIDe2300309. [PMID: 38320521 DOI: 10.1056/evide2300309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Adaptive clinical trials allow researchers to make preplanned modifications based on accumulating data from an ongoing trial while preserving the trial's integrity and validity. These modifications may include early termination in cases of successes or lack of efficacy, refining the sample size, altering treatments or doses, or focusing recruitment efforts on individuals most likely to benefit. In this issue of NEJM Evidence, Geisler et al.1 report results from the Apixaban for Treatment of Embolic Stroke of Undetermined Source (ATTICUS) trial, a multicenter randomized trial of apixaban compared with aspirin in patients with cardioembolism risk factors.
Collapse
Affiliation(s)
- Rui Wang
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston
| | - Cyrus Mehta
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston
- Cytel Corporation, Cambridge, MA
| |
Collapse
|
22
|
Geisler T, Keller T, Martus P, Poli K, Serna-Higuita LM, Schreieck J, Gawaz M, Tünnerhoff J, Bombach P, Nägele T, Klose U, Aidery P, Groga-Bada P, Kraft A, Hoffmann F, Hobohm C, Naupold K, Niehaus L, Wolf M, Bäzner H, Liman J, Wachter R, Kimmig H, Jung W, Huber R, Feurer R, Lindner A, Althaus K, Bode FJ, Petzold GC, Nguyen TN, Mac Grory B, Schrag M, Purrucker JC, Zuern CS, Ziemann U, Poli S. Apixaban versus Aspirin for Embolic Stroke of Undetermined Source. NEJM EVIDENCE 2024; 3:EVIDoa2300235. [PMID: 38320511 DOI: 10.1056/evidoa2300235] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
BACKGROUND: Rivaroxaban and dabigatran were not superior to aspirin in trials of patients with embolic stroke of undetermined source (ESUS). It is unknown whether apixaban is superior to aspirin in patients with ESUS and known risk factors for cardioembolism. METHODS: We conducted a multicenter, randomized, open-label, blinded-outcome trial of apixaban (5 mg twice daily) compared with aspirin (100 mg once daily) initiated within 28 days after ESUS in patients with at least one predictive factor for atrial fibrillation or a patent foramen ovale. Cardiac monitoring was mandatory, and aspirin treatment was switched to apixaban in case of atrial fibrillation detection. The primary outcome was any new ischemic lesion on brain magnetic resonance imaging (MRI) during 12-month follow-up. Secondary outcomes included major and clinically relevant nonmajor bleeding. RESULTS: A total of 352 patients were randomly assigned to receive apixaban (178 patients) or aspirin (174 patients) at a median of 8 days after ESUS. At 12-month follow-up, MRI follow-up was available in 325 participants (92.3%). New ischemic lesions occurred in 23 of 169 (13.6%) participants in the apixaban group and in 25 of 156 (16.0%) participants in the aspirin group (adjusted odds ratio, 0.79; 95% confidence interval, 0.42 to 1.48; P=0.57). Major and clinically relevant nonmajor bleeding occurred in five and seven participants, respectively (1-year cumulative incidences, 2.9 and 4.2; hazard ratio, 0.68; 95% confidence interval, 0.22 to 2.16). Serious adverse event rates were 43.9 per 100 person-years in those given apixaban and 45.7 per 100 person-years in those given aspirin. The Apixaban for the Treatment of Embolic Stroke of Undetermined Source trial was terminated after a prespecified interim analysis as a result of futility. CONCLUSIONS: Apixaban treatment was not superior to cardiac monitoring-guided aspirin in preventing new ischemic lesions in an enriched ESUS population. (Funded by Bristol-Myers Squibb and Medtronic Europe; ClinicalTrials.gov number, NCT02427126.)
Collapse
Affiliation(s)
- Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Timea Keller
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biometry, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Khouloud Poli
- Department of Neurology & Stroke, Eberhard Karls University Tübingen, Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Lina Maria Serna-Higuita
- Institute for Clinical Epidemiology and Applied Biometry, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Juergen Schreieck
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Johannes Tünnerhoff
- Department of Neurology & Stroke, Eberhard Karls University Tübingen, Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Paula Bombach
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Neurology and Interdisciplinary Neuro-Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Thomas Nägele
- Department of Diagnostic and Interventional Neuroradiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Uwe Klose
- Department of Diagnostic and Interventional Neuroradiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Parwez Aidery
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Patrick Groga-Bada
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andrea Kraft
- Department of Neurology, Krankenhaus Martha-Maria Halle-Doelau, Halle (Saale), Germany
| | - Frank Hoffmann
- Department of Neurology, Krankenhaus Martha-Maria Halle-Doelau, Halle (Saale), Germany
| | - Carsten Hobohm
- Department of Neurology, Carl-von-Basedow Klinikum Merseburg, Merseburg, Germany
| | - Katrin Naupold
- Department of Neurology, Carl-von-Basedow Klinikum Merseburg, Merseburg, Germany
| | - Ludwig Niehaus
- Department of Neurology, Rems-Murr Kliniken, Winnenden, Germany
| | - Marc Wolf
- Department of Neurology, Klinikum Stuttgart, Stuttgart, Germany
| | - Hansjörg Bäzner
- Department of Neurology, Klinikum Stuttgart, Stuttgart, Germany
| | - Jan Liman
- Department of Neurology, Klinikum Nürnberg, Nürnberg, Germany
- Clinic for Neurology, University Hospital Göttingen, Göttingen, Germany
| | - Rolf Wachter
- Department of Cardiology, University Hospital Leipzig, Leipzig, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research, Göttingen, Germany
| | - Hubert Kimmig
- Department of Neurology, Schwarzwald-Baar Klinikum, Villingen-Schwenningen, Germany
| | - Werner Jung
- Department of Cardiology, Schwarzwald-Baar Klinikum, Villingen-Schwenningen, Germany
| | - Roman Huber
- Department of Neurology, Klinikum Friedrichshafen, Friedrichshafen, Germany
| | - Regina Feurer
- Department of Neurology, Klinikum Friedrichshafen, Friedrichshafen, Germany
| | - Alfred Lindner
- Department of Neurology, Marienhospital Stuttgart, Stuttgart, Germany
| | | | - Felix J Bode
- Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Gabor C Petzold
- Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Thanh N Nguyen
- Department of Radiology, Boston Medical Center, Boston
- Department of Neurology, Boston Medical Center, Boston
| | - Brian Mac Grory
- Duke Clinical Research Institute, Durham, NC
- Department of Neurology, Duke University School of Medicine, Durham, NC
| | - Matthew Schrag
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN
| | - Jan C Purrucker
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christine S Zuern
- Department of Neurology, Klinikum Nürnberg, Nürnberg, Germany
- Department of Cardiology, Universitätsspital Basel, Basel, Switzerland
| | - Ulf Ziemann
- Department of Neurology & Stroke, Eberhard Karls University Tübingen, Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sven Poli
- Department of Neurology & Stroke, Eberhard Karls University Tübingen, Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
23
|
Poli S, Mbroh J, Baron JC, Singhal AB, Strbian D, Molina C, Lemmens R, Turc G, Mikulik R, Michel P, Tatlisumak T, Audebert HJ, Dichgans M, Veltkamp R, Hüsing J, Graessner H, Fiehler J, Montaner J, Adeyemi AK, Althaus K, Arenillas JF, Bender B, Benedikt F, Broocks G, Burghaus I, Cardona P, Deb-Chatterji M, Cviková M, Defreyne L, De Herdt V, Detante O, Ernemann U, Flottmann F, García Guillamón L, Glauch M, Gomez-Exposito A, Gory B, Sylvie Grand S, Haršány M, Hauser TK, Heck O, Hemelsoet D, Hennersdorf F, Hoppe J, Kalmbach P, Kellert L, Köhrmann M, Kowarik M, Lara-Rodríguez B, Legris L, Lindig T, Luntz S, Lusk J, Mac Grory B, Manger A, Martinez-Majander N, Mengel A, Meyne J, Müller S, Mundiyanapurath S, Naggara O, Nedeltchev K, Nguyen TN, Nilsson MA, Obadia M, Poli K, Purrucker JC, Räty S, Richard S, Richter H, Schilte C, Schlemm E, Stöhr L, Stolte B, Sykora M, Thomalla G, Tomppo L, van Horn N, Zeller J, Ziemann U, Zuern CS, Härtig F, Tuennerhoff J. Penumbral Rescue by normobaric O = O administration in patients with ischemic stroke and target mismatch proFile (PROOF): Study protocol of a phase IIb trial. Int J Stroke 2024; 19:120-126. [PMID: 37515459 PMCID: PMC10759237 DOI: 10.1177/17474930231185275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/12/2023] [Indexed: 07/30/2023]
Abstract
RATIONALE Oxygen is essential for cellular energy metabolism. Neurons are particularly vulnerable to hypoxia. Increasing oxygen supply shortly after stroke onset could preserve the ischemic penumbra until revascularization occurs. AIMS PROOF investigates the use of normobaric oxygen (NBO) therapy within 6 h of symptom onset/notice for brain-protective bridging until endovascular revascularization of acute intracranial anterior-circulation occlusion. METHODS AND DESIGN Randomized (1:1), standard treatment-controlled, open-label, blinded endpoint, multicenter adaptive phase IIb trial. STUDY OUTCOMES Primary outcome is ischemic core growth (mL) from baseline to 24 h (intention-to-treat analysis). Secondary efficacy outcomes include change in NIHSS from baseline to 24 h, mRS at 90 days, cognitive and emotional function, and quality of life. Safety outcomes include mortality, intracranial hemorrhage, and respiratory failure. Exploratory analyses of imaging and blood biomarkers will be conducted. SAMPLE SIZE Using an adaptive design with interim analysis at 80 patients per arm, up to 456 participants (228 per arm) would be needed for 80% power (one-sided alpha 0.05) to detect a mean reduction of ischemic core growth by 6.68 mL, assuming 21.4 mL standard deviation. DISCUSSION By enrolling endovascular thrombectomy candidates in an early time window, the trial replicates insights from preclinical studies in which NBO showed beneficial effects, namely early initiation of near 100% inspired oxygen during short temporary ischemia. Primary outcome assessment at 24 h on follow-up imaging reduces variability due to withdrawal of care and early clinical confounders such as delayed extubation and aspiration pneumonia. TRIAL REGISTRATIONS ClinicalTrials.gov: NCT03500939; EudraCT: 2017-001355-31.
Collapse
Affiliation(s)
- Sven Poli
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard-Karls University, Tubingen, Germany
| | - Joshua Mbroh
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
| | - Jean-Claude Baron
- Department of Neurology, Hopital Sainte-Anne, Universite de Paris, Paris, France
| | - Aneesh B Singhal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Strbian
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Carlos Molina
- Department of Neurology, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Robin Lemmens
- Department of Neurosciences, Experimental Neurology, KU Leuven, University of Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Guillaume Turc
- Department of Neurology, Hopital Sainte-Anne, Universite de Paris, Paris, France
- Department of Neurology, GHU Paris Psychiatrie et Neurosciences INSERM U1266 Universite Paris Cite FHU NeuroVasc, Paris, France
| | - Robert Mikulik
- Department of Neurology, St. Anne’s University Hospital Brno and Masaryk University, Brno, Czech Republic
| | - Patrik Michel
- Neurosciences Cliniques, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Turgut Tatlisumak
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Heinrich J Audebert
- Department of Neurology and Center for Stroke Research Berlin, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
- German Centre for Cardiovascular Research (DZHK, Munich), Munich, Germany
| | - Roland Veltkamp
- Department of Neurology, Alfried Krupp Hospital, Essen, Germany
- Department of Brain Sciences, Imperial College London, London, UK
| | - Johannes Hüsing
- Coordinating Centre for Clinical Trials, University of Heidelberg, Heidelberg, Germany
- Landeskrebsregister Nordrhein-Westfalen, Bochum, Germany
| | - Holm Graessner
- Center for Rare Diseases, Eberhard-Karls University, Tubingen, Germany
| | - Jens Fiehler
- Neuroradiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Eppdata GmbH, Hamburg, Germany
| | - Joan Montaner
- Vall d’Hebron Institut de Recerca, Neurovascular Research Lab, Barcelona, Spain
| | | | | | | | - Benjamin Bender
- Department of Diagnostic and Interventional Neuroradiology, Eberhard-Karls University, Tubingen, Germany
| | - Frank Benedikt
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Gabriel Broocks
- Department of Neuroradiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ina Burghaus
- Coordinating Centre for Clinical Trials, University of Heidelberg, Heidelberg, Germany
| | - Pere Cardona
- Department of Neurology, Hospital University de Bellvitge, Barcelona, Spain
| | - Milani Deb-Chatterji
- Department of Neurology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Cviková
- Department of Neurology, St. Anne’s University Hospital in Brno, Faculty of Medicine Masaryk University, Brno, Czech Republic
| | - Luc Defreyne
- Department of Vascular and Interventional Radiology, Ghent University Hospital, Ghent, Belgium
| | - Veerle De Herdt
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Olivier Detante
- Neurology, CHU Grenoble Alpes, Grenoble, France
- Inserm, U1216, Grenoble Institut Neurosciences, Université Grenoble Alpes, Grenoble, France
| | - Ulrike Ernemann
- Department of Diagnostic and Interventional Neuroradiology, Eberhard-Karls University, Tubingen, Germany
| | - Fabian Flottmann
- Department of Neuroradiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Monika Glauch
- Center for Rare Diseases, Eberhard-Karls University, Tubingen, Germany
| | - Alexandra Gomez-Exposito
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
| | - Benjamin Gory
- Department of Diagnostic and Therapeutic Neuroradiology, Centre Hospital Regional Universitaire de Nancy, Universite de Lorraine, INSERM U1254, Nancy, France
| | - Sylvie Sylvie Grand
- Inserm, U1216, Grenoble Institut Neurosciences, Université Grenoble Alpes, Grenoble, France
- Neuroradiology / MRI Department, CHU Grenoble Alpes, Grenoble, France
| | - Michal Haršány
- Department of Neurology, St. Anne’s University Hospital in Brno, Faculty of Medicine Masaryk University, Brno, Czech Republic
- International Clinical Research Centre, St. Anne’s University Hospital in Brno, Brno, Czech Republic
| | - Till Karsten Hauser
- Department of Diagnostic and Interventional Neuroradiology, Eberhard-Karls University, Tubingen, Germany
| | - Olivier Heck
- Neuroradiology / MRI Department, CHU Grenoble Alpes, Grenoble, France
| | | | - Florian Hennersdorf
- Department of Diagnostic and Interventional Neuroradiology, Eberhard-Karls University, Tubingen, Germany
| | - Julia Hoppe
- Department of Neurology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Pia Kalmbach
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
| | - Lars Kellert
- Department of Neurology, Ludwig Maximilian University (LMU), Munich, Germany
| | - Martin Köhrmann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Markus Kowarik
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard-Karls University, Tubingen, Germany
| | | | - Loic Legris
- Neurology, CHU Grenoble Alpes, Grenoble, France
- Inserm, U1216, Grenoble Institut Neurosciences, Université Grenoble Alpes, Grenoble, France
| | - Tobias Lindig
- Department of Diagnostic and Interventional Neuroradiology, Eberhard-Karls University, Tubingen, Germany
| | - Steffen Luntz
- Coordinating Centre for Clinical Trials, University of Heidelberg, Heidelberg, Germany
| | - Jay Lusk
- Duke University School of Medicine, Durham, NC, USA
| | - Brian Mac Grory
- Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Andreas Manger
- Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls University, Tubingen, Germany
| | | | - Annerose Mengel
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
| | - Johannes Meyne
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Susanne Müller
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | | | - Olivier Naggara
- Department of Neuroradiology, GHU Paris Psychiatrie et Neurosciences INSERM U1266 Universite Paris Cite FHU NeuroVasc, Paris, France
| | - Krassen Nedeltchev
- Department of Neurology, KSA Kantonsspital Aarau and University of Bern, Bern, Switzerland
| | - Thanh N Nguyen
- Department of Radiology, Boston Medical Center, Boston, MA, USA
- Department of Neurology, Boston Medical Center, Boston, MA, USA
| | - Maike A Nilsson
- Coordinating Centre for Clinical Trials, University of Heidelberg, Heidelberg, Germany
| | - Michael Obadia
- Department of Neurology and Stroke Center, Hopital fondation Adolphe de Rothschild, Paris, France
| | - Khouloud Poli
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
| | - Jan C Purrucker
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Silja Räty
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | - Hardy Richter
- Department of Infectiology, Eberhard-Karls-University, Tuebingen, Germany
| | - Clotilde Schilte
- Department of Anaesthesia and Critical Care, CHU Grenoble Alpes, Grenoble, France
| | - Eckhard Schlemm
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Stöhr
- European Clinical Research Infrastructure Network (ECRIN), Paris, France
| | - Benjamin Stolte
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Marek Sykora
- Department of Neurology, St. John’s Hospital, Vienna, Austria
| | - Götz Thomalla
- Department of Neurology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Liisa Tomppo
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Noel van Horn
- Department of Neuroradiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Zeller
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
| | - Ulf Ziemann
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard-Karls University, Tubingen, Germany
| | - Christine S Zuern
- Department of Cardiology, Universitatsspital Basel, Basel, Switzerland
| | - Florian Härtig
- Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls University, Tubingen, Germany
| | - Johannes Tuennerhoff
- Department of Neurology & Stroke, Eberhard-Karls University, University Hospital, Tubingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard-Karls University, Tubingen, Germany
| |
Collapse
|
24
|
Polychronidis G, Rahbari NN, Bruckner T, Sander A, Sommer F, Usta S, Hermann J, Albers MB, Sargut M, Knebel P, Klotz R. Continuous versus interrupted abdominal wall closure after emergency midline laparotomy: CONTINT: a randomized controlled trial [NCT00544583]. World J Emerg Surg 2023; 18:51. [PMID: 37848901 PMCID: PMC10583371 DOI: 10.1186/s13017-023-00517-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/23/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND High-level evidence regarding the technique of abdominal wall closure for patients undergoing emergency midline laparotomy is sparse. Therefore, we conducted a randomized controlled trial (RCT) to evaluate the efficacy and safety of two commonly applied abdominal wall closure strategies after primary emergency midline laparotomy. METHODS/DESIGN CONTINT was a multi-center pragmatic open-label exploratory randomized controlled parallel trial. Two different abdominal wall closure strategies in patients undergoing primary midline laparotomy for an emergency surgical intervention with a suspected septic focus in the abdominal cavity were compared: the continuous, all-layer suture and the interrupted suture technique. The primary composite endpoint was burst abdomen within 30 days after surgery or incisional hernia within 12 months. As reliable data on this composite primary endpoint were not available for patients undergoing emergency surgery, it was planned to initially recruit 80 patients and conduct an interim analysis after these had completed the 12 months follow-up. RESULTS From August 31, 2009, to June 28, 2012, 124 patients were randomized of whom 119 underwent surgery and were analyzed according to the intention-to-treat (ITT) principal. The primary composite endpoint did not differ between the continuous suture (C: 27.1%) and the interrupted suture group (I: 30.0%). None of the individual components of the primary endpoint (reoperation due to burst abdomen after 30 days (C: 13.5%, I: 15.1%) and reoperation due to incisional hernia (C: 3.0%, I:11.1%)) differed between groups. Time needed for fascial closure was longer in the interrupted suture group (C: 12.8 ± 4.5 min, I: 17.4 ± 6.1 min). BMI was associated with burst abdomen during the first 30 days with an OR of 1.17 (95% CI 1.04-1.32). CONCLUSION This RCT showed no difference between continuous suture with slowly absorbable suture versus interrupted rapidly absorbable sutures after primary emergency midline laparotomy in rates of postoperative burst abdomen and incisional hernia after one year. However, the trial was stopped after the interim analysis due to futility as there was no chance to show superiority of one suture technique.
Collapse
Affiliation(s)
- Georgios Polychronidis
- Department of General, Visceral and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Study Centre of the German Surgical Society (SDGC), Heidelberg, Germany
| | - Nuh N Rahbari
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Bruckner
- Institute of Medical Biometry (IMBI), University of Heidelberg, Heidelberg, Germany
| | - Anja Sander
- Institute of Medical Biometry (IMBI), University of Heidelberg, Heidelberg, Germany
| | - Florian Sommer
- Department of General and Visceral Surgery, Augsburg University Medical Center, Augsburg, Germany
| | - Selami Usta
- Department for General and Visceral Surgery, St. Josefs-Hospital, Dortmund, Germany
| | - Janssen Hermann
- Department of General, Visceral, Vascular and Thoracic Surgery, Düren Hospital, Düren, Germany
| | - Max Benjamin Albers
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Mine Sargut
- Department of Surgery, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Rosa Klotz
- Department of General, Visceral and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany.
- Study Centre of the German Surgical Society (SDGC), Heidelberg, Germany.
| |
Collapse
|
25
|
Zhu H, Wong WK. An Overview of Adaptive Designs and Some of Their Challenges, Benefits, and Innovative Applications. J Med Internet Res 2023; 25:e44171. [PMID: 37843888 PMCID: PMC10616728 DOI: 10.2196/44171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/25/2023] [Accepted: 05/16/2023] [Indexed: 10/17/2023] Open
Abstract
Adaptive designs are increasingly developed and used to improve all phases of clinical trials and in biomedical studies in various ways to address different statistical issues. We first present an overview of adaptive designs and note their numerous advantages over traditional clinical trials. In particular, we provide a concrete demonstration that shows how recent adaptive design strategies can further improve an adaptive trial implemented 13 years ago. Despite their usefulness, adaptive designs are still not widely implemented in clinical trials. We offer a few possible reasons and propose some ways to use them more broadly in practice, which include greater availability of software tools and interactive websites to generate optimal adaptive trials freely and effectively, including the use of metaheuristics to facilitate the search for an efficient trial design. To this end, we present several web-based tools for finding various adaptive and nonadaptive optimal designs and discuss nature-inspired metaheuristics. Metaheuristics are assumptions-free general purpose optimization algorithms widely used in computer science and engineering to tackle all kinds of challenging optimization problems, and their use in designing clinical trials is just emerging. We describe a few recent such applications and some of their capabilities for designing various complex trials. Particle swarm optimization is an exemplary nature-inspired algorithm, and similar to others, it has a simple definition but many moving parts, making it hard to study its properties analytically. We investigated one of its hitherto unstudied issues on how to bring back out-of-range candidates during the search for the optimum of the search domain and show that different strategies can impact the success and time of the search. We conclude with a few caveats on the use of metaheuristics for a successful search.
Collapse
Affiliation(s)
- Hongjian Zhu
- Statistical Innovation Group, AbbVie Inc., Virtual Office, Sugar Land, TX, United States
| | - Weng Kee Wong
- Department of Biostatistics, Fielding School of Public Health, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
26
|
Deforth M, Micheloud C, Roes KC, Held L. Combining evidence from clinical trials in conditional or accelerated approval. Pharm Stat 2023. [PMID: 37114714 DOI: 10.1002/pst.2302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/24/2023] [Indexed: 04/29/2023]
Abstract
Conditional (European Medicines Agency) or accelerated (U.S. Food and Drug Administration) approval of drugs allows earlier access to promising new treatments that address unmet medical needs. Certain post-marketing requirements must typically be met in order to obtain full approval, such as conducting a new post-market clinical trial. We study the applicability of the recently developed harmonic mean χ 2 $$ {\chi}^2 $$ -test to this conditional or accelerated approval framework. The proposed approach can be used both to support the design of the post-market trial and the analysis of the combined evidence provided by both trials. Other methods considered are the two-trials rule, Fisher's criterion and Stouffer's method. In contrast to some of the traditional methods, the harmonic mean χ 2 $$ {\chi}^2 $$ -test always requires a post-market clinical trial. If the p $$ p $$ -value from the pre-market clinical trial is ≪ 0.025 $$ \ll 0.025 $$ , a smaller sample size for the post-market clinical trial is needed than with the two-trials rule. For illustration, we apply the harmonic mean χ 2 $$ {\chi}^2 $$ -test to a drug which received conditional (and later full) market licensing by the EMA. A simulation study is conducted to study the operating characteristics of the harmonic mean χ 2 $$ {\chi}^2 $$ -test and two-trials rule in more detail. We finally investigate the applicability of these two methods to compute the power at interim of an ongoing post-market trial. These results are expected to aid in the design and assessment of the required post-market studies in terms of the level of evidence required for full approval.
Collapse
Affiliation(s)
- Manja Deforth
- Department of Biostatistics at the Epidemiology, Biostatistics and Prevention Institute (EBPI) and Center for Reproducible Science (CRS), University of Zurich, Zurich, Switzerland
| | - Charlotte Micheloud
- Department of Biostatistics at the Epidemiology, Biostatistics and Prevention Institute (EBPI) and Center for Reproducible Science (CRS), University of Zurich, Zurich, Switzerland
| | - Kit C Roes
- Department of Health Evidence, Section Biostatistics, Radboud University Medical Center, Radboud University, Nijmegen, The Netherlands
| | - Leonhard Held
- Department of Biostatistics at the Epidemiology, Biostatistics and Prevention Institute (EBPI) and Center for Reproducible Science (CRS), University of Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Baldi Antognini A, Frieri R, Zagoraiou M. New insights into adaptive enrichment designs. Stat Pap (Berl) 2023. [DOI: 10.1007/s00362-023-01433-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
AbstractThe transition towards personalized medicine is happening and the new experimental framework is raising several challenges, from a clinical, ethical, logistical, regulatory, and statistical perspective. To face these challenges, innovative study designs with increasing complexity have been proposed. In particular, adaptive enrichment designs are becoming more attractive for their flexibility. However, these procedures rely on an increasing number of parameters that are unknown at the planning stage of the clinical trial, so the study design requires particular care. This review is dedicated to adaptive enrichment studies with a focus on design aspects. While many papers deal with methods for the analysis, the sample size determination and the optimal allocation problem have been overlooked. We discuss the multiple aspects involved in adaptive enrichment designs that contribute to their advantages and disadvantages. The decision-making process of whether or not it is worth enriching should be driven by clinical and ethical considerations as well as scientific and statistical concerns.
Collapse
|
28
|
Stallard N. Adaptive enrichment designs with a continuous biomarker. Biometrics 2023; 79:9-19. [PMID: 35174875 DOI: 10.1111/biom.13644] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/23/2021] [Indexed: 12/01/2022]
Abstract
A popular design for clinical trials assessing targeted therapies is the two-stage adaptive enrichment design with recruitment in stage 2 limited to a biomarker-defined subgroup chosen based on data from stage 1. The data-dependent selection leads to statistical challenges if data from both stages are used to draw inference on treatment effects in the selected subgroup. If subgroups considered are nested, as when defined by a continuous biomarker, treatment effect estimates in different subgroups follow the same distribution as estimates in a group-sequential trial. This result is used to obtain tests controlling the familywise type I error rate (FWER) for six simple subgroup selection rules, one of which also controls the FWER for any selection rule. Two approaches are proposed: one based on multivariate normal distributions suitable if the number of possible subgroups, k, is small, and one based on Brownian motion approximations suitable for large k. The methods, applicable in the wide range of settings with asymptotically normal test statistics, are illustrated using survival data from a breast cancer trial.
Collapse
Affiliation(s)
- Nigel Stallard
- Statistics and Epidemiology, Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
29
|
Flournoy N, Tarima S. Discussion on "Adaptive enrichment designs with a continuous biomarker" by Nigel Stallard. Biometrics 2023; 79:31-35. [PMID: 35290671 DOI: 10.1111/biom.13641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/23/2021] [Indexed: 11/27/2022]
Abstract
We congratulate Dr. Nigel Stallard on his stimulating paper on adaptive enrichment designs with a continuous biomarker. Dr. Stallard details a framework for a large and interesting class of enrichment procedures. His work has motivated us to offer some thoughts in response. Dr. Stallard's strategy is to use the maximum of a test statistic over a set of possible threshold values to define the enriched population to be sampled in a second stage. This reminds us of procedures for identifying a change point, a biomarker value beyond which the effect of treatment is increased. For simplicity we focus our comments on Dr. Stallard's Rule 1 for selecting the second-stage sampling threshold. Using this rule, we present the likelihood ratio approach for adaptive testing and compare it to Dr. Stallard's approach for a few scenarios.
Collapse
Affiliation(s)
- Nancy Flournoy
- Department of Statistics, University of Missouri, Columbia, Missouri
| | - Sergey Tarima
- Institute for Health and Equity, Medical College of Wisconsin, Wauwatosa, Wisconsin
| |
Collapse
|
30
|
Phillips RV, van der Laan MJ. Discussion on "Adaptive enrichment designs with a continuous biomarker" by Nigel Stallard. Biometrics 2023; 79:20-22. [PMID: 35332936 DOI: 10.1111/biom.13640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Rachael V Phillips
- Division of Biostatistics, University of California at Berkeley, Berkeley, California, USA
| | - Mark J van der Laan
- Division of Biostatistics, University of California at Berkeley, Berkeley, California, USA
| |
Collapse
|
31
|
Wason JMS. Discussion on "Adaptive enrichment designs with a continuous biomarker" by Nigel Stallard. Biometrics 2023; 79:23-25. [PMID: 35266548 DOI: 10.1111/biom.13643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 11/28/2022]
Affiliation(s)
- James M S Wason
- Biostatistics Research Group, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
32
|
Jennison C. Discussion on "Adaptive enrichment designs with a continuous biomarker" by N. Stallard. Biometrics 2023; 79:26-30. [PMID: 35344206 DOI: 10.1111/biom.13642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022]
|
33
|
Xu H, Liu Y, Beckman RA. Adaptive Endpoints Selection with Application in Rare Disease. Stat Biopharm Res 2023. [DOI: 10.1080/19466315.2023.2183252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Heng Xu
- Nektar Therapeutics, San Francisco, USA
| | - Yi Liu
- Nektar Therapeutics, San Francisco, USA
| | - Robert A. Beckman
- Departments of Oncology and of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center
| |
Collapse
|
34
|
Labenz J, Anschütz M, Walstab J, Wedemeyer RS, Wolters H, Schug B. Heartburn relief with bicarbonate-rich mineral water: results of the randomised, placebo-controlled phase-III trial STOMACH STILL. BMJ Open Gastroenterol 2023; 10:bmjgast-2022-001048. [PMID: 36849190 PMCID: PMC9972411 DOI: 10.1136/bmjgast-2022-001048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/09/2023] [Indexed: 03/01/2023] Open
Abstract
OBJECTIVE We assessed whether the bicarbonate-rich mineral water Staatl. Fachingen STILL is superior over conventional mineral water in relieving heartburn. DESIGN Multicentre, double-blind, randomised, placebo-controlled trial STOMACH STILL in adult patients with frequent heartburn episodes since ≥6 months and without moderate/severe reflux oesophagitis. Patients drank 1.5 L/day verum or placebo over the course of the day for 6 weeks. Primary endpoint was the percentage of patients with reduction of ≥5 points in the Reflux Disease Questionnaire (RDQ) score for 'heartburn'. Secondary endpoints included symptom reduction (RDQ), health-related quality of life (HRQOL, Quality of Life in Reflux and Dyspepsia (QOLRAD)), intake of rescue medication and safety/tolerability. RESULTS Of 148 randomised patients (verum: n=73, placebo: n=75), 143 completed the trial. Responder rates were 84.72% in the verum and 63.51% in the placebo group (p=0.0035, number needed to treat=5). Symptoms improved under verum compared with placebo for the dimension 'heartburn' (p=0.0003) and the RDQ total score (p=0.0050). HRQOL improvements under verum compared with placebo were reported for 3 of 5 QOLRAD domains, that is, 'food/drink problems' (p=0.0125), 'emotional distress' (p=0.0147) and 'vitality' (p=0.0393). Mean intake of rescue medication decreased from 0.73 tablets/day at baseline to 0.47 tablets/day in week 6 in the verum group, whereas in the placebo group it remained constant during the trial. Only three patients had treatment-related adverse events (verum: n=1, placebo: n=2). CONCLUSION STOMACH STILL is the first controlled clinical trial demonstrating superiority of a mineral water over placebo in relieving heartburn, accompanied by an improved HRQOL. TRIAL REGISTRATION NUMBER EudraCT 2017-001100-30.
Collapse
Affiliation(s)
| | - Maria Anschütz
- SocraTec R&D Concepts in Drug Research and Development GmbH, Oberursel, Germany
| | - Jutta Walstab
- SocraTec R&D Concepts in Drug Research and Development GmbH, Erfurt, Germany
| | - Ralph-Steven Wedemeyer
- SocraTec R&D Concepts in Drug Research and Development GmbH, Oberursel, Germany.,SocraMetrics GmbH, Erfurt, Germany
| | - Heiner Wolters
- Fachingen Heil- und Mineralbrunnen GmbH, Birlenbach OT Fachingen/Lahn, Germany
| | - Barbara Schug
- SocraTec R&D Concepts in Drug Research and Development GmbH, Oberursel, Germany.,SocraMetrics GmbH, Erfurt, Germany
| |
Collapse
|
35
|
Placzek M, Friede T. Blinded sample size recalculation in adaptive enrichment designs. Biom J 2023; 65:e2000345. [PMID: 35983952 DOI: 10.1002/bimj.202000345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 09/24/2021] [Accepted: 11/07/2021] [Indexed: 12/17/2022]
Abstract
In the precision medicine era, (prespecified) subgroup analyses are an integral part of clinical trials. Incorporating multiple populations and hypotheses in the design and analysis plan, adaptive designs promise flexibility and efficiency in such trials. Adaptations include (unblinded) interim analyses (IAs) or blinded sample size reviews. An IA offers the possibility to select promising subgroups and reallocate sample size in further stages. Trials with these features are known as adaptive enrichment designs. Such complex designs comprise many nuisance parameters, such as prevalences of the subgroups and variances of the outcomes in the subgroups. Additionally, a number of design options including the timepoint of the sample size review and timepoint of the IA have to be selected. Here, for normally distributed endpoints, we propose a strategy combining blinded sample size recalculation and adaptive enrichment at an IA, that is, at an early timepoint nuisance parameters are reestimated and the sample size is adjusted while subgroup selection and enrichment is performed later. We discuss implications of different scenarios concerning the variances as well as the timepoints of blinded review and IA and investigate the design characteristics in simulations. The proposed method maintains the desired power if planning assumptions were inaccurate and reduces the sample size and variability of the final sample size when an enrichment is performed. Having two separate timepoints for blinded sample size review and IA improves the timing of the latter and increases the probability to correctly enrich a subgroup.
Collapse
Affiliation(s)
- Marius Placzek
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| |
Collapse
|
36
|
Robertson DS, Choodari‐Oskooei B, Dimairo M, Flight L, Pallmann P, Jaki T. Point estimation for adaptive trial designs I: A methodological review. Stat Med 2023; 42:122-145. [PMID: 36451173 PMCID: PMC7613995 DOI: 10.1002/sim.9605] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/21/2022] [Accepted: 11/01/2022] [Indexed: 12/02/2022]
Abstract
Recent FDA guidance on adaptive clinical trial designs defines bias as "a systematic tendency for the estimate of treatment effect to deviate from its true value," and states that it is desirable to obtain and report estimates of treatment effects that reduce or remove this bias. The conventional end-of-trial point estimates of the treatment effects are prone to bias in many adaptive designs, because they do not take into account the potential and realized trial adaptations. While much of the methodological developments on adaptive designs have tended to focus on control of type I error rates and power considerations, in contrast the question of biased estimation has received relatively less attention. This article is the first in a two-part series that studies the issue of potential bias in point estimation for adaptive trials. Part I provides a comprehensive review of the methods to remove or reduce the potential bias in point estimation of treatment effects for adaptive designs, while part II illustrates how to implement these in practice and proposes a set of guidelines for trial statisticians. The methods reviewed in this article can be broadly classified into unbiased and bias-reduced estimation, and we also provide a classification of estimators by the type of adaptive design. We compare the proposed methods, highlight available software and code, and discuss potential methodological gaps in the literature.
Collapse
Affiliation(s)
| | | | - Munya Dimairo
- School of Health and Related Research (ScHARR)University of SheffieldSheffieldUK
| | - Laura Flight
- School of Health and Related Research (ScHARR)University of SheffieldSheffieldUK
| | | | - Thomas Jaki
- MRC Biostatistics UnitUniversity of CambridgeCambridgeUK
- Faculty of Informatics and Data ScienceUniversity of RegensburgRegensburgGermany
| |
Collapse
|
37
|
Ko FS. The Simon’s two-stage design accounting for genetic heterogeneity. COMMUN STAT-THEOR M 2022. [DOI: 10.1080/03610926.2022.2148469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Feng-shou Ko
- KF Statistical Consulting Company, Kaohsiung, Taiwan R.O.C
| |
Collapse
|
38
|
Abstract
Covariate adjustment via a regression approach is known to increase the precision of statistical inference when fixed trial designs are employed in randomized controlled studies. When an adaptive multi-arm design is employed with the ability to select treatments, it is unclear how covariate adjustment affects various aspects of the study. Consider the design framework that relies on pre-specified treatment selection rule(s) and a combination test approach for hypothesis testing. It is our primary goal to evaluate the impact of covariate adjustment on adaptive multi-arm designs with treatment selection. Our secondary goal is to show how the Uniformly Minimum Variance Conditionally Unbiased Estimator can be extended to account for covariate adjustment analytically. We find that adjustment with different sets of covariates can lead to different treatment selection outcomes and hence probabilities of rejecting hypotheses. Nevertheless, we do not see any negative impact on the control of the familywise error rate when covariates are included in the analysis model. When adjusting for covariates that are moderately or highly correlated with the outcome, we see various benefits to the analysis of the design. Conversely, there is negligible impact when including covariates that are uncorrelated with the outcome. Overall, pre-specification of covariate adjustment is recommended for the analysis of adaptive multi-arm design with treatment selection. Having the statistical analysis plan in place prior to the interim and final analyses is crucial, especially when a non-collapsible measure of treatment effect is considered in the trial.
Collapse
Affiliation(s)
- Kim May Lee
- Institute of Psychiatry, Psychology and Neuroscience, King’s College
London, London, UK
| | | | - Thomas Jaki
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
39
|
Wolfrum S, Roedl K, Hanebutte A, Pfeifer R, Kurowski V, Riessen R, Daubmann A, Braune S, Söffker G, Bibiza-Freiwald E, Wegscheider K, Schunkert H, Thiele H, Kluge S. Temperature Control After In-Hospital Cardiac Arrest: A Randomized Clinical Trial. Circulation 2022; 146:1357-1366. [PMID: 36168956 DOI: 10.1161/circulationaha.122.060106] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND This study was conducted to determine the effect of hypothermic temperature control after in-hospital cardiac arrest (IHCA) on mortality and functional outcome as compared with normothermia. METHODS An investigator initiated, open-label, blinded-outcome-assessor, multicenter, randomized controlled trial comparing hypothermic temperature control (32-34°C) for 24 h with normothermia after IHCA in 11 hospitals in Germany. The primary endpoint was all-cause mortality after 180 days. Secondary end points included in-hospital mortality and favorable functional outcome using the Cerebral Performance Category scale after 180 days. A Cerebral Performance Category score of 1 or 2 was defined as a favorable functional outcome. RESULTS A total of 1055 patients were screened for eligibility and 249 patients were randomized: 126 were assigned to hypothermic temperature control and 123 to normothermia. The mean age of the cohort was 72.6±10.4 years, 64% (152 of 236) were male, 73% (166 of 227) of cardiac arrests were witnessed, 25% (57 of 231) had an initial shockable rhythm, and time to return of spontaneous circulation was 16.4±10.5 minutes. Target temperature was reached within 4.2±2.8 hours after randomization in the hypothermic group and temperature was controlled for 48 hours at 37.0°±0.9°C in the normothermia group. Mortality by day 180 was 72.5% (87 of 120) in hypothermic temperature control arm, compared with 71.2% (84 of 118) in the normothermia group (relative risk, 1.03 [95% CI, 0.79-1.40]; P=0.822). In-hospital mortality was 62.5% (75 of 120) in the hypothermic temperature control as compared with 57.6% (68 of 118) in the normothermia group (relative risk, 1.11 [95% CI, 0.86-1.46, P=0.443). Favorable functional outcome (Cerebral Performance Category 1 or 2) by day 180 was 22.5% (27 of 120) in the hypothermic temperature control, compared with 23.7% (28 of 118) in the normothermia group (relative risk, 1.04 [95% CI, 0.78-1.44]; P=0.822). The study was prematurely terminated because of futility. CONCLUSIONS Hypothermic temperature control as compared with normothermia did not improve survival nor functional outcome at day 180 in patients presenting with coma after IHCA. The HACA in-hospital trial (Hypothermia After Cardiac Arrest in-hospital) was underpowered and may have failed to detect clinically important differences between hypothermic temperature control and normothermia. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique Identifier: NCT00457431.
Collapse
Affiliation(s)
- Sebastian Wolfrum
- Emergency Department (S.W., A.H.), University of Luebeck, Germany.,Medical Clinic II, Department of Cardiology, Angiology and Intensive Care Medicine (S.W., A.H., V.K., H.S.), University of Luebeck, Germany
| | - Kevin Roedl
- Department of Intensive Care Medicine (K.R., S.B., G.S., S.K.), University Medical Centre Hamburg-Eppendorf, Germany
| | - Alexia Hanebutte
- Emergency Department (S.W., A.H.), University of Luebeck, Germany.,Medical Clinic II, Department of Cardiology, Angiology and Intensive Care Medicine (S.W., A.H., V.K., H.S.), University of Luebeck, Germany
| | - Rüdiger Pfeifer
- Department of Internal Medicine 1, University Hospital of Jena, Germany (R.P.)
| | - Volkhard Kurowski
- Department of Intensive Care Medicine (K.R., S.B., G.S., S.K.), University Medical Centre Hamburg-Eppendorf, Germany.,Department of Cardiology and Intensive Care Medicine, DRK Hospital, Ratzeburg, Germany (V.K.)
| | - Reimer Riessen
- Department of Medicine, Medical Intensive Care Unit, University of Tübingen, Germany (R.R.)
| | - Anne Daubmann
- Institute of Medical Biometry and Epidemiology (A.D., E.B.-F.' K.W.), University Medical Centre Hamburg-Eppendorf, Germany
| | - Stephan Braune
- Department of Intensive Care Medicine (K.R., S.B., G.S., S.K.), University Medical Centre Hamburg-Eppendorf, Germany
| | - Gerold Söffker
- Department of Intensive Care Medicine (K.R., S.B., G.S., S.K.), University Medical Centre Hamburg-Eppendorf, Germany
| | - Eric Bibiza-Freiwald
- Institute of Medical Biometry and Epidemiology (A.D., E.B.-F.' K.W.), University Medical Centre Hamburg-Eppendorf, Germany
| | - Karl Wegscheider
- Institute of Medical Biometry and Epidemiology (A.D., E.B.-F.' K.W.), University Medical Centre Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK e.V.)' Partner Site Hamburg/Kiel/Lübeck' Hamburg' Germany (K.W.)
| | - Heribert Schunkert
- Medical Clinic II, Department of Cardiology, Angiology and Intensive Care Medicine (S.W., A.H., V.K., H.S.), University of Luebeck, Germany.,German Heart Center Munich, Department of Cardiology' Technical University of Munich' German Center for Cardiovascular Research (DZHK) - Munich Heart Alliance (H.S.)
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Germany (H.T.)
| | - Stefan Kluge
- Department of Intensive Care Medicine (K.R., S.B., G.S., S.K.), University Medical Centre Hamburg-Eppendorf, Germany
| | | |
Collapse
|
40
|
Shen L, Zhang J, DeLucca P. Sample size calculation and timing of dose selection in an adaptive multiple-dose clinical trial. Stat Biopharm Res 2022. [DOI: 10.1080/19466315.2022.2116101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
41
|
Reference curve sampling variability in one–sample log–rank tests. PLoS One 2022; 17:e0271094. [PMID: 35862473 PMCID: PMC9302761 DOI: 10.1371/journal.pone.0271094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
The one–sample log–rank test is the method of choice for single–arm Phase II trials with time–to–event endpoint. It allows to compare the survival of patients to a reference survival curve that typically represents the expected survival under standard of care. The one–sample log–rank test, however, assumes that the reference survival curve is known. This ignores that the reference curve is commonly estimated from historic data and thus prone to sampling error. Ignoring sampling variability of the reference curve results in type I error rate inflation. We study this inflation in type I error rate analytically and by simulation. Moreover we derive the actual distribution of the one–sample log–rank test statistic, when the sampling variability of the reference curve is taken into account. In particular, we provide a consistent estimate of the factor by which the true variance of the one-sample log–rank statistic is underestimated when reference curve sampling variability is ignored. Our results are further substantiated by a case study using a real world data example in which we demonstrate how to estimate the error rate inflation in the planning stage of a trial.
Collapse
|
42
|
Liu B. Statistical Analysis of Employment Education in Colleges and Universities Based on Improved Clustering Algorithm. SECURITY AND COMMUNICATION NETWORKS 2022; 2022:1-12. [DOI: 10.1155/2022/5776831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
In order to improve the effect of employment education in colleges and universities, this study combines the improved clustering algorithm to carry out statistical analysis of employment education in colleges and universities and analyzes the current situation of employment education in colleges and universities. In cluster analysis, considering the complexity of the distribution of statistics after the reestimation of the sample size and the possible complex correlation between the series of statistics in the group sequential design, on the premise that the statistics used will not cause type I error expansion after sample size adjustment, some rules for reestimating the sample size can be formulated. Through the simulation analysis, it can be seen that the statistical analysis system of college employment education based on the improved clustering algorithm proposed in this study has a good effect in the clustering of employment education data and has a certain role in promoting the employment of college graduates.
Collapse
Affiliation(s)
- Bin Liu
- Xinyang Vocational and Technical College, Xinyang 464000, China
| |
Collapse
|
43
|
Zhang W, Ro S, Jiang Q, Li X, Liu R, Lu C'C, Marchenko O, Zhao J, Xu Z. Statistical and Operational Considerations for 2-Stage Adaptive Designs with Simultaneous Evaluation of Overall and Marker-Selected Populations in Oncology Confirmatory Trials. Ther Innov Regul Sci 2022; 56:552-560. [PMID: 35503503 DOI: 10.1007/s43441-022-00407-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
In biomarker enrichment study designs that start with an all-comer population, simultaneous evaluation of the entire and the marker-selected populations can be more desirable than pre-specifying the testing order, when the degree of marker predictiveness is uncertain. While there has been substantial research on this approach, our goal is to provide a complete overview and guidance in all aspects of this approach, including the interim analysis potentially using different endpoints, combination tests with associated multiplicity control, and the final treatment effect estimation. Regulatory/operational aspects and actual cases demonstrating the potential advantage of this approach are also described.
Collapse
Affiliation(s)
| | - Sunhee Ro
- Sierra Oncology, Inc., San Mateo, CA, USA
| | | | | | - Rong Liu
- Bristol Myers Squibb, Co., New York, NY, USA
| | | | | | - Jing Zhao
- Merck & Co, Inc., Kenilworth, NJ, USA
| | - Zhenzhen Xu
- Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
44
|
Takahashi K, Ishii R, Maruo K, Gosho M. Statistical tests for two-stage adaptive seamless design using short- and long-term binary outcomes. Stat Med 2022; 41:4130-4142. [PMID: 35713225 DOI: 10.1002/sim.9500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 11/09/2022]
Abstract
The adaptive seamless design combining phases II and III into a single trial has been shown growing interest for improving the efficiency of drug development, becoming the most frequent adaptive design type. It typically consists of two stages, the trial objectives being often different in each stage. The primary objectives are to select optimal experimental treatment group(s) in the first stage and compare the efficacy between the selected treatment and control groups in the second stage. In this article, we focus on a two-stage adaptive seamless design, for which treatment selection is based on the short-term binary endpoint and treatment comparison is based on the long-term binary endpoint. We thus propose an exact conditional test as a final analysis, based on the bivariate binomial distribution and given the selected treatment with the most promising short-term endpoint response rate from an interim analysis. Additionally, the mid- p $$ p $$ approach is incorporated to improve conservativeness for an exact test. Simulation studies were conducted to compare the proposed methods with a method based on the combination test. The proposed exact method controlled for type I error rate at the nominal level, regardless of the number of initial treatments or the correlation between short- and long-term endpoints. In terms of the treatment comparison power, the proposed methods are more powerful than that based on the combination test in the scenarios, with only one treatment being effective.
Collapse
Affiliation(s)
- Kenichi Takahashi
- Japan Development, MSD K. K., Tokyo, Japan.,Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Ryota Ishii
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazushi Maruo
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masahiko Gosho
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
45
|
Pilz M, Herrmann C, Rauch G, Kieser M. Optimal unplanned design modification in adaptive two-stage trials. Pharm Stat 2022; 21:1121-1137. [PMID: 35604767 DOI: 10.1002/pst.2228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 02/01/2022] [Accepted: 04/24/2022] [Indexed: 11/08/2022]
Abstract
Adaptive planning of clinical trials allows modifying the entire trial design at any time point mid-course. In this paper, we consider the case when a trial-external update of the planning assumptions during the ongoing trial makes an unforeseen design adaptation necessary. We take up the idea to construct adaptive designs with defined features by solving an optimization problem and apply it to the situation of unplanned design reassessment. By using the conditional error principle, we present an approach on how to optimally modify the trial design at an unplanned interim analysis while at the same time strictly protecting the type I error rate. This linking of optimal design planning and the conditional error principle allows sound reactions to unforeseen events that make a design reassessment necessary.
Collapse
Affiliation(s)
- Maximilian Pilz
- Institute of Medical Biometry, University Medical Center Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Carolin Herrmann
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Meinhard Kieser
- Institute of Medical Biometry, University Medical Center Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| |
Collapse
|
46
|
Nelson BS, Liu L, Mehta C. A simulation-based comparison of estimation methods for adaptive and classical group sequential clinical trials. Pharm Stat 2022; 21:599-611. [PMID: 34957677 DOI: 10.1002/pst.2188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/19/2021] [Accepted: 12/12/2021] [Indexed: 11/12/2022]
Abstract
Statistical methods for controlling the type-I error of hypothesis tests in adaptive group sequential clinical trials are well established and well understood. However, methods for obtaining statistically valid point estimates and confidence intervals for adaptive designs are not as well established or as well understood. At the end of an adaptive trial, one may calculate the repeated confidence interval (RCI), which provides conservative coverage of δ , or the backward image confidence interval (BWCI), which provides exact coverage of δ and is an extension of the stagewise adjusted confidence interval (SWCI, used in classical group sequential designs). The BWCI can also provide a median unbiased estimate (MUE) of δ . There is a need to better understand the coverage and possible biases associated with these methods. We conducted a simulation study exploring parameter estimation following sample size reestimation based on testing methods with strong control of type-I error. Generally, the BWCI provided exact coverage, the naïve CI provided inconsistent coverage, and the RCI provided conservative coverage. Additionally, we note considerable asymmetry in the coverage from above/from below for the RCI, although we did not see any instance where the 95% RCI excluded the true parameter more than 2.5% on either side. At the end of an adaptive group sequential trial, we strongly recommend the use of the BWCI (and associated MUE), with the RCI computed during interim looks; the naïve CI should be avoided. These results and conclusions also hold true for classical group sequential designs.
Collapse
Affiliation(s)
- Bryan S Nelson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Lingyun Liu
- Vertex Pharmaceuticals, Boston, Massachusetts, USA
| | - Cyrus Mehta
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Cytel Corporation, Cambridge, Massachusetts, USA
| |
Collapse
|
47
|
Herrmann C, Kieser M, Rauch G, Pilz M. Optimization of adaptive designs with respect to a performance score. Biom J 2022; 64:989-1006. [PMID: 35426460 DOI: 10.1002/bimj.202100166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 11/08/2022]
Abstract
Adaptive designs are an increasingly popular method for the adaptation of design aspects in clinical trials, such as the sample size. Scoring different adaptive designs helps to make an appropriate choice among the numerous existing adaptive design methods. Several scores have been proposed to evaluate adaptive designs. Moreover, it is possible to determine optimal two-stage adaptive designs with respect to a customized objective score by solving a constrained optimization problem. In this paper, we use the conditional performance score by Herrmann et al. (2020) as the optimization criterion to derive optimal adaptive two-stage designs. We investigate variations of the original performance score, for example, by assigning different weights to the score components and by incorporating prior assumptions on the effect size. We further investigate a setting where the optimization framework is extended by a global power constraint, and additional optimization of the critical value function next to the stage-two sample size is performed. Those evaluations with respect to the sample size curves and the resulting design's performance can contribute to facilitate the score's usage in practice.
Collapse
Affiliation(s)
- Carolin Herrmann
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Meinhard Kieser
- Institute of Medical Biometry, University Hospital Heidelberg, Heidelberg, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maximilian Pilz
- Institute of Medical Biometry, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
48
|
Li X, Hu F. Sample size re-estimation for response-adaptive randomized clinical trials. Pharm Stat 2022; 21:1058-1073. [PMID: 35191605 DOI: 10.1002/pst.2199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/23/2022] [Accepted: 02/02/2022] [Indexed: 11/10/2022]
Abstract
Clinical trials usually take a period of time to recruit volunteers, and they become a steady accumulation of data. Traditionally, the sample size of a trial is determined in advance and data is collected before analysis proceeds. Over the past decades, many strategies have been proposed and rigorous theoretical groundings have been provided to conduct sample size re-estimation. However, the application of these methodologies has not been well extended to take care of trials with adaptive designs. Therefore, we aim to fill the gap by proposing a sample size re-estimation procedure on response-adaptive randomized trial. For ethical and economical concerns, we use multiple stopping criteria with the allowance of early termination. Statistical inference is studied for the hypothesis testing under doubly-adaptive biased coin design. We also prove that the test statistics for each stage are asymptotic independently normally distributed, though dependency exists between the two stages. We find that under our methods, compared to fixed sample size design and other commonly used randomization procedures: (1) power is increased for all scenarios with adjusted sample size; (2) sample size is reduced up to 40% when underestimating the treatment effect; (3) the duration of trials is shortened. These advantages are evidenced by numerical studies and real examples.
Collapse
Affiliation(s)
- Xin Li
- Department of Statistics, George Washington University, Washington, District of Columbia, USA
| | - Feifang Hu
- Department of Statistics, George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
49
|
Arjas E, Gasbarra D. Adaptive treatment allocation and selection in multi-arm clinical trials: a Bayesian perspective. BMC Med Res Methodol 2022; 22:50. [PMID: 35184731 PMCID: PMC8858379 DOI: 10.1186/s12874-022-01526-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adaptive designs offer added flexibility in the execution of clinical trials, including the possibilities of allocating more patients to the treatments that turned out more successful, and early stopping due to either declared success or futility. Commonly applied adaptive designs, such as group sequential methods, are based on the frequentist paradigm and on ideas from statistical significance testing. Interim checks during the trial will have the effect of inflating the Type 1 error rate, or, if this rate is controlled and kept fixed, lowering the power. RESULTS The purpose of the paper is to demonstrate the usefulness of the Bayesian approach in the design and in the actual running of randomized clinical trials during phase II and III. This approach is based on comparing the performance of the different treatment arms in terms of the respective joint posterior probabilities evaluated sequentially from the accruing outcome data, and then taking a control action if such posterior probabilities fall below a pre-specified critical threshold value. Two types of actions are considered: treatment allocation, putting on hold at least temporarily further accrual of patients to a treatment arm, and treatment selection, removing an arm from the trial permanently. The main development in the paper is in terms of binary outcomes, but extensions for handling time-to-event data, including data from vaccine trials, are also discussed. The performance of the proposed methodology is tested in extensive simulation experiments, with numerical results and graphical illustrations documented in a Supplement to the main text. As a companion to this paper, an implementation of the methods is provided in the form of a freely available R package 'barts'. CONCLUSION The proposed methods for trial design provide an attractive alternative to their frequentist counterparts.
Collapse
Affiliation(s)
- Elja Arjas
- University of Helsinki, Helsinki, Finland.
- University of Oslo, Oslo, Norway.
| | | |
Collapse
|
50
|
Zhan T, Kang J. Finite-Sample Two-Group Composite Hypothesis Testing via Machine Learning. J Comput Graph Stat 2022; 31:856-865. [PMID: 36506350 PMCID: PMC9733814 DOI: 10.1080/10618600.2021.2020128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 09/28/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022]
Abstract
In the problem of composite hypothesis testing, identifying the potential uniformly most powerful (UMP) unbiased test is of great interest. Beyond typical hypothesis settings with exponential family, it is usually challenging to prove the existence and further construct such UMP unbiased tests with finite sample size. For example in the COVID-19 pandemic with limited previous assumptions on the treatment for investigation and the standard of care, adaptive clinical trials are appealing due to ethical considerations, and the ability to accommodate uncertainty while conducting the trial. Although several methods have been proposed to control Type I error rates, how to find a more powerful hypothesis testing strategy is still an open question. Motivated by this problem, we propose an automatic framework of constructing test statistics and corresponding critical values via machine learning methods to enhance power in a finite sample. In this article, we particularly illustrate the performance using Deep Neural Networks (DNN) and discuss its advantages. Simulations and two case studies of adaptive designs demonstrate that our method is automatic, general and prespecified to construct statistics with satisfactory power in finite-sample. Supplemental materials are available online including R code and an R shiny app.
Collapse
Affiliation(s)
- Tianyu Zhan
- Data and Statistical Sciences, AbbVie Inc., North Chicago, IL
| | - Jian Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| |
Collapse
|