Cargando…

A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold

Tumor mutation burden (TMB) is a recognized stratification biomarker for immunotherapy. Nevertheless, the general TMB-high threshold is unstandardized due to severe clinical controversies, with the underlying cause being inconsistency between multiple assessment criteria and imprecision of the TMB v...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yixuan, Lai, Xin, Wang, Jiayin, Xu, Ying, Zhang, Xuanping, Zhu, Xiaoyan, Liu, Yuqian, Shao, Yang, Zhang, Li, Fang, Wenfeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9386083/
https://www.ncbi.nlm.nih.gov/pubmed/35991549
http://dx.doi.org/10.3389/fgene.2022.915839
_version_ 1784769723824603136
author Wang, Yixuan
Lai, Xin
Wang, Jiayin
Xu, Ying
Zhang, Xuanping
Zhu, Xiaoyan
Liu, Yuqian
Shao, Yang
Zhang, Li
Fang, Wenfeng
author_facet Wang, Yixuan
Lai, Xin
Wang, Jiayin
Xu, Ying
Zhang, Xuanping
Zhu, Xiaoyan
Liu, Yuqian
Shao, Yang
Zhang, Li
Fang, Wenfeng
author_sort Wang, Yixuan
collection PubMed
description Tumor mutation burden (TMB) is a recognized stratification biomarker for immunotherapy. Nevertheless, the general TMB-high threshold is unstandardized due to severe clinical controversies, with the underlying cause being inconsistency between multiple assessment criteria and imprecision of the TMB value. The existing methods for determining TMB thresholds all consider only a single dimension of clinical benefit and ignore the interference of the TMB error. Our research aims to determine the TMB threshold optimally based on multifaceted clinical efficacies accounting for measurement errors. We report a multi-endpoint joint model as a generalized method for inferring the TMB thresholds, facilitating consistent statistical inference using an iterative numerical estimation procedure considering mis-specified covariates. The model optimizes the division by combining objective response rate and time-to-event outcomes, which may be interrelated due to some shared traits. We augment previous works by enabling subject-specific random effects to govern the communication among distinct endpoints. Our simulations show that the proposed model has advantages over the standard model in terms of precision and stability in parameter estimation and threshold determination. To validate the feasibility of the proposed thresholds, we pool a cohort of 73 patients with non-small-cell lung cancer and 64 patients with nasopharyngeal carcinoma who underwent anti-PD-(L)1 treatment, as well as validation cohorts of 943 patients. Analyses revealed that our approach could grant clinicians a holistic efficacy assessment, culminating in a robust determination of the TMB screening threshold for superior patients. Our methodology has the potential to yield innovative insights into therapeutic selection and support precision immuno-oncology.
format Online
Article
Text
id pubmed-9386083
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-93860832022-08-19 A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold Wang, Yixuan Lai, Xin Wang, Jiayin Xu, Ying Zhang, Xuanping Zhu, Xiaoyan Liu, Yuqian Shao, Yang Zhang, Li Fang, Wenfeng Front Genet Genetics Tumor mutation burden (TMB) is a recognized stratification biomarker for immunotherapy. Nevertheless, the general TMB-high threshold is unstandardized due to severe clinical controversies, with the underlying cause being inconsistency between multiple assessment criteria and imprecision of the TMB value. The existing methods for determining TMB thresholds all consider only a single dimension of clinical benefit and ignore the interference of the TMB error. Our research aims to determine the TMB threshold optimally based on multifaceted clinical efficacies accounting for measurement errors. We report a multi-endpoint joint model as a generalized method for inferring the TMB thresholds, facilitating consistent statistical inference using an iterative numerical estimation procedure considering mis-specified covariates. The model optimizes the division by combining objective response rate and time-to-event outcomes, which may be interrelated due to some shared traits. We augment previous works by enabling subject-specific random effects to govern the communication among distinct endpoints. Our simulations show that the proposed model has advantages over the standard model in terms of precision and stability in parameter estimation and threshold determination. To validate the feasibility of the proposed thresholds, we pool a cohort of 73 patients with non-small-cell lung cancer and 64 patients with nasopharyngeal carcinoma who underwent anti-PD-(L)1 treatment, as well as validation cohorts of 943 patients. Analyses revealed that our approach could grant clinicians a holistic efficacy assessment, culminating in a robust determination of the TMB screening threshold for superior patients. Our methodology has the potential to yield innovative insights into therapeutic selection and support precision immuno-oncology. Frontiers Media S.A. 2022-08-04 /pmc/articles/PMC9386083/ /pubmed/35991549 http://dx.doi.org/10.3389/fgene.2022.915839 Text en Copyright © 2022 Wang, Lai, Wang, Xu, Zhang, Zhu, Liu, Shao, Zhang and Fang. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Genetics
Wang, Yixuan
Lai, Xin
Wang, Jiayin
Xu, Ying
Zhang, Xuanping
Zhu, Xiaoyan
Liu, Yuqian
Shao, Yang
Zhang, Li
Fang, Wenfeng
A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold
title A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold
title_full A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold
title_fullStr A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold
title_full_unstemmed A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold
title_short A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold
title_sort joint model considering measurement errors for optimally identifying tumor mutation burden threshold
topic Genetics
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9386083/
https://www.ncbi.nlm.nih.gov/pubmed/35991549
http://dx.doi.org/10.3389/fgene.2022.915839
work_keys_str_mv AT wangyixuan ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT laixin ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT wangjiayin ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT xuying ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT zhangxuanping ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT zhuxiaoyan ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT liuyuqian ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT shaoyang ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT zhangli ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT fangwenfeng ajointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT wangyixuan jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT laixin jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT wangjiayin jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT xuying jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT zhangxuanping jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT zhuxiaoyan jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT liuyuqian jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT shaoyang jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT zhangli jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold
AT fangwenfeng jointmodelconsideringmeasurementerrorsforoptimallyidentifyingtumormutationburdenthreshold