1
|
Moon KZ, Rahman MH, Alam MJ, Hossain MA, Hwang S, Kang S, Moon S, Park MN, Ahn CH, Kim B. Unraveling the interplay between cardiovascular diseases and alcohol use disorder: A bioinformatics and network-based exploration of shared molecular pathways and key biomarkers validation via western blot analysis. Comput Biol Chem 2025; 115:108338. [PMID: 39778286 DOI: 10.1016/j.compbiolchem.2024.108338] [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: 09/23/2024] [Revised: 12/02/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Clinical observations indicate a pronounced exacerbation of Cardiovascular Diseases (CVDs) in individuals grappling with Alcohol Use Disorder (AUD), suggesting an intricate interplay between these maladies. Pinpointing shared risk factors for both conditions has proven elusive. To address this, we pioneered a sophisticated bioinformatics framework and network-based strategy to unearth genes exhibiting aberrant expression patterns in both AUD and CVDs. In heart tissue samples from patients battling both AUD and CVDs, our study identified 76 Differentially Expressed Genes (DEGs) further used for retrieving important Gene Ontology (GO) keywords and metabolic pathways, highlighting mechanisms like proinflammatory cascades, T-cell cytotoxicity, antigen processing and presentation. By using Protein-Protein Interaction (PPI) analysis, we were able to identify key hub proteins that have a significant impact on the pathophysiology of these illnesses. Several hub proteins were identified include PTGS2, VCAM1, CCL2, CXCL8, IL7R, among these only CDH1 was covered in 10 algorithms of cytoHubba plugin. Furthermore, we pinpointed several Transcription Factors (TFs), including SOD2, CXCL8, THBS2, GREM1, CCL2, and PTGS2, alongside potential microRNAs (miRNAs) such as hsa-mir-203a-3p, hsa-mir-23a-3p, hsa-mir-98-5p, and hsa-mir-7-5p, which exert critical regulatory control over gene expression… In vitro study investigates the effect of alcohol on E-cadherin (CDH1) expression in HepG2 and Hep3B cells, showing a significant decrease in expression following ethanol treatment. These findings suggest that alcohol exposure may disrupt cell adhesion, potentially contributing to cellular changes associated with cardiovascular diseases. Our innovative approach has unveiled distinctive biomarkers delineating the dynamic interplay between AUD and various cardiovascular conditions for future therapeutic exploration.
Collapse
Affiliation(s)
- Kamelia Zaman Moon
- Department of Computer Science and Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh.
| | - Md Jahangir Alam
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh
| | - Md Arju Hossain
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Sungho Hwang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chi-Hoon Ahn
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
2
|
Dekker EN, Janssen QP, van Dam JL, Strijk GJ, Verkolf EMM, Kandala S, Dumas J, Fellah A, O'Reilly EM, Besselink MG, van Eijck CHJ, Homs MYV, van Tienhoven GJ, Wilmink JW, Mustafa DAM, Groot Koerkamp B. Blood Sample Collection in Randomized Controlled Trials for Biomarker Discovery and Validation: Experience of the PREOPANC-2 Trial. Ann Surg Oncol 2025:10.1245/s10434-025-16890-0. [PMID: 39907876 DOI: 10.1245/s10434-025-16890-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND This study aimed to investigate the feasibility and yield of blood sample collection in an investigator-initiated nationwide randomized controlled trial (RCT). METHODS In the PREOPANC-2 trial, 375 patients with (borderline) resectable pancreatic cancer were randomly assigned to two neoadjuvant regiments in 19 centers in the Netherlands (2018-2021). Blood sample collection was scheduled at seven time points before, during, and after treatment. The primary outcome was the proportion of successfully collected blood samples at each scheduled time point. RESULTS Of the 375 randomized patients, 12 were excluded from blood sample collection before any treatment. From the remaining 363 patients, 1513 (87 %) of 1748 blood samples were collected, processed, mailed, and centrally stored. The blood samples were collected before treatment from 347 (96 %) of the 363 patients, after the first neoadjuvant cycle from 322 (94 %) of 343 patients, after neoadjuvant treatment (i.e., before surgery) from 260 (83 %) of 313 patients, and after surgery from 210 (77 %) of 271 patients. During the follow-up visits, blood samples were collected from 147 (82 %) of 179 patients 12 months after randomization and from 83 (77 %) of 108 patients after 24 months. A total of 220 samples (13 %) were missing. The most common causes for missing blood samples were scheduling oversights, unsuccessful blood draw attempts, and mailing failures (151 times, 69 %). Blood sample collection was canceled 69 times (31 %) due to COVID-19. CONCLUSION Blood sample collection in the PREOPANC-2 trial had a yield of 96 % before treatment and an overall yield of 87 %. Collection of blood samples for biomarker studies is feasible in a nationwide RCT.
Collapse
Affiliation(s)
- Esther N Dekker
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Quisette P Janssen
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jacob L van Dam
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Gaby J Strijk
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Eva M M Verkolf
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sridhar Kandala
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jasper Dumas
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, Rotterdam, The Netherlands
| | - Amine Fellah
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, Rotterdam, The Netherlands
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc G Besselink
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Marjolein Y V Homs
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Geert-Jan van Tienhoven
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Akhunzianov AA, Rozhina EV, Filina YV, Rizvanov AA, Miftakhova RR. Resistance to Radiotherapy in Cancer. Diseases 2025; 13:22. [PMID: 39851486 PMCID: PMC11764699 DOI: 10.3390/diseases13010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Radiation therapy or radiotherapy is a medical treatment that uses high doses of ionizing radiation to eliminate cancer cells and shrink tumors. It works by targeting the DNA within the tumor cells restricting their proliferation. Radiotherapy has been used for treating cancer for more than 100 years. Along with surgery and chemotherapy, it is one of the three main and most common approaches used in cancer therapy. Nowadays, radiotherapy has become a standard treatment option for a wide range of cancers around the world, including lung, breast, cervical, and colorectal cancers. Around 50% of all patients will require radiotherapy, 60% of whom are treated with curative intent. Moreover, it is commonly used for palliative treatment. Radiotherapy provides 5-year local control and overall survival benefit in 10.4% and 2.4% of all cancer patients, respectively. The highest local control benefit is reported for cervical (33%), head and neck (32%), and prostate (26%) cancers. But no benefit is observed in pancreas, ovary, liver, kidney, and colon cancers. Such relatively low efficiency is related to the development of radiation resistance, which results in cancer recurrence, metastatic dissemination, and poor prognosis. The identification of radioresistance biomarkers allows for improving the treatment outcome. These biomarkers mainly include proteins involved in metabolism and cell signaling pathways.
Collapse
Affiliation(s)
- Almaz A. Akhunzianov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Elvira V. Rozhina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Yuliya V. Filina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| | - Regina R. Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
4
|
Taylor C, Patterson KM, Friedman D, Bacot SM, Feldman GM, Wang T. Mechanistic Insights into the Successful Development of Combination Therapy of Enfortumab Vedotin and Pembrolizumab for the Treatment of Locally Advanced or Metastatic Urothelial Cancer. Cancers (Basel) 2024; 16:3071. [PMID: 39272928 PMCID: PMC11393896 DOI: 10.3390/cancers16173071] [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: 07/31/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Antibody-drug conjugates (ADCs) consist of an antibody backbone that recognizes and binds to a target antigen expressed on tumor cells and a small molecule chemotherapy payload that is conjugated to the antibody via a linker. ADCs are one of the most promising therapeutic modalities for the treatment of various cancers. However, many patients have developed resistance to this form of therapy. Extensive efforts have been dedicated to identifying an effective combination of ADCs with other types of anticancer therapies to potentially overcome this resistance. A recent clinical study demonstrated that a combination of the ADC enfortumab vedotin (EV) with the immune checkpoint inhibitor (ICI) pembrolizumab can achieve remarkable clinical efficacy as the first-line therapy for the treatment of locally advanced or metastatic urothelial carcinoma (la/mUC)-leading to the first approval of a combination therapy of an ADC with an ICI for the treatment of cancer patients. In this review, we highlight knowledge and understanding gained from the successful development of EV and the combination therapy of EV with ICI for the treatment of la/mUC. Using urothelial carcinoma as an example, we will focus on dissecting the underlying mechanisms necessary for the development of this type of combination therapy for a variety of cancers.
Collapse
Affiliation(s)
- Caroline Taylor
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kamai M Patterson
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Devira Friedman
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Silvia M Bacot
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Gerald M Feldman
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Tao Wang
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
5
|
Wang D, Wang SJ, Lababidi S. The impact of misclassification errors on the performance of biomarkers based on next-generation sequencing, a simulation study. J Biopharm Stat 2024; 34:700-718. [PMID: 37819021 DOI: 10.1080/10543406.2023.2269251] [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: 05/07/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
The development of next-generation sequencing (NGS) opens opportunities for new applications such as liquid biopsy, in which tumor mutation genotypes can be determined by sequencing circulating tumor DNA after blood draws. However, with highly diluted samples like those obtained with liquid biopsy, NGS invariably introduces a certain level of misclassification, even with improved technology. Recently, there has been a high demand to use mutation genotypes as biomarkers for predicting prognosis and treatment selection. Many methods have also been proposed to build classifiers based on multiple loci with machine learning algorithms as biomarkers. How the higher misclassification rate introduced by liquid biopsy will affect the performance of these biomarkers has not been thoroughly investigated. In this paper, we report the results from a simulation study focused on the clinical utility of biomarkers when misclassification is present due to the current technological limit of NGS in the liquid biopsy setting. The simulation covers a range of performance profiles for current NGS platforms with different machine learning algorithms and uses actual patient genotypes. Our results show that, at the high end of the performance spectrum, the misclassification introduced by NGS had very little effect on the clinical utility of the biomarker. However, in more challenging applications with lower accuracy, misclassification could have a notable effect on clinical utility. The pattern of this effect can be complex, especially for machine learning-based classifiers. Our results show that simulation can be an effective tool for assessing different scenarios of misclassification.
Collapse
Affiliation(s)
- Dong Wang
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Sue-Jane Wang
- Office of Biostatistics, Center for Drug Evaluation Research, FDA, Maryland, USA
| | - Samir Lababidi
- Office of Data, Analytics and Research, Office of Digital Transformation, Office of Commissioner, FDA, Maryland, USA
| |
Collapse
|
6
|
Verschoor N, Bos MK, Oomen-de Hoop E, Martens JWM, Sleijfer S, Jager A, Beije N. A review of trials investigating ctDNA-guided adjuvant treatment of solid tumors: The importance of trial design. Eur J Cancer 2024; 207:114159. [PMID: 38878446 DOI: 10.1016/j.ejca.2024.114159] [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: 04/16/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 07/14/2024]
Abstract
Circulating tumor DNA (ctDNA) holds promise as a biomarker for guiding adjuvant treatment decisions in solid tumors. This review systematically assembles ongoing and published trials investigating ctDNA-directed adjuvant treatment strategies. A total of 57 phase II/III trials focusing on ctDNA in minimal residual disease (MRD) detection were identified, with a notable increase in initiation over recent years. Most trials target stage II or III colon/colorectal cancer, followed by breast cancer and non-small cell lung cancer. Trial methodologies vary, with some randomizing ctDNA-positive patients between standard-of-care (SoC) treatment and intensified regimens, while others aim to de-escalate therapy in ctDNA-negative patients. Challenges in trial design include the need for randomized controlled trials to establish clinical utility for ctDNA, ensuring adherence to standard treatment in control arms, and addressing the ethical dilemma of withholding treatment in high-risk ctDNA-positive patients. Longitudinal ctDNA surveillance emerges as a strategy to improve sensitivity for recurrence, particularly in less proliferative tumor types. However, ctDNA as longitudinal marker is often not validated yet. Ultimately, designing effective ctDNA interventional trials requires careful consideration of feasibility, meaningful outcomes, and potential impact on patient care.
Collapse
Affiliation(s)
- Noortje Verschoor
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, the Netherlands.
| | - Manouk K Bos
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, the Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, the Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, the Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, the Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, the Netherlands
| | - Nick Beije
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, the Netherlands
| |
Collapse
|
7
|
Chen Y, Lin Y, Lu SE, Shih WJ, Quan H. Two-stage stratified designs with survival outcomes and adjustment for misclassification in predictive biomarkers. Stat Med 2024; 43:1883-1904. [PMID: 38634277 PMCID: PMC11068307 DOI: 10.1002/sim.10048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 09/09/2023] [Accepted: 02/12/2024] [Indexed: 04/19/2024]
Abstract
Biomarker stratified clinical trial designs are versatile tools to assess biomarker clinical utility and address its relationship with clinical endpoints. Due to imperfect assays and/or classification rules, biomarker status is prone to errors. To account for biomarker misclassification, we consider a two-stage stratified design for survival outcomes with an adjustment for misclassification in predictive biomarkers. Compared to continuous and/or binary outcomes, the test statistics for survival outcomes with an adjustment for biomarker misclassification is much more complicated and needs to take special care. We propose to use the information from the observed biomarker status strata to construct adjusted log-rank statistics for true biomarker status strata. These adjusted log-rank statistics are then used to develop sequential tests for the global (composite) hypothesis and component-wise hypothesis. We discuss the power analysis with the control of the type-I error rate by using the correlations between the adjusted log-rank statistics within and between the design stages. Our method is illustrated with examples of the recent successful development of immunotherapy in nonsmall-cell lung cancer.
Collapse
Affiliation(s)
- Yanping Chen
- Global Biometrics and Data Sciences, Bristol Myers Squibb,
Berkeley Heights, New Jersey, USA
| | - Yong Lin
- Biostatistics and Epidemiology Department, School of Public
Health, Rutgers University, Piscataway, New Jersey, USA
- Biometrics Division, Rutgers Cancer Institute of New
Jersey, New Brunswick, New Jersey, USA
| | - Shou-En Lu
- Biostatistics and Epidemiology Department, School of Public
Health, Rutgers University, Piscataway, New Jersey, USA
- Biometrics Division, Rutgers Cancer Institute of New
Jersey, New Brunswick, New Jersey, USA
| | - Weichung J. Shih
- Biostatistics and Epidemiology Department, School of Public
Health, Rutgers University, Piscataway, New Jersey, USA
- Biometrics Division, Rutgers Cancer Institute of New
Jersey, New Brunswick, New Jersey, USA
| | - Hui Quan
- Biostatistics and Programming, Sanofi, Bridgewater, New
Jersey, USA
| |
Collapse
|
8
|
Freidlin B, Korn EL. Efficiency of Biomarker-Driven Clinical Trial Designs. J Clin Oncol 2024; 42:1454-1455. [PMID: 38437594 PMCID: PMC11095868 DOI: 10.1200/jco.23.02581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/05/2024] [Indexed: 03/06/2024] Open
Affiliation(s)
- Boris Freidlin
- Boris Freidlin, PhD and Edward L. Korn, PhD, Biostatistics Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Edward L. Korn
- Boris Freidlin, PhD and Edward L. Korn, PhD, Biostatistics Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| |
Collapse
|
9
|
Vinnat V, Annane D, Chevret S. Bayesian Sequential Design for Identifying and Ranking Effective Patient Subgroups in Precision Medicine in the Case of Counting Outcome Data with Inflated Zeros. J Pers Med 2023; 13:1560. [PMID: 38003875 PMCID: PMC10672716 DOI: 10.3390/jpm13111560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/23/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Precision medicine is revolutionizing health care, particularly by addressing patient variability due to different biological profiles. As traditional treatments may not always be appropriate for certain patient subsets, the rise of biomarker-stratified clinical trials has driven the need for innovative methods. We introduced a Bayesian sequential scheme to evaluate therapeutic interventions in an intensive care unit setting, focusing on complex endpoints characterized by an excess of zeros and right truncation. By using a zero-inflated truncated Poisson model, we efficiently addressed this data complexity. The posterior distribution of rankings and the surface under the cumulative ranking curve (SUCRA) approach provided a comprehensive ranking of the subgroups studied. Different subsets of subgroups were evaluated depending on the availability of biomarker data. Interim analyses, accounting for early stopping for efficacy, were an integral aspect of our design. The simulation study demonstrated a high proportion of correct identification of the subgroup which is the most predictive of the treatment effect, as well as satisfactory false positive and true positive rates. As the role of personalized medicine grows, especially in the intensive care setting, it is critical to have designs that can manage complicated endpoints and that can control for decision error. Our method seems promising in this challenging context.
Collapse
Affiliation(s)
- Valentin Vinnat
- ECSTRRA Team, INSERM U1153, Université Paris Cité, 75010 Paris, France;
| | - Djillali Annane
- Intensive Care Unit, Raymond Poincaré Hospital, 78266 Garches, France;
| | - Sylvie Chevret
- ECSTRRA Team, INSERM U1153, Université Paris Cité, 75010 Paris, France;
- Institut Universitaire de France (IUF), 75231 Paris, France
| |
Collapse
|
10
|
Alderuccio JP, Kuker RA, Yang F, Moskowitz CH. Quantitative PET-based biomarkers in lymphoma: getting ready for primetime. Nat Rev Clin Oncol 2023; 20:640-657. [PMID: 37460635 DOI: 10.1038/s41571-023-00799-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 08/20/2023]
Abstract
The use of functional quantitative biomarkers extracted from routine PET-CT scans to characterize clinical responses in patients with lymphoma is gaining increased attention, and these biomarkers can outperform established clinical risk factors. Total metabolic tumour volume enables individualized estimation of survival outcomes in patients with lymphoma and has shown the potential to predict response to therapy suitable for risk-adapted treatment approaches in clinical trials. The deployment of machine learning tools in molecular imaging research can assist in recognizing complex patterns and, with image classification, in tumour identification and segmentation of data from PET-CT scans. Initial studies using fully automated approaches to calculate metabolic tumour volume and other PET-based biomarkers have demonstrated appropriate correlation with calculations from experts, warranting further testing in large-scale studies. The extraction of computer-based quantitative tumour characterization through radiomics can provide a comprehensive view of phenotypic heterogeneity that better captures the molecular and functional features of the disease. Additionally, radiomics can be integrated with genomic data to provide more accurate prognostic information. Further improvements in PET-based biomarkers are imminent, although their incorporation into clinical decision-making currently has methodological shortcomings that need to be addressed with confirmatory prospective validation in selected patient populations. In this Review, we discuss the current knowledge, challenges and opportunities in the integration of quantitative PET-based biomarkers in clinical trials and the routine management of patients with lymphoma.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Russ A Kuker
- Department of Radiology, Division of Nuclear Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fei Yang
- Department of Radiation Oncology, Division of Medical Physics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Craig H Moskowitz
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
11
|
Davey A, Thor M, van Herk M, Faivre-Finn C, Rimner A, Deasy JO, McWilliam A. Predicting cancer relapse following lung stereotactic radiotherapy: an external validation study using real-world evidence. Front Oncol 2023; 13:1156389. [PMID: 37503315 PMCID: PMC10369005 DOI: 10.3389/fonc.2023.1156389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Purpose For patients receiving lung stereotactic ablative radiotherapy (SABR), evidence suggests that high peritumor density predicts an increased risk of microscopic disease (MDE) and local-regional failure, but only if there is low or heterogenous incidental dose surrounding the tumor (GTV). A data-mining method (Cox-per-radius) has been developed to investigate this dose-density interaction. We apply the method to predict local relapse (LR) and regional failure (RF) in patients with non-small cell lung cancer. Methods 199 patients treated in a routine setting were collated from a single institution for training, and 76 patients from an external institution for validation. Three density metrics (mean, 90th percentile, standard deviation (SD)) were studied in 1mm annuli between 0.5cm inside and 2cm outside the GTV boundary. Dose SD and fraction of volume receiving less than 30Gy were studied in annuli 0.5-2cm outside the GTV to describe incidental MDE dosage. Heat-maps were created that correlate with changes in LR and RF rates due to the interaction between dose heterogeneity and density at each distance combination. Regions of significant improvement were studied in Cox proportional hazards models, and explored with and without re-fitting in external data. Correlations between the dose component of the interaction and common dose metrics were reported. Results Local relapse occurred at a rate of 6.5% in the training cohort, and 18% in the validation cohort, which included larger and more centrally located tumors. High peritumor density in combination with high dose variability (0.5 - 1.6cm) predicts LR. No interactions predicted RF. The LR interaction improved the predictive ability compared to using clinical variables alone (optimism-adjusted C-index; 0.82 vs 0.76). Re-fitting model coefficients in external data confirmed the importance of this interaction (C-index; 0.86 vs 0.76). Dose variability in the 0.5-1.6 cm annular region strongly correlates with heterogeneity inside the target volume (SD; ρ = 0.53 training, ρ = 0.65 validation). Conclusion In these real-world cohorts, the combination of relatively high peritumor density and high dose variability predicts increase in LR, but not RF, following lung SABR. This external validation justifies potential use of the model to increase low-dose CTV margins for high-risk patients.
Collapse
Affiliation(s)
- Angela Davey
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Maria Thor
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Marcel van Herk
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Joseph O. Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Alan McWilliam
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
12
|
Vinnat V, Chiche JD, Demoule A, Chevret S. Simulation study for evaluating an adaptive-randomisation Bayesian hybrid trial design with enrichment. Contemp Clin Trials Commun 2023; 33:101141. [PMID: 37397429 PMCID: PMC10313856 DOI: 10.1016/j.conctc.2023.101141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 03/22/2023] [Accepted: 04/12/2023] [Indexed: 07/04/2023] Open
Abstract
Background As we enter the era of precision medicine, the role of adaptive designs, such as response-adaptive randomisation or enrichment designs in drug discovery and development, has become increasingly important to identify the treatment given to a patient based on one or more biomarkers. Tailoring the ventilation supply technique according to the responsiveness of patients to positive end-expiratory pressure is a suitable setting for such a design. Methods In the setting of marker-strategy design, we propose a Bayesian response-adaptive randomisation with enrichment design based on group sequential analyses. This design combines the elements of enrichment design and response-adaptive randomisation. Concerning the enrichment strategy, Bayesian treatment-by-subset interaction measures were used to adaptively enrich the patients most likely to benefit from an experimental treatment while controlling the false-positive rate.The operating characteristics of the design were assessed by simulation and compared to those of alternate designs. Results The results obtained allowed the detection of the superiority of one treatment over another and the presence of a treatment-by-subgroup interaction while keeping the false-positive rate at approximately 5\% and reducing the average number of included patients. In addition, simulation studies identified that the number of interim analyses and the burn-in period may have an impact on the performance of the scheme. Conclusion The proposed design highlights important objectives of precision medicine, such as determining whether the experimental treatment is superior to another and identifying wheter such an efficacy could depend on patient profile.
Collapse
Affiliation(s)
- Valentin Vinnat
- ECSTRRA team, INSERM U1153, Université Paris Cité, Paris, France
| | - Jean-Daniel Chiche
- Service de médecine intensive adulte, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Alexandre Demoule
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie Respiratoire Expérimentale et Clinique, France
- AP-HP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Service de Médecine Intensive et Réanimation (Département R3S), Paris, France
| | - Sylvie Chevret
- ECSTRRA team, INSERM U1153, Université Paris Cité, Paris, France
| |
Collapse
|
13
|
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
|
14
|
Liang F, Peng L, Wu Z, Giamas G, Stebbing J. Design and reporting of phase III oncology trials with prospective biomarker validation. J Natl Cancer Inst 2023; 115:174-180. [PMID: 36448689 PMCID: PMC9905966 DOI: 10.1093/jnci/djac210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/14/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Phase III trials with prospective biomarker validation are essential to drug development in the era of personalized oncology. However, concerns have emerged regarding the design and reporting of phase III trials with prospective biomarker validation. METHODS We searched MEDLINE for phase III oncology trials with prospective biomarker validation published in high-impact medical journals from 2011 to 2020. Information regarding trial design and reporting were extracted. Descriptive methods were used to summarize the results. RESULTS We identified 45 phase III trials with prospective biomarker validation. There was a trend for increasing use of biomarker validation phase III trials (from 1 trial in 2011 to 12 trials in 2020). For 39 (86.7%) trials, results in biomarker-negative population were either listed as an exploratory subgroup analysis (62.2%) or not mentioned in the methods (24.4%). Twenty-one (46.7%) trials were originally designed without biomarker validation but were then apparently modified to incorporate prospective biomarker validation after trial commencement, albeit only 15 (33.3%) trials reported this change. Treatment effect and primary outcome values in biomarker-negative patients were not reported in 24.4% and 40.0% trials, respectively. For 18 trials with statistically significant results in the overall population, only 7 trials reported a hazard ratio less than 0.8 in the biomarker-negative population. CONCLUSIONS Although biomarker validation in phase III trials have been increasingly used in the past decade, issues regarding changes in trial design after commencement without disclosure, underreporting of results in biomarker-negative groups, and recommending treatment in biomarker negative groups despite modest effects require substantial improvement.
Collapse
Affiliation(s)
- Fei Liang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
- Clinical Research Unit, Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhengyu Wu
- Department of Biostatistics, School of Public Health, Key Laboratory of Public Health Safety and Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai, China
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| | - Justin Stebbing
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
- Department of Biomedical Sciences, Anglia Ruskin University, Cambridge, UK
| |
Collapse
|
15
|
Bioinformatics approach to identify the core ontologies, pathways, signature genes and drug molecules of prostate cancer. INFORMATICS IN MEDICINE UNLOCKED 2023. [DOI: 10.1016/j.imu.2023.101179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
16
|
Corbett M, Ramessur R, Marshall D, Acencio ML, Ostaszewski M, Barbosa IA, Dand N, Di Meglio P, Haddad S, Jensen AH, Koopmann W, Mahil SK, Rahmatulla S, Rastrick J, Saklatvala J, Weidinger S, Wright K, Eyerich K, Barker JN, Ndlovu M, Conrad C, Skov L, Smith CH, BIOMAP consortium. Biomarkers of systemic treatment response in people with psoriasis: a scoping review. Br J Dermatol 2022; 187:494-506. [PMID: 35606928 PMCID: PMC9796396 DOI: 10.1111/bjd.21677] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Responses to the systemic treatments commonly used to treat psoriasis vary. Biomarkers that accurately predict effectiveness and safety would enable targeted treatment selection, improved patient outcomes and more cost-effective healthcare. OBJECTIVES To perform a scoping review to identify and catalogue candidate biomarkers of systemic treatment response in psoriasis for the translational research community. METHODS A systematic search of CENTRAL, Embase, LILACS and MEDLINE was performed for relevant articles published between 1990 and December 2021. Eligibility criteria were studies involving patients with psoriasis (any age, n ≥ 50) reporting biomarkers associated with systemic treatment response. The main outcomes were any measure of systemic treatment efficacy or safety. Data were extracted by one reviewer and checked by a second; studies meeting minimal quality criteria (use of methods to control for confounding) were formally assessed for bias. Candidate biomarkers were identified by an expert multistakeholder group using a majority voting consensus exercise and mapped to relevant cellular and molecular pathways. RESULTS Of 71 included studies (67 studying effectiveness outcomes and eight safety outcomes; four studied both), most reported genomic or proteomic biomarkers associated with response to biologics (48 studies). Methodological or reporting limitations frequently compromised the interpretation of findings, including inadequate control for key covariates, lack of adjustment for multiple testing, and selective outcome reporting. We identified candidate biomarkers of efficacy to tumour necrosis factor inhibitors [variation in CARD14, CDKAL1, IL1B, IL12B and IL17RA loci, and lipopolysaccharide-induced phosphorylation of nuclear factor (NF)-κB in type 2 dendritic cells] and ustekinumab (HLA-C*06:02 and variation in an IL1B locus). None were supported by sufficient evidence for clinical use without further validation studies. Candidate biomarkers were found to be involved in the immune cellular crosstalk implicated in psoriasis pathogenesis, most notably antigen presentation, T helper (Th)17 cell differentiation, positive regulation of NF-κB, and Th17 cell activation. CONCLUSIONS This comprehensive catalogue provides a key resource for researchers and reveals a diverse range of biomarker types and outcomes in the included studies. The candidate biomarkers identified require further evaluation in methodologically robust studies to establish potential clinical utility. Future studies should aim to address the common methodological limitations highlighted in this review to expedite discovery and validation of biomarkers for clinical use. What is already known about this topic? Responses to the systemic treatments commonly used to treat psoriasis vary. Biomarkers that accurately predict effectiveness and safety would enable targeted treatment selection, improved patient outcomes and more cost-effective healthcare. What does this study add? This review provides a comprehensive catalogue of investigated biomarkers of systemic treatment response in psoriasis. A diverse range of biomarker types and outcomes was found in the included studies, serving as a key resource for the translational research community.
Collapse
Affiliation(s)
- Mark Corbett
- Centre for Reviews and DisseminationUniversity of YorkYorkUK
| | - Ravi Ramessur
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - David Marshall
- Centre for Reviews and DisseminationUniversity of YorkYorkUK
| | - Marcio L. Acencio
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Marek Ostaszewski
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Ines A. Barbosa
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Nick Dand
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Paola Di Meglio
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | | | | | - Witte Koopmann
- Department of Translational MedicineLEO Pharma A/SBallerupDenmark
| | - Satveer K. Mahil
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | | | - Joe Rastrick
- Department of Immunology ResearchUCBBrusselsBelgium
| | - Jake Saklatvala
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Stephan Weidinger
- Department of Dermatology and AllergyUniversity Hospital Schleswig‐HolsteinKielGermany
| | - Kath Wright
- Centre for Reviews and DisseminationUniversity of YorkYorkUK
| | - Kilian Eyerich
- Department of Dermatology and AllergyTechnical University of MunichMunichGermany
- Division of Dermatology, Department of MedicineKarolinska InstitutetStockholmSweden
| | - Jonathan N. Barker
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | | | - Curdin Conrad
- Department of DermatologyLausanne University Hospital CHUV & University of LausanneLausanneSwitzerland
| | - Lone Skov
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Catherine H. Smith
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | | |
Collapse
|
17
|
Toyoizumi K, Matsui S. Bias correction based on weighted likelihood for conditional estimation of subgroup effects in randomized clinical trials. Stat Med 2022; 41:5276-5289. [PMID: 36055340 DOI: 10.1002/sim.9567] [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: 11/11/2021] [Revised: 07/07/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022]
Abstract
Currently, many confirmatory randomized clinical trials (RCTs) with predictive markers have taken the all-comers approach because of the difficulty in developing predictive markers that are biologically compelling enough to apply the enrichment approach to restrict the patient population to a marker-defined subgroup. However, such a RCT with weak marker credentials can conclude that the new treatment is efficacious only in the subgroup, especially when the primary analysis demonstrates some treatment efficacy in the subgroup, but the overall treatment efficacy is not significant under a control of study-wise alpha rate. In this article, we consider conditional estimation of subgroup treatment effects, given the negative result in testing the overall treatment efficacy in the trial. To address the problem of unstable estimation due to the truncation in the distribution of the test statistic on overall treatment efficacy, we propose a new approach based on a weighted likelihood for the truncated distribution. The weighted likelihood can be derived by invoking a randomized test with a smooth critical function for the overall test. Our approach allows for point and interval estimations of the conditional effects consistently based on the standard maximum likelihood inference. Numerical evaluations, including simulations and application to real clinical trials, and guidelines for implementing our methods with R-codes, are provided.
Collapse
Affiliation(s)
- Kiichiro Toyoizumi
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Statistics & Decision Sciences Department, Janssen Pharmaceutical K. K, Tokyo, Japan
| | - Shigeyuki Matsui
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Data Science, The Institute of Statistical Mathematics, Tokyo, Japan
| |
Collapse
|
18
|
Ho L, Xu Y, Zhang NL, Ho FF, Wu IXY, Chen S, Liu X, Wong CHL, Ching JYL, Cheong PK, Yeung WF, Wu JCY, Chung VCH. Quantification of prevalence, clinical characteristics, co-existence, and geographic variations of traditional Chinese medicine diagnostic patterns via latent tree analysis-based differentiation rules among functional dyspepsia patients. Chin Med 2022; 17:101. [PMID: 36038888 PMCID: PMC9425972 DOI: 10.1186/s13020-022-00656-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Traditional Chinese Medicine (TCM) treatment strategies are guided by pattern differentiation, as documented in the eleventh edition of the International Classification of Diseases (ICD). However, no standards for pattern differentiation are proposed to ensure inter-rater agreement. Without standardisation, research on associations between TCM diagnostic patterns, clinical features, and geographical characteristics is also not feasible. This diagnostic cross-sectional study aimed to (i) establish the pattern differentiation rules of functional dyspepsia (FD) using latent tree analysis (LTA); (ii) compare the prevalence of diagnostic patterns in Hong Kong and Hunan; (iii) discover the co-existence of diagnostic patterns; and (iv) reveal the associations between diagnostic patterns and FD common comorbidities. METHODS A total of 250 and 150 participants with FD consecutively sampled in Hong Kong and Hunan, respectively, completed a questionnaire on TCM clinical features. LTA was performed to reveal TCM diagnostic patterns of FD and derive relevant pattern differentiation rules. Multivariate regression analyses were performed to quantify correlations between different diagnostic patterns and between diagnostic patterns and clinical and geographical variables. RESULTS At least one TCM diagnostic pattern was differentiated in 70.7%, 73.6%, and 64.0% of the participants in the overall (n = 400), Hong Kong (n = 250), and Hunan (n = 150) samples, respectively, using the eight pattern differentiation rules derived. 52.7% to 59.6% of the participants were diagnosed with two or more diagnostic patterns. Cold-heat complex (59.8%) and spleen-stomach dampness-heat (77.1%) were the most prevalent diagnostic patterns in Hong Kong and Hunan, respectively. Spleen-stomach deficiency cold was highly likely to co-exist with spleen-stomach qi deficiency (adjusted odds ratio (AOR): 53.23; 95% confidence interval (CI): 21.77 to 130.16). Participants with severe anxiety tended to have liver qi invading the stomach (AOR: 1.20; 95% CI: 1.08 to 1.33). CONCLUSIONS Future updates of the ICD, textbooks, and guidelines should emphasise the importance of clinical and geographical variations in TCM diagnosis. Location-specific pattern differentiation rules should be derived from local data using LTA. In future, patients' pattern differentiation results, local prevalence of TCM diagnostic patterns, and corresponding TCM treatment choices should be accessible to practitioners on online clinical decision support systems to streamline service delivery.
Collapse
Affiliation(s)
- Leonard Ho
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yulong Xu
- School of Information Technology, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Nevin L Zhang
- Department of Computer Science and Engineering, School of Engineering, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Fai Fai Ho
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Irene X Y Wu
- Xiangya School of Public Health, Central South University, 5/F, 238 Shang-Ma-Yuan-Ling Alley, Kai-Fu District, Changsha, Hunan, China.
| | - Shuijiao Chen
- Department of Gastroenterology, Xiangya Hospital, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer-Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer-Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
| | - Charlene H L Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Jessica Y L Ching
- Institute of Digestive Diseases, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Pui Kuan Cheong
- Institute of Digestive Diseases, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Fai Yeung
- School of Nursing, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Justin C Y Wu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Vincent C H Chung
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
19
|
Iyer V, Yang Z, Ko J, Weissleder R, Issadore D. Advancing microfluidic diagnostic chips into clinical use: a review of current challenges and opportunities. LAB ON A CHIP 2022; 22:3110-3121. [PMID: 35674283 PMCID: PMC9798730 DOI: 10.1039/d2lc00024e] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Microfluidic diagnostic (μDX) technologies miniaturize sensors and actuators to the length-scales that are relevant to biology: the micrometer scale to interact with cells and the nanometer scale to interrogate biology's molecular machinery. This miniaturization allows measurements of biomarkers of disease (cells, nanoscale vesicles, molecules) in clinical samples that are not detectable using conventional technologies. There has been steady progress in the field over the last three decades, and a recent burst of activity catalyzed by the COVID-19 pandemic. In this time, an impressive and ever-growing set of technologies have been successfully validated in their ability to measure biomarkers in clinical samples, such as blood and urine, with sensitivity and specificity not possible using conventional tests. Despite our field's many accomplishments to date, very few of these technologies have been successfully commercialized and brought to clinical use where they can fulfill their promise to improve medical care. In this paper, we identify three major technological trends in our field that we believe will allow the next generation of μDx to have a major impact on the practice of medicine, and which present major opportunities for those entering the field from outside disciplines: 1. the combination of next generation, highly multiplexed μDx technologies with machine learning to allow complex patterns of multiple biomarkers to be decoded to inform clinical decision points, for which conventional biomarkers do not necessarily exist. 2. The use of micro/nano devices to overcome the limits of binding affinity in complex backgrounds in both the detection of sparse soluble proteins and nucleic acids in blood and rare circulating extracellular vesicles. 3. A suite of recent technologies that obviate the manual pre-processing and post-processing of samples before they are measured on a μDX chip. Additionally, we discuss economic and regulatory challenges that have stymied μDx translation to the clinic, and highlight strategies for successfully navigating this challenging space.
Collapse
Affiliation(s)
- Vasant Iyer
- Electrical and Systems Engineering Department, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Zijian Yang
- Mechanical Engineering Department, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jina Ko
- Bioengineering Department, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, 185 Cambridge Street, Boston, Massachusetts, USA
| | - David Issadore
- Electrical and Systems Engineering Department, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
- Bioengineering Department, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
The potential of predictive and prognostic breast MRI (P2-bMRI). Eur Radiol Exp 2022; 6:42. [PMID: 35989400 PMCID: PMC9393116 DOI: 10.1186/s41747-022-00291-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 06/08/2022] [Indexed: 11/10/2022] Open
Abstract
Magnetic resonance imaging (MRI) is an important part of breast cancer diagnosis and multimodal workup. It provides unsurpassed soft tissue contrast to analyse the underlying pathophysiology, and it is adopted for a variety of clinical indications. Predictive and prognostic breast MRI (P2-bMRI) is an emerging application next to these indications. The general objective of P2-bMRI is to provide predictive and/or prognostic biomarkers in order to support personalisation of breast cancer treatment. We believe P2-bMRI has a great clinical potential, thanks to the in vivo examination of the whole tumour and of the surrounding tissue, establishing a link between pathophysiology and response to therapy (prediction) as well as patient outcome (prognostication). The tools used for P2-bMRI cover a wide spectrum: standard and advanced multiparametric pulse sequences; structured reporting criteria (for instance BI-RADS descriptors); artificial intelligence methods, including machine learning (with emphasis on radiomics data analysis); and deep learning that have shown compelling potential for this purpose. P2-bMRI reuses the imaging data of examinations performed in the current practice. Accordingly, P2-bMRI could optimise clinical workflow, enabling cost savings and ultimately improving personalisation of treatment. This review introduces the concept of P2-bMRI, focusing on the clinical application of P2-bMRI by using semantic criteria.
Collapse
|
21
|
Buitelaar J, Bölte S, Brandeis D, Caye A, Christmann N, Cortese S, Coghill D, Faraone SV, Franke B, Gleitz M, Greven CU, Kooij S, Leffa DT, Rommelse N, Newcorn JH, Polanczyk GV, Rohde LA, Simonoff E, Stein M, Vitiello B, Yazgan Y, Roesler M, Doepfner M, Banaschewski T. Toward Precision Medicine in ADHD. Front Behav Neurosci 2022; 16:900981. [PMID: 35874653 PMCID: PMC9299434 DOI: 10.3389/fnbeh.2022.900981] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
Attention-Deficit Hyperactivity Disorder (ADHD) is a complex and heterogeneous neurodevelopmental condition for which curative treatments are lacking. Whilst pharmacological treatments are generally effective and safe, there is considerable inter-individual variability among patients regarding treatment response, required dose, and tolerability. Many of the non-pharmacological treatments, which are preferred to drug-treatment by some patients, either lack efficacy for core symptoms or are associated with small effect sizes. No evidence-based decision tools are currently available to allocate pharmacological or psychosocial treatments based on the patient's clinical, environmental, cognitive, genetic, or biological characteristics. We systematically reviewed potential biomarkers that may help in diagnosing ADHD and/or stratifying ADHD into more homogeneous subgroups and/or predict clinical course, treatment response, and long-term outcome across the lifespan. Most work involved exploratory studies with cognitive, actigraphic and EEG diagnostic markers to predict ADHD, along with relatively few studies exploring markers to subtype ADHD and predict response to treatment. There is a critical need for multisite prospective carefully designed experimentally controlled or observational studies to identify biomarkers that index inter-individual variability and/or predict treatment response.
Collapse
Affiliation(s)
- Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands.,Karakter Child and Adolescent Psychiatry University Center, Nijmegen, Netherlands
| | - Sven Bölte
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.,Child and Adolescent Psychiatry, Stockholm Health Care Services, Stockholm, Sweden.,Curtin Autism Research Group, School of Occupational Therapy, Social Work and Speech Pathology, Curtin University, Perth, WA, Australia
| | - Daniel Brandeis
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany.,Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Arthur Caye
- Department of Psychiatry, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Nina Christmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Samuele Cortese
- Centre for Innovation in Mental Health, Academic Unit of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom.,Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Solent National Health System Trust, Southampton, United Kingdom.,Hassenfeld Children's Hospital at NYU Langone, New York University Child Study Center, New York, NY, United States.,Division of Psychiatry and Applied Psychology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - David Coghill
- Departments of Paediatrics and Psychiatry, Royal Children's Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen V Faraone
- Departments of Psychiatry, Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, NY, United States
| | - Barbara Franke
- Departments of Human Genetics and Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Markus Gleitz
- Medice Arzneimittel Pütter GmbH & Co. KG, Iserlohn, Germany
| | - Corina U Greven
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, Netherlands.,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands.,King's College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| | - Sandra Kooij
- Amsterdam University Medical Center, Location VUMc, Amsterdam, Netherlands.,PsyQ, Expertise Center Adult ADHD, The Hague, Netherlands
| | - Douglas Teixeira Leffa
- Department of Psychiatry, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Nanda Rommelse
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, Netherlands.,Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jeffrey H Newcorn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Guilherme V Polanczyk
- Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Luis Augusto Rohde
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil.,ADHD Outpatient Program and Developmental Psychiatry Program, Hospital de Clinica de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Emily Simonoff
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - Mark Stein
- Department of Psychiatry and Behavioral Sciences, Seattle, WA, United States
| | - Benedetto Vitiello
- Department of Public Health and Pediatric Sciences, Section of Child and Adolescent Neuropsychiatry, University of Turin, Turin, Italy.,Department of Public Health, Johns Hopkins University, Baltimore, MA, United States
| | - Yanki Yazgan
- GuzelGunler Clinic, Istanbul, Turkey.,Yale Child Study Center, New Haven, CT, United States
| | - Michael Roesler
- Institute for Forensic Psychology and Psychiatry, Neurocenter, Saarland, Germany
| | - Manfred Doepfner
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Faculty of the University of Cologne, Cologne, Germany.,School for Child and Adolescent Cognitive Behavioural Therapy, University Hospital of Cologne, Cologne, Germany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| |
Collapse
|
22
|
Cho D, Cheyne S, Lord SJ, Simes J, Lee CK. Extrapolating evidence for molecularly targeted therapies from common to rare cancers: a scoping review of methodological guidance. BMJ Open 2022; 12:e058350. [PMID: 35820758 PMCID: PMC9274540 DOI: 10.1136/bmjopen-2021-058350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 06/23/2022] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES Cancer is increasingly classified according to biomarkers that drive tumour growth and therapies developed to target them. In rare biomarker-defined cancers, randomised controlled trials to adequately assess targeted therapies may be infeasible. Extrapolating existing evidence of targeted therapy from common cancers to rare cancers sharing the same biomarker may reduce evidence requirements for regulatory approval in rare cancers. It is unclear whether guidelines exist for extrapolation. We sought to identify methodological guidance for extrapolating evidence from targeted therapies used for common cancers to rare biomarker-defined cancers. DESIGN Scoping review. DATA SOURCES Websites of health technology assessment agencies, regulatory bodies, research groups, scientific societies and industry. EBM Reviews-Cochrane Methodology Register and Health Technology Assessment, Embase and MEDLINE databases (1946 to 11 May 2022). ELIGIBILITY CRITERIA Papers proposing a framework or recommendations for extrapolating evidence for rare cancers, small populations and biomarker-defined cancers. DATA EXTRACTION AND SYNTHESIS We extracted framework details where available and guidance for components of extrapolation. We used these components to structure and summarise recommendations. RESULTS We identified 23 papers. One paper provided an extrapolation framework but was not cancer specific. Extrapolation recommendations addressed six distinct components: strategies for grouping cancers as the same biomarker-defined disease; analytical validation requirements of a biomarker test to use across cancer types; strategies to generate control data when a randomised concurrent control arm is infeasible; sources to inform biomarker clinical utility assessment in the absence of prospective clinical evidence; requirements for surrogate endpoints chosen for the rare cancer; and assessing and augmenting safety data in the rare cancer. CONCLUSIONS In the absence of an established framework, our recommendations for components of extrapolation can be used to guide discussions about interpreting evidence to support extrapolation. The review can inform the development of an extrapolation framework for biomarker-targeted therapies in rare cancers.
Collapse
Affiliation(s)
- Doah Cho
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Saskia Cheyne
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Sarah J Lord
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- School of Medicine, The University of Notre Dame Australia, Sydney Campus, Darlinghurst, New South Wales, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Chee Khoon Lee
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- Cancer Care Centre, St George Hospital, Kogarah, New South Wales, Australia
| |
Collapse
|
23
|
Rousseau B, Bieche I, Pasmant E, Hamzaoui N, Leulliot N, Michon L, de Reynies A, Attignon V, Foote MB, Masliah-Planchon J, Svrcek M, Cohen R, Simmet V, Augereau P, Malka D, Hollebecque A, Pouessel D, Gomez-Roca C, Guimbaud R, Bruyas A, Guillet M, Grob JJ, Duluc M, Cousin S, de la Fouchardiere C, Flechon A, Rolland F, Hiret S, Saada-Bouzid E, Bouche O, Andre T, Pannier D, El Hajbi F, Oudard S, Tournigand C, Soria JC, Champiat S, Gerber DG, Stephens D, Lamendola-Essel MF, Maron SB, Diplas BH, Argiles G, Krishnan AR, Tabone-Eglinger S, Ferrari A, Segal NH, Cercek A, Hoog-Labouret N, Legrand F, Simon C, Lamrani-Ghaouti A, Diaz LA, Saintigny P, Chevret S, Marabelle A. PD-1 Blockade in Solid Tumors with Defects in Polymerase Epsilon. Cancer Discov 2022; 12:1435-1448. [PMID: 35398880 PMCID: PMC9167784 DOI: 10.1158/2159-8290.cd-21-0521] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 03/09/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022]
Abstract
Missense mutations in the polymerase epsilon (POLE) gene have been reported to generate proofreading defects resulting in an ultramutated genome and to sensitize tumors to checkpoint blockade immunotherapy. However, many POLE-mutated tumors do not respond to such treatment. To better understand the link between POLE mutation variants and response to immunotherapy, we prospectively assessed the efficacy of nivolumab in a multicenter clinical trial in patients bearing advanced mismatch repair-proficient POLE-mutated solid tumors. We found that only tumors harboring selective POLE pathogenic mutations in the DNA binding or catalytic site of the exonuclease domain presented high mutational burden with a specific single-base substitution signature, high T-cell infiltrates, and a high response rate to anti-PD-1 monotherapy. This study illustrates how specific DNA repair defects sensitize to immunotherapy. POLE proofreading deficiency represents a novel agnostic biomarker for response to PD-1 checkpoint blockade therapy. SIGNIFICANCE POLE proofreading deficiency leads to high tumor mutational burden with high tumor-infiltrating lymphocytes and predicts anti-PD-1 efficacy in mismatch repair-proficient tumors. Conversely, tumors harboring POLE mutations not affecting proofreading derived no benefit from PD-1 blockade. POLE proofreading deficiency is a new tissue-agnostic biomarker for cancer immunotherapy. This article is highlighted in the In This Issue feature, p. 1397.
Collapse
Affiliation(s)
- Benoit Rousseau
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivan Bieche
- Department of Genetics, Institut Curie, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
| | - Eric Pasmant
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, AP-HP.Centre-Université de Paris, Paris, France
| | - Nadim Hamzaoui
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
- Fédération de Génétique et Médecine Génomique, Hôpital Cochin, AP-HP.Centre-Université de Paris, Paris, France
| | - Nicolas Leulliot
- Cibles Thérapeutiques et Conception de Médicaments, CNRS UMR8015, Université de Paris, UFR de Pharmacie de Paris, Paris, France
| | - Lucas Michon
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Aurelien de Reynies
- Université de Paris, Centre de Recherche des Cordeliers, UMRS1138, AP-HP, SeqOIA-IT, Paris, France
| | | | - Michael B. Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Magali Svrcek
- Pathology department, Saint Antoine Hospital
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, F-75012 Paris, France
| | - Romain Cohen
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, F-75012 Paris, France
- Medical Oncology Department, Hôpital Saint-Antoine, Paris, France
| | - Victor Simmet
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest (ICO), Angers, France
| | - Paule Augereau
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest (ICO), Angers, France
| | - David Malka
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Antoine Hollebecque
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Damien Pouessel
- Department of Medical Oncology, Institut Claudius Regaud / IUCT Oncopole, Toulouse, France
| | - Carlos Gomez-Roca
- Department of Medical Oncology, Institut Claudius Regaud / IUCT Oncopole, Toulouse, France
| | | | - Amandine Bruyas
- Department of Medical Oncology, Hôpital de la Croix-Rousse, Lyon, France
| | - Marielle Guillet
- Department of Gastroenterology and Digestive Oncology, Hôpital de la Croix-Rousse, Lyon, France
| | | | - Muriel Duluc
- Dermatology and Oncology, Hôpital de la Timone, Marseille, France
| | | | | | - Aude Flechon
- Department of medical Oncology, Centre Leon Berard, Lyon, France
| | - Frederic Rolland
- Department of Medical Oncology, ICO Institut de Cancerologie de l’Ouest René Gauducheau, Saint-Herblain, France
| | - Sandrine Hiret
- Department of Medical Oncology, ICO Institut de Cancerologie de l’Ouest René Gauducheau, Saint-Herblain, France
| | - Esma Saada-Bouzid
- Medical Oncology, Centre Anticancer Antoine Lacassagne, Nice, France
| | - Olivier Bouche
- Gastroenterology and Digestive Oncology, CHU de Reims - Hôpital Robert Debré, Reims, France
| | - Thierry Andre
- Medical Oncology Department, Hôpital Saint-Antoine, Paris, France
| | | | | | - Stephane Oudard
- Oncology, Hopital Europeen Georges Pompidou, AP-HP, Paris, France
| | | | - Jean-Charles Soria
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Stephane Champiat
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Drew G. Gerber
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dennis Stephens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Steven B. Maron
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bill H. Diplas
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Guillem Argiles
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Asha R. Krishnan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Anthony Ferrari
- Platform of Bioinformatics Gilles Thomas-Synergie Lyon Cancer, Centre Léon Bérard, Lyon
| | - Neil H. Segal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Frederic Legrand
- Research and Innovation, Institut National du Cancer, Boulogne-Billancourt, France
| | | | | | - Luis A. Diaz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pierre Saintigny
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of medical Oncology, Centre Leon Berard, Lyon, France
| | | | - Aurelien Marabelle
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
- U1015 & CIC1428, Institut national de la santé et de la recherche médicale (INSERM), Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicetre, France
| |
Collapse
|
24
|
Hertz DL, McShane LM, Hayes DF. Defining Clinical Utility of Germline Indicators of Toxicity Risk: A Perspective. J Clin Oncol 2022; 40:1721-1731. [PMID: 35324346 PMCID: PMC9148690 DOI: 10.1200/jco.21.02209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI
| | - Lisa M McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Daniel F Hayes
- Stuart B. Padnos Professor of Breast Cancer Research, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| |
Collapse
|
25
|
Buccisano F, Palmieri R, Piciocchi A, Arena V, Maurillo L, Del Principe MI, Paterno G, Irno-Consalvo MA, Ottone T, Divona M, Conti C, Fraboni D, Lavorgna S, Arcese W, Voso MT, Venditti A. Clinical relevance of an objective flow cytometry approach based on limit of detection and limit of quantification for measurable residual disease assessment in acute myeloid leukemia. A post-hoc analysis of the GIMEMA AML1310 trial. Haematologica 2022; 107:2823-2833. [PMID: 35295076 PMCID: PMC9713557 DOI: 10.3324/haematol.2021.279777] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Using a multiparametric flow cytometry assay, we assessed the predictive power of a threshold calculated applying the criteria of limit of detection (LOD) and limit of quantitation (LOQ) in adult patients with acute myeloid leukemia. This was a post-hoc analysis of 261 patients enrolled in the GIMEMA AML1310 prospective trial. According to the protocol design, using the predefined measurable residual disease (MRD) threshold of 0.035% bone marrow residual leukemic cells (RLC) calculated on mononuclear cells, 154 (59%) of the 261 patients were negative (MRD <0.035%) and 107 (41%) were positive (MRD ≥0.035%). Using LOD and LOQ, we selected the following categories of patients: (i) LODneg if RLC were below the LOD (74; 28.4%); (ii) LODpos-LOQneg if RLC were between the LOD and LOQ (43; 16.5%); and (iii) LOQpos if RLC were above the LOQ (144; 54.4%). Two-year overall survival of these three categories of patients was 75.4%, 79.8% and 66.4%, respectively (P=0.1197). Given their superimposable outcomes, the LODneg and LODpos-LOQneg categories were combined. Two-year overall survival of LODneg/LODpos-LOQneg patients was 77.0% versus 66.4% of LOQpos individuals (P=0.043). This figure was challenged in univariate analysis (P=0.046, hazard ratio=1.6, 95% confidence interval: 1.01-2.54) which confirmed the independent role of the LOD-LOQ approach in determining overall survival. In the AML1310 protocol, using the threshold of 0.035%, 2-year overall survival of patients with MRD <0.035% and MRD ≥0.035% was 74.5% versus 66.4%, respectively (P=0.3521). In conclusion, the use of the LOD-LOQ method results in more sensitive detection of MRD that, in turn, translates into a more accurate recognition of patients with different outcomes.
Collapse
Affiliation(s)
- Francesco Buccisano
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,FB and RP contributed equally as co-first authors
| | - Raffaele Palmieri
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,FB and RP contributed equally as co-first authors
| | | | | | - Luca Maurillo
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | | | | | | | - Tiziana Ottone
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Mariadomenica Divona
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Consuelo Conti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Daniela Fraboni
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Serena Lavorgna
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - William Arcese
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,Rome Transplant Network, Rome, Italy
| | - Maria Teresa Voso
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Adriano Venditti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| |
Collapse
|
26
|
Park Y. Personalized Risk-Based Screening Design for Comparative Two-Arm Group Sequential Clinical Trials. J Pers Med 2022; 12:448. [PMID: 35330448 PMCID: PMC8953575 DOI: 10.3390/jpm12030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Personalized medicine has been emerging to take into account individual variability in genes and environment. In the era of personalized medicine, it is critical to incorporate the patients' characteristics and improve the clinical benefit for patients. The patients' characteristics are incorporated in adaptive randomization to identify patients who are expected to get more benefit from the treatment and optimize the treatment allocation. However, it is challenging to control potential selection bias from using observed efficacy data and the effect of prognostic covariates in adaptive randomization. This paper proposes a personalized risk-based screening design using Bayesian covariate-adjusted response-adaptive randomization that compares the experimental screening method to a standard screening method based on indicators of having a disease. Personalized risk-based allocation probability is built for adaptive randomization, and Bayesian adaptive decision rules are calibrated to preserve error rates. A simulation study shows that the proposed design controls error rates and yields a much smaller number of failures and a larger number of patients allocated to a better intervention compared to existing randomized controlled trial designs. Therefore, the proposed design performs well for randomized controlled clinical trials under personalized medicine.
Collapse
Affiliation(s)
- Yeonhee Park
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
27
|
Determination of molecular signatures and pathways common to brain tissues of autism spectrum disorder: Insights from comprehensive bioinformatics approach. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
28
|
Kammer MN, Lakhani DA, Balar AB, Antic SL, Kussrow AK, Webster RL, Mahapatra S, Barad U, Shah C, Atwater T, Diergaarde B, Qian J, Kaizer A, New M, Hirsch E, Feser WJ, Strong J, Rioth M, Miller YE, Balagurunathan Y, Rowe DJ, Helmey S, Chen SC, Bauza J, Deppen SA, Sandler K, Maldonado F, Spira A, Billatos E, Schabath MB, Gillies RJ, Wilson DO, Walker RC, Landman B, Chen H, Grogan EL, Barón AE, Bornhop DJ, Massion PP. Integrated Biomarkers for the Management of Indeterminate Pulmonary Nodules. Am J Respir Crit Care Med 2021; 204:1306-1316. [PMID: 34464235 PMCID: PMC8786067 DOI: 10.1164/rccm.202012-4438oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 08/27/2021] [Indexed: 01/06/2023] Open
Abstract
Rationale: Patients with indeterminate pulmonary nodules (IPNs) at risk of cancer undergo high rates of invasive, costly, and morbid procedures. Objectives: To train and externally validate a risk prediction model that combined clinical, blood, and imaging biomarkers to improve the noninvasive management of IPNs. Methods: In this prospectively collected, retrospective blinded evaluation study, probability of cancer was calculated for 456 patient nodules using the Mayo Clinic model, and patients were categorized into low-, intermediate-, and high-risk groups. A combined biomarker model (CBM) including clinical variables, serum high sensitivity CYFRA 21-1 level, and a radiomic signature was trained in cohort 1 (n = 170) and validated in cohorts 2-4 (total n = 286). All patients were pooled to recalibrate the model for clinical implementation. The clinical utility of the CBM compared with current clinical care was evaluated in 2 cohorts. Measurements and Main Results: The CBM provided improved diagnostic accuracy over the Mayo Clinic model with an improvement in area under the curve of 0.124 (95% bootstrap confidence interval, 0.091-0.156; P < 2 × 10-16). Applying 10% and 70% risk thresholds resulted in a bias-corrected clinical reclassification index for cases and control subjects of 0.15 and 0.12, respectively. A clinical utility analysis of patient medical records estimated that a CBM-guided strategy would have reduced invasive procedures from 62.9% to 50.6% in the intermediate-risk benign population and shortened the median time to diagnosis of cancer from 60 to 21 days in intermediate-risk cancers. Conclusions: Integration of clinical, blood, and image biomarkers improves noninvasive diagnosis of patients with IPNs, potentially reducing the rate of unnecessary invasive procedures while shortening the time to diagnosis.
Collapse
Affiliation(s)
- Michael N. Kammer
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Chemistry, and
| | - Dhairya A. Lakhani
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Aneri B. Balar
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Sanja L. Antic
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Amanda K. Kussrow
- Department of Chemistry, and
- Vanderbilt Institute for Chemical Biology, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | | | - Shayan Mahapatra
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | | | | | - Thomas Atwater
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Brenda Diergaarde
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Jun Qian
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Alexander Kaizer
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Erin Hirsch
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - William J. Feser
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jolene Strong
- Biomedical Informatics and Personalized Medicine, and
| | - Matthew Rioth
- Medical Oncology and Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| | | | | | - Dianna J. Rowe
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Sherif Helmey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph Bauza
- American College of Radiology, Philadelphia, Pennsylvania
| | - Stephen A. Deppen
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kim Sandler
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Fabien Maldonado
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Avrum Spira
- Department of Medicine, Boston University, Boston, Massachusetts
| | - Ehab Billatos
- Department of Medicine, Boston University, Boston, Massachusetts
| | | | | | - David O. Wilson
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | | | - Bennett Landman
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Heidi Chen
- American College of Radiology, Philadelphia, Pennsylvania
| | - Eric L. Grogan
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Anna E. Barón
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Darryl J. Bornhop
- Department of Chemistry, and
- Vanderbilt Institute for Chemical Biology, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Pierre P. Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee
- Pulmonary Section, Medical Service, Tennessee Valley Healthcare Systems Nashville Campus, Nashville, Tennessee
| |
Collapse
|
29
|
Scherz V, Greub G, Bertelli C. Building up a clinical microbiota profiling: a quality framework proposal. Crit Rev Microbiol 2021; 48:356-375. [PMID: 34752719 DOI: 10.1080/1040841x.2021.1975642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Extensive characterization of the human microbiota has revealed promising relationships between microbial composition and health or disease, generating interest in biomarkers derived from microbiota profiling. However, microbiota complexity and technical challenges strongly influencing the results limit the generalization of microbiota profiling and question its clinical utility. In addition, no quality management scheme has been adapted to the specificities of microbiota profiling, notably due to the heterogeneity in methods and results. In this review, we discuss possible adaptation of classical quality management tools routinely used in diagnostic laboratories to microbiota profiling and propose a specific framework. Multiple quality controls are needed to cover all steps, from sampling to data processing. Standard operating procedures, primarily developed for wet lab analyses, must be adapted to the use of bioinformatic tools. Finally, requirements for test validation and proficiency testing must take into account expected discrepancies in results due to the heterogeneity of the processes. The proposed quality management framework should support the implementation of routine microbiota profiling by clinical laboratories to support patient care. Furthermore, its use in research laboratories would improve publication reproducibility as well as transferability of methods and results to routine practice.
Collapse
Affiliation(s)
- Valentin Scherz
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gilbert Greub
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Claire Bertelli
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
Rahman R, Ventz S, McDunn J, Louv B, Reyes-Rivera I, Polley MYC, Merchant F, Abrey LE, Allen JE, Aguilar LK, Aguilar-Cordova E, Arons D, Tanner K, Bagley S, Khasraw M, Cloughesy T, Wen PY, Alexander BM, Trippa L. Leveraging external data in the design and analysis of clinical trials in neuro-oncology. Lancet Oncol 2021; 22:e456-e465. [PMID: 34592195 PMCID: PMC8893120 DOI: 10.1016/s1470-2045(21)00488-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 01/20/2023]
Abstract
Integration of external control data, with patient-level information, in clinical trials has the potential to accelerate the development of new treatments in neuro-oncology by contextualising single-arm studies and improving decision making (eg, early stopping decisions). Based on a series of presentations at the 2020 Clinical Trials Think Tank hosted by the Society of Neuro-Oncology, we provide an overview on the use of external control data representative of the standard of care in the design and analysis of clinical trials. High-quality patient-level records, rigorous methods, and validation analyses are necessary to effectively leverage external data. We review study designs, statistical methods, risks, and potential distortions in using external data from completed trials and real-world data, as well as data sources, data sharing models, ongoing work, and applications in glioblastoma.
Collapse
Affiliation(s)
- Rifaquat Rahman
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA.
| | - Steffen Ventz
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Jon McDunn
- Project Data Sphere, Morrisville, NC, USA
| | - Bill Louv
- Project Data Sphere, Morrisville, NC, USA
| | | | - Mei-Yin C Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | - David Arons
- National Brain Tumor Society, Newton, MA, USA
| | - Kirk Tanner
- National Brain Tumor Society, Newton, MA, USA
| | - Stephen Bagley
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mustafa Khasraw
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| | - Timothy Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA; Foundation Medicine, Cambridge, MA, USA
| | - Lorenzo Trippa
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T H Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
31
|
Lin Z, Flournoy N, Rosenberger WF. Inference for a two-stage enrichment design. Ann Stat 2021. [DOI: 10.1214/21-aos2051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Zhantao Lin
- Department of Statistics, George Mason University
| | - Nancy Flournoy
- Department of Statistics, University of Missouri, Columbia
| | | |
Collapse
|
32
|
Toward the Next Generation of High-Grade Glioma Clinical Trials in the Era of Precision Medicine. Cancer J 2021; 27:410-415. [PMID: 34570456 DOI: 10.1097/ppo.0000000000000549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT In the era of precision medicine, there is a desire to harness our improved understanding of genomic and molecular underpinnings of gliomas to develop therapies that can be tailored to individual patients and tumors. With the rapid development of novel therapies, there has been a growing need to develop smart clinical trials that are designed to efficiently test promising agents, identify therapies likely to benefit patients, and discard ineffective therapies. We review clinical trial design in gliomas and developments designed to address the unique challenges of precision medicine. To provide an overview of this topic, we examine considerations for endpoints and response assessment, biomarkers, and novel clinical trial designs such as adaptive platform trials in the testing of new therapies for glioma patients.
Collapse
|
33
|
Gray M, Marland JRK, Murray AF, Argyle DJ, Potter MA. Predictive and Diagnostic Biomarkers of Anastomotic Leakage: A Precision Medicine Approach for Colorectal Cancer Patients. J Pers Med 2021; 11:471. [PMID: 34070593 PMCID: PMC8229046 DOI: 10.3390/jpm11060471] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Development of an anastomotic leak (AL) following intestinal surgery for the treatment of colorectal cancers is a life-threatening complication. Failure of the anastomosis to heal correctly can lead to contamination of the abdomen with intestinal contents and the development of peritonitis. The additional care that these patients require is associated with longer hospitalisation stays and increased economic costs. Patients also have higher morbidity and mortality rates and poorer oncological prognosis. Unfortunately, current practices for AL diagnosis are non-specific, which may delay diagnosis and have a negative impact on patient outcome. To overcome these issues, research is continuing to identify AL diagnostic or predictive biomarkers. In this review, we highlight promising candidate biomarkers including ischaemic metabolites, inflammatory markers and bacteria. Although research has focused on the use of blood or peritoneal fluid samples, we describe the use of implantable medical devices that have been designed to measure biomarkers in peri-anastomotic tissue. Biomarkers that can be used in conjunction with clinical status, routine haematological and biochemical analysis and imaging have the potential to help to deliver a precision medicine package that could significantly enhance a patient's post-operative care and improve outcomes. Although no AL biomarker has yet been validated in large-scale clinical trials, there is confidence that personalised medicine, through biomarker analysis, could be realised for colorectal cancer intestinal resection and anastomosis patients in the years to come.
Collapse
Affiliation(s)
- Mark Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, UK;
| | - Jamie R. K. Marland
- School of Engineering, Institute for Integrated Micro and Nano Systems, University of Edinburgh, Scottish Microelectronics Centre, King’s Buildings, Edinburgh EH9 3FF, UK;
| | - Alan F. Murray
- School of Engineering, Institute for Bioengineering, University of Edinburgh, Faraday Building, The King’s Buildings, Edinburgh EH9 3DW, UK;
| | - David J. Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, UK;
| | - Mark A. Potter
- Department of Surgery, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK;
| |
Collapse
|
34
|
Jung SH. Design and Analysis of Cancer Clinical Trials for Personalized Medicine. J Pers Med 2021; 11:jpm11050376. [PMID: 34064394 PMCID: PMC8147797 DOI: 10.3390/jpm11050376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/31/2022] Open
Abstract
Biomarkers play a key role in the development of personalized medicine. Cancer clinical trials with biomarker should be appropriately designed and analyzed reflecting the various factors, such as the phase of trials, the type of biomarker, the study objectives, and whether the used biomarker is already validated or not. In this paper, we demonstrate design and analysis of two phase II cancer clinical trials, one with a predictive biomarker and the other with a prognostic biomarker. A statistical testing method and its sample size calculation method are presented for each of the trials. We assume that the primary endpoint of these trials is a time to event variable, but this concept can be used for any type of endpoint with associated testing methods. The test statistics and their sample size formulas are derived using the large sample approximation based on the martingale central limit theorem. Using simulations, we find that the test statistics control the type I error rate accurately and the sample sizes calculated using the formulas maintain the statistical power specified at the design stage.
Collapse
Affiliation(s)
- Sin-Ho Jung
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710, USA
| |
Collapse
|
35
|
Ballarini NM, Burnett T, Jaki T, Jennison C, König F, Posch M. Optimizing subgroup selection in two-stage adaptive enrichment and umbrella designs. Stat Med 2021; 40:2939-2956. [PMID: 33783020 PMCID: PMC8251960 DOI: 10.1002/sim.8949] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 01/11/2021] [Accepted: 02/28/2021] [Indexed: 12/11/2022]
Abstract
We design two‐stage confirmatory clinical trials that use adaptation to find the subgroup of patients who will benefit from a new treatment, testing for a treatment effect in each of two disjoint subgroups. Our proposal allows aspects of the trial, such as recruitment probabilities of each group, to be altered at an interim analysis. We use the conditional error rate approach to implement these adaptations with protection of overall error rates. Applying a Bayesian decision‐theoretic framework, we optimize design parameters by maximizing a utility function that takes the population prevalence of the subgroups into account. We show results for traditional trials with familywise error rate control (using a closed testing procedure) as well as for umbrella trials in which only the per‐comparison type 1 error rate is controlled. We present numerical examples to illustrate the optimization process and the effectiveness of the proposed designs.
Collapse
Affiliation(s)
- Nicolás M Ballarini
- Section for Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Thomas Burnett
- Department of Mathematics and Statistics, Lancaster University, Lancaster, UK
| | - Thomas Jaki
- Department of Mathematics and Statistics, Lancaster University, Lancaster, UK.,MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | | | - Franz König
- Section for Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Martin Posch
- Section for Medical Statistics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Thiruthaneeswaran N, Bibby BAS, Yang L, Hoskin PJ, Bristow RG, Choudhury A, West C. Lost in application: Measuring hypoxia for radiotherapy optimisation. Eur J Cancer 2021; 148:260-276. [PMID: 33756422 DOI: 10.1016/j.ejca.2021.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
The history of radiotherapy is intertwined with research on hypoxia. There is level 1a evidence that giving hypoxia-targeting treatments with radiotherapy improves locoregional control and survival without compromising late side-effects. Despite coming in and out of vogue over decades, there is now an established role for hypoxia in driving molecular alterations promoting tumour progression and metastases. While tumour genomic complexity and immune profiling offer promise, there is a stronger evidence base for personalising radiotherapy based on hypoxia status. Despite this, there is only one phase III trial targeting hypoxia modification with full transcriptomic data available. There are no biomarkers in routine use for patients undergoing radiotherapy to aid management decisions, and a roadmap is needed to ensure consistency and provide a benchmark for progression to application. Gene expression signatures address past limitations of hypoxia biomarkers and could progress biologically optimised radiotherapy. Here, we review recent developments in generating hypoxia gene expression signatures and highlight progress addressing the challenges that must be overcome to pave the way for their clinical application.
Collapse
Affiliation(s)
- Niluja Thiruthaneeswaran
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Becky A S Bibby
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Lingjang Yang
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
37
|
Wang X, Piantadosi S, Le-Rademacher J, Mandrekar SJ. Statistical Considerations for Subgroup Analyses. J Thorac Oncol 2021; 16:375-380. [PMID: 33373692 PMCID: PMC7920926 DOI: 10.1016/j.jtho.2020.12.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/08/2020] [Accepted: 12/12/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Xiaofei Wang
- Department of Biostatistics & Bioinformatics, Duke University, Durham, North Carolina.
| | - Steven Piantadosi
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | | | | |
Collapse
|
38
|
Riechelmann H, Steinbichler TB, Sprung S, Santer M, Runge A, Ganswindt U, Gamerith G, Dudas J. The Epithelial-Mesenchymal Transcription Factor Slug Predicts Survival Benefit of Up-Front Surgery in Head and Neck Cancer. Cancers (Basel) 2021; 13:cancers13040772. [PMID: 33673269 PMCID: PMC7918715 DOI: 10.3390/cancers13040772] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/06/2021] [Accepted: 02/09/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary In preclinical studies, the epithelial-to-mesenchymal transition (EMT)-related transcription factor Slug indicated radio- and chemoresistance in head and neck squamous cell carcinoma (HNSCC). Here we show that Slug is a biomarker associated with treatment failure in HNSCC patients treated with primary radio- or radiochemotherapy, but not in patients undergoing upfront surgery and postoperative radio- or chemoradiotherapy. Slug may thus serve as a predictive biomarker to identify HNSCC patients who will benefit from upfront surgery. Slug status is an immunohistochemical (IHC) parameter that is easy to determine. If the predictive value observed here can be confirmed in validation studies with independent data, Slug immunohistochemistry may have significant clinical relevance in treatment planning for HNSCC patients. Abstract EMT promotes radio- and chemotherapy resistance in HNSCC in vitro. As EMT has been correlated to the transcription factor Slug in tumor specimens from HNSCC patients, we assessed whether Slug overexpression predicts radio- and chemotherapy resistance and favors upfront surgery in HNSCC patients. Slug expression was determined by IHC scoring in tumor specimens from patients with incident HNSCC. Patients were treated with either definitive radiotherapy or chemoradiotherapy (primary RT/CRT) or upfront surgery with or without postoperative RT or CRT (upfront surgery/PORT). Treatment failure rates and overall survival (OS) were compared between RT/CRT and upfront surgery/PORT in Slug-positive and Slug-negative patients. Slug IHC was positive in 91/354 HNSCC patients. Primary RT/CRT showed inferior response rates (univariate odds ratio (OR) for treatment failure, 3.6; 95% CI, 1.7 to 7.9; p = 0.001) and inferior 5-year OS (univariate, p < 0.001) in Slug-positive patients. The independent predictive value of Slug expression status was confirmed in a multivariable Cox model (p = 0.017). Slug-positive patients had a 3.3 times better chance of survival when treated with upfront surgery/PORT versus primary RT/CRT. For HNSCC patients, Slug IHC represents a novel and feasible predictive biomarker to support upfront surgery.
Collapse
Affiliation(s)
- Herbert Riechelmann
- Department for Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.R.); (M.S.); (A.R.); (J.D.)
| | - Teresa Bernadette Steinbichler
- Department for Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.R.); (M.S.); (A.R.); (J.D.)
- Correspondence:
| | - Susanne Sprung
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Matthias Santer
- Department for Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.R.); (M.S.); (A.R.); (J.D.)
| | - Annette Runge
- Department for Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.R.); (M.S.); (A.R.); (J.D.)
| | - Ute Ganswindt
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Gabriele Gamerith
- Department of Hematology and Oncology, Medical University Innsbruck, 6020 Innsbruck, Austria;
| | - Jozsef Dudas
- Department for Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.R.); (M.S.); (A.R.); (J.D.)
| |
Collapse
|
39
|
Llovet JM, Villanueva A, Marrero JA, Schwartz M, Meyer T, Galle PR, Lencioni R, Greten TF, Kudo M, Mandrekar SJ, Zhu AX, Finn RS, Roberts LR. Trial Design and Endpoints in Hepatocellular Carcinoma: AASLD Consensus Conference. Hepatology 2021; 73 Suppl 1:158-191. [PMID: 32430997 DOI: 10.1002/hep.31327] [Citation(s) in RCA: 269] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Josep M Llovet
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.,Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institució Catalana d'Estudis Avançats (ICREA), Barcelona, Spain
| | - Augusto Villanueva
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Myron Schwartz
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tim Meyer
- Department Oncology, University College London Cancer Institute, London, UK
| | - Peter R Galle
- Department of Internal Medicine, Mainz University Medical Center, Mainz, Germany
| | - Riccardo Lencioni
- Department of Radiology, University of Pisa School of Medicine, Pisa, Italy.,Miami Cancer Institute, Miami, FL
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | | | - Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA.,Jiahui International Cancer Center, Shanghai, China
| | | | - Lewis R Roberts
- Gastroenterology & Hepatology Department, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
40
|
Is It Possible to Personalize the Diagnosis and Treatment of Breast Cancer during Pregnancy? J Pers Med 2020; 11:jpm11010018. [PMID: 33379383 PMCID: PMC7823967 DOI: 10.3390/jpm11010018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022] Open
Abstract
The main goal of precision medicine in patients with breast cancer is to tailor the treatment according to the particular genetic makeup and the genetic changes in the cancer cells. Breast cancer occurring during pregnancy (BCP) is a complex and difficult clinical problem. Although it is not very common, both maternal and fetal outcome must be always considered when planning treatment. Pregnancy represents a significant barrier to the implementation of personalized treatment for breast cancer. Tailoring therapy mainly takes into account the stage of pregnancy, the subtype of cancer, the stage of cancer, and the patient’s preference. Results of the treatment of breast cancer in pregnancy are as yet not very satisfactory because of often delayed diagnosis, and it usually has an unfavorable outcome. Treatment of patients with pregnancy-associated breast cancer should be centralized. Centralization may result in increased experience in diagnosis and treatment and accumulated data may help us to optimize the treatment approaches, modify general treatment recommendations, and improve the survival and quality of life of the patients.
Collapse
|
41
|
Lin R, Yang Z, Yuan Y, Yin G. Sample size re-estimation in adaptive enrichment design. Contemp Clin Trials 2020; 100:106216. [PMID: 33246098 DOI: 10.1016/j.cct.2020.106216] [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: 07/06/2020] [Revised: 10/23/2020] [Accepted: 11/10/2020] [Indexed: 10/22/2022]
Abstract
Clinical trial participants are often heterogeneous, which is a fundamental problem in the rapidly developing field of precision medicine. Participants heterogeneity causes considerable difficulty in the current phase III trial designs. Adaptive enrichment designs provide a flexible and intuitive solution. At the interim analysis, we enrich the subgroup of trial participants who have a higher likelihood to benefit from the new treatment. However, it is critical to control the level of the test size and maintain adequate power after enrichment of certain subgroup of participants. We develop two adaptive enrichment strategies with sample size re-estimation and verify their feasibility and practicability through extensive simulations and sensitivity analyses. The simulation studies show that the proposed methods can control the overall type I error rate and exhibit competitive improvement in terms of statistical power and expected sample size. The proposed designs are exemplified with a real trial application.
Collapse
Affiliation(s)
- Ruitao Lin
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhao Yang
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Ying Yuan
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| |
Collapse
|
42
|
Nelson PG, Promislow DEL, Masel J. Biomarkers for Aging Identified in Cross-sectional Studies Tend to Be Non-causative. J Gerontol A Biol Sci Med Sci 2020; 75:466-472. [PMID: 31353411 DOI: 10.1093/gerona/glz174] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Indexed: 12/14/2022] Open
Abstract
Biomarkers are important tools for diagnosis, prognosis, and identification of the causal factors of physiological conditions. Biomarkers are typically identified by correlating biological measurements with the status of a condition in a sample of subjects. Cross-sectional studies sample subjects at a single timepoint, whereas longitudinal studies follow a cohort through time. Identifying biomarkers of aging is subject to unique challenges. Individuals who age faster have intrinsically higher mortality rates and so are preferentially lost over time, in a phenomenon known as cohort selection. In this article, we use simulations to show that cohort selection biases cross-sectional analysis away from identifying causal loci of aging, to the point where cross-sectional studies are less likely to identify loci that cause aging than if loci had been chosen at random. We go on to show this bias can be corrected by incorporating correlates of mortality identified from longitudinal studies, allowing cross-sectional studies to effectively identify the causal factors of aging.
Collapse
Affiliation(s)
- Paul G Nelson
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson
| | | | - Joanna Masel
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson
| |
Collapse
|
43
|
Zhao SG, Yu M, Spratt DE, Chang SL, Feng FY, Kim MM, Speers CW, Carlson BL, Mladek AC, Lawrence TS, Sarkaria JN, Wahl DR. Xenograft-based, platform-independent gene signatures to predict response to alkylating chemotherapy, radiation, and combination therapy for glioblastoma. Neuro Oncol 2020; 21:1141-1149. [PMID: 31121035 DOI: 10.1093/neuonc/noz090] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Predictive molecular biomarkers to select optimal treatment for patients with glioblastoma and other cancers are lacking. New strategies are needed when large randomized trials with correlative molecular data are not feasible. METHODS Gene signatures (GS) were developed from 31 orthotopic glioblastoma patient-derived xenografts (PDXs), treated with standard therapies, to predict benefit from radiotherapy (RT-GS), temozolomide (Chemo-GS), or the combination (ChemoRT-GS). Independent validation was performed in a heterogeneously treated clinical cohort of 502 glioblastoma patients with overall survival as the primary endpoint. Multivariate Cox analysis was used to adjust for confounding variables and evaluate interactions between signatures and treatment. RESULTS PDX models recapitulated the clinical heterogeneity of glioblastoma patients. RT-GS, Chemo-GS, and ChemoRT-GS were correlated with benefit from treatment in the PDX models. In independent clinical validation, higher RT-GS scores were associated with increased survival only in patients receiving RT (P = 0.0031, hazard ratio [HR] = 0.78 [0.66-0.92]), higher Chemo-GS scores were associated with increased survival only in patients receiving chemotherapy (P < 0.0001, HR = 0.66 [0.55-0.8]), and higher ChemoRT-GS scores were associated with increased survival only in patients receiving ChemoRT (P = 0.0001, HR = 0.54 [0.4-0.74]). RT-GS and ChemoRT-GS had significant interactions with treatment on multivariate analysis (P = 0.0009 and 0.02, respectively), indicating that they are bona fide predictive biomarkers. CONCLUSIONS Using a novel PDX-driven methodology, we developed and validated 3 platform-independent molecular signatures that predict benefit from standard of care therapies for glioblastoma. These signatures may be useful to personalize glioblastoma treatment in the clinic and this approach may be a generalizable method to identify predictive biomarkers without resource-intensive randomized trials.
Collapse
Affiliation(s)
- Shuang G Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Menggang Yu
- Department of Biostatistics, University of Wisconsin, Madison, Wisconsin
| | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - S Laura Chang
- Department of Urology, Medicine, and Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Felix Y Feng
- Department of Urology, Medicine, and Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Corey W Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
44
|
Arfè A, Fell G, Alexander B, Awad MM, Rodig SJ, Trippa L, Schoenfeld JD. Meta-Analysis of PD-L1 Expression As a Predictor of Survival After Checkpoint Blockade. JCO Precis Oncol 2020; 4:1196-1206. [DOI: 10.1200/po.20.00150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Programmed cell death receptor ligand 1 (PD-L1) expression is the most studied biomarker to predict the efficacy of immune checkpoint inhibitors (ICIs), but its clinical significance is controversial. We estimated the distribution of PD-L1 expression scores (ie, tumor proportion score or combined proportion score) and the relationship between PD-L1 levels and ICIs’ impact on overall survival (OS). METHODS We reconstructed, pooled, and analyzed individual-level data on 7,617 patients with cancer from 14 randomized clinical trials. The effects of ICIs were quantified using differences in 24-month restricted mean survival times (ΔRMSTs; ie, the increase in life expectancy truncated at 2 years associated with ICI therapy). In a simulation study, we compared standard randomized clinical trial designs with a trial design that leverages meta-analytic results like ours. RESULTS Approximately 93% of patients had a PD-L1 expression ≤ 5% (66% of patients) or > 50% (27% of patients). OS improves with ICIs regardless of PD-L1 expression level, which predicts the benefits’ magnitude. For patients with non–small-cell lung cancer (NSCLC), ΔRMSTs ranged from 1.4 months (95% probability interval [PI], 0.7 to 2.2 months) for PD-L1 expression ≤ 1% to 4.1 months (95% PI, 3.2 to 5.2 months) for PD-L1 expression > 80%. For patients with non-NSCLC tumors, ΔRMSTs ranged from 0.8 months (95% PI, −0.1 to 1.7 months) to 2.3 months (95% PI, 1.3 to 4.4 months), again for PD-L1 expression levels of ≤ 1% and > 80%, respectively. Simulations suggested that designs tailored to meta-analytic results can detect the effects of ICIs in PD-L1 subgroups with higher probability (> 15%) than standard designs. CONCLUSION The practice of dichotomizing the range of PD-L1 expression scores is inadequate for patient stratification. Meta-analytic estimates of the distribution of PD-L1 scores and subgroup-specific treatment effects can improve the designs of future trials of ICIs.
Collapse
Affiliation(s)
- Andrea Arfè
- Harvard-MIT Center for Regulatory Science, Harvard Medical School, Boston, MA
| | - Geoffrey Fell
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | | | - Mark M. Awad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Scott J. Rodig
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Lorenzo Trippa
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | | |
Collapse
|
45
|
Wang T, Wang X, George SL, Zhou H. Design and analysis of biomarker-integrated clinical trials with adaptive threshold detection and flexible patient enrichment. J Biopharm Stat 2020; 30:1060-1076. [PMID: 33175640 PMCID: PMC7954851 DOI: 10.1080/10543406.2020.1832110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/12/2020] [Indexed: 10/23/2022]
Abstract
We propose a new adaptive threshold detection and enrichment design in which the biomarker threshold is adaptively estimated and updated by optimizing a trade-off between the size of the biomarker positive population and the magnitude of the treatment effect in that population. Enrichment is based on an enrollment criterion that accounts for the uncertainty in estimation of the threshold. Early termination for futility is allowed based on predictive success probability. Valid testing and estimation techniques for the treatment effect overall and inpatient subgroups are studied. Simulations and an example demonstrate advantages of the proposed design over existing designs.
Collapse
Affiliation(s)
- Ting Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, U.S.A
| | - Xiaofei Wang
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, U.S.A
| | - Stephen L George
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, U.S.A
| | - Haibo Zhou
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, U.S.A
| |
Collapse
|
46
|
Cavalcante P, Mantegazza R, Bernasconi P. Pharmacogenetic and pharmaco-miR biomarkers for tailoring and monitoring myasthenia gravis treatments. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1804865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Paola Cavalcante
- Neurology IV Unit ‒ Neuroimmunology and Neuromuscular Diseases, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | - Renato Mantegazza
- Neurology IV Unit ‒ Neuroimmunology and Neuromuscular Diseases, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | - Pia Bernasconi
- Neurology IV Unit ‒ Neuroimmunology and Neuromuscular Diseases, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
47
|
Han Y, Yuan Y, Cao S, Li M, Zang Y. On the Use of Marker Strategy Design to Detect Predictive Marker Effect in Cancer Immunotherapy and Targeted Therapy. STATISTICS IN BIOSCIENCES 2020. [DOI: 10.1007/s12561-019-09255-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
48
|
Principles of Good Clinical Trial Design. J Thorac Oncol 2020; 15:1277-1280. [PMID: 32417343 DOI: 10.1016/j.jtho.2020.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/13/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
Clinical trials are a fundamental component of medical research and serve as the main route to obtain evidence of the safety and efficacy of treatment before its approval. A trial's ability to provide the intended evidence hinges on appropriate design, background knowledge, trial rationale to sample size, and interim monitoring rules. In this article, we present some general design principles for investigators and their research teams to consider when planning to conduct a trial.
Collapse
|
49
|
Meehan J, Gray M, Martínez-Pérez C, Kay C, Pang LY, Fraser JA, Poole AV, Kunkler IH, Langdon SP, Argyle D, Turnbull AK. Precision Medicine and the Role of Biomarkers of Radiotherapy Response in Breast Cancer. Front Oncol 2020; 10:628. [PMID: 32391281 PMCID: PMC7193869 DOI: 10.3389/fonc.2020.00628] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy remains an important treatment modality in nearly two thirds of all cancers, including the primary curative or palliative treatment of breast cancer. Unfortunately, largely due to tumor heterogeneity, tumor radiotherapy response rates can vary significantly, even between patients diagnosed with the same tumor type. Although in recent years significant technological advances have been made in the way radiation can be precisely delivered to tumors, it is proving more difficult to personalize radiotherapy regimens based on cancer biology. Biomarkers that provide prognostic or predictive information regarding a tumor's intrinsic radiosensitivity or its response to treatment could prove valuable in helping to personalize radiation dosing, enabling clinicians to make decisions between different treatment options whilst avoiding radiation-induced toxicity in patients unlikely to gain therapeutic benefit. Studies have investigated numerous ways in which both patient and tumor radiosensitivities can be assessed. Tumor molecular profiling has been used to develop radiosensitivity gene signatures, while the assessment of specific intracellular or secreted proteins, including circulating tumor cells, exosomes and DNA, has been performed to identify prognostic or predictive biomarkers of radiation response. Finally, the investigation of biomarkers related to radiation-induced toxicity could provide another means by which radiotherapy could become personalized. In this review, we discuss studies that have used these methods to identify or develop prognostic/predictive signatures of radiosensitivity, and how such assays could be used in the future as a means of providing personalized radiotherapy.
Collapse
Affiliation(s)
- James Meehan
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Gray
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Martínez-Pérez
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Charlene Kay
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Lisa Y Pang
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jennifer A Fraser
- School of Applied Science, Sighthill Campus, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Amy V Poole
- School of Applied Science, Sighthill Campus, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Arran K Turnbull
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
50
|
Stroes CI, Schokker S, Creemers A, Molenaar RJ, Hulshof MC, van der Woude SO, Bennink RJ, Mathôt RA, Krishnadath KK, Punt CJ, Verhoeven RH, van Oijen MG, Creemers GJ, Nieuwenhuijzen GA, van der Sangen MJ, Beerepoot LV, Heisterkamp J, Los M, Slingerland M, Cats A, Hospers GA, Bijlsma MF, van Berge Henegouwen MI, Meijer SL, van Laarhoven HW. Phase II Feasibility and Biomarker Study of Neoadjuvant Trastuzumab and Pertuzumab With Chemoradiotherapy for Resectable Human Epidermal Growth Factor Receptor 2-Positive Esophageal Adenocarcinoma: TRAP Study. J Clin Oncol 2020; 38:462-471. [PMID: 31809243 PMCID: PMC7007286 DOI: 10.1200/jco.19.01814] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2019] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Approximately 15% to 43% of esophageal adenocarcinomas (EACs) are human epidermal growth factor receptor 2 (HER2) positive. Because dual-agent HER2 blockade demonstrated a survival benefit in breast cancer, we conducted a phase II feasibility study of trastuzumab and pertuzumab added to neoadjuvant chemoradiotherapy (nCRT) in patients with EAC. PATIENTS AND METHODS Patients with resectable HER2-positive EAC received standard nCRT with carboplatin and paclitaxel and 41.4 Gy of radiotherapy, with 4 mg/kg of trastuzumab on day 1, 2 mg/kg per week during weeks 2 to 6, and 6 mg/kg per week during weeks 7, 10, and 13 and 840 mg of pertuzumab every 3 weeks. The primary end point was feasibility, defined as ≥ 80% completion of treatment with both trastuzumab and pertuzumab. An exploratory comparison of survival with a propensity score-matched cohort receiving standard nCRT was performed, as were exploratory pharmacokinetic and biomarker analyses. RESULTS Of the 40 enrolled patients (78% men; median age, 63 years), 33 (83%) completed treatment with trastuzumab and pertuzumab. No unexpected safety events were observed. R0 resection was achieved in all patients undergoing surgery, with pathologic complete response in 13 patients (34%). Three-year progression-free and overall survival (OS) were 57% and 71%, respectively (median follow-up, 32.1 months). Compared with the propensity score-matched cohort, a significantly longer OS was observed with HER2 blockade (hazard ratio, 0.58; 95% CI, 0.34 to 0.97). Results of pharmacokinetic analysis and activity on [18F]fluorodeoxyglucose positron emission tomography scans did not correlate with survival or pathologic response. Patients with HER2 3+ overexpression or growth factor receptor-bound protein 7 (Grb7) -positive tumors at baseline demonstrated significantly better survival (P = .007) or treatment response (P = .016), respectively. CONCLUSION Addition of trastuzumab and pertuzumab to nCRT in patients with HER2-positive EAC is feasible and demonstrates potentially promising activity compared with historical controls. HER2 3+ overexpression and Grb7 positivity are potentially predictive for survival and treatment response, respectively.
Collapse
Affiliation(s)
- Charlotte I. Stroes
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sandor Schokker
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Aafke Creemers
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Remco J. Molenaar
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Maarten C.C.M. Hulshof
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Stephanie O. van der Woude
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Roel J. Bennink
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ron A.A. Mathôt
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Kausilia K. Krishnadath
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Cornelis J.A. Punt
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | - Martijn G.H. van Oijen
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | - Maartje Los
- Sint Antonius Hospital, Nieuwegein, the Netherlands
| | | | - Annemieke Cats
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Maarten F. Bijlsma
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Mark I. van Berge Henegouwen
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sybren L. Meijer
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Hanneke W.M. van Laarhoven
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|