Cargando…

TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits

A high tumor mutation burden (TMB) is known to drive the response to immune checkpoint inhibitors (ICI) and is associated with favorable prognoses. However, because it is a one-dimensional numerical representation of non-synonymous genetic alterations, TMB suffers from clinical challenges due to its...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yixuan, Wang, Jiayin, Fang, Wenfeng, Xiao, Xiao, Wang, Quan, Zhao, Jian, Liu, Jingjing, Yang, Shuanying, Liu, Yuqian, Lai, Xin, Song, Xiaofeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10208409/
https://www.ncbi.nlm.nih.gov/pubmed/37234148
http://dx.doi.org/10.3389/fimmu.2023.1151755
_version_ 1785046661619253248
author Wang, Yixuan
Wang, Jiayin
Fang, Wenfeng
Xiao, Xiao
Wang, Quan
Zhao, Jian
Liu, Jingjing
Yang, Shuanying
Liu, Yuqian
Lai, Xin
Song, Xiaofeng
author_facet Wang, Yixuan
Wang, Jiayin
Fang, Wenfeng
Xiao, Xiao
Wang, Quan
Zhao, Jian
Liu, Jingjing
Yang, Shuanying
Liu, Yuqian
Lai, Xin
Song, Xiaofeng
author_sort Wang, Yixuan
collection PubMed
description A high tumor mutation burden (TMB) is known to drive the response to immune checkpoint inhibitors (ICI) and is associated with favorable prognoses. However, because it is a one-dimensional numerical representation of non-synonymous genetic alterations, TMB suffers from clinical challenges due to its equal quantification. Since not all mutations elicit the same antitumor rejection, the effect on immunity of neoantigens encoded by different types or locations of somatic mutations may vary. In addition, other typical genomic features, including complex structural variants, are not captured by the conventional TMB metric. Given the diversity of cancer subtypes and the complexity of treatment regimens, this paper proposes that tumor mutations capable of causing various degrees of immunogenicity should be calculated separately. TMB should therefore, be segmented into more exact, higher dimensional feature vectors to exhaustively measure the foreignness of tumors. We systematically reviewed patients’ multifaceted efficacy based on a refined TMB metric, investigated the association between multidimensional mutations and integrative immunotherapy outcomes, and developed a convergent categorical decision-making framework, TMBserval (Statistical Explainable machine learning with Regression-based VALidation). TMBserval integrates a multiple-instance learning concept with statistics to create a statistically interpretable model that addresses the broad interdependencies between multidimensional mutation burdens and decision endpoints. TMBserval is a pan-cancer-oriented many-to-many nonlinear regression model with discrimination and calibration power. Simulations and experimental analyses using data from 137 actual patients both demonstrated that our method could discriminate between patient groups in a high-dimensional feature space, thereby rationally expanding the beneficiary population of immunotherapy.
format Online
Article
Text
id pubmed-10208409
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-102084092023-05-25 TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits Wang, Yixuan Wang, Jiayin Fang, Wenfeng Xiao, Xiao Wang, Quan Zhao, Jian Liu, Jingjing Yang, Shuanying Liu, Yuqian Lai, Xin Song, Xiaofeng Front Immunol Immunology A high tumor mutation burden (TMB) is known to drive the response to immune checkpoint inhibitors (ICI) and is associated with favorable prognoses. However, because it is a one-dimensional numerical representation of non-synonymous genetic alterations, TMB suffers from clinical challenges due to its equal quantification. Since not all mutations elicit the same antitumor rejection, the effect on immunity of neoantigens encoded by different types or locations of somatic mutations may vary. In addition, other typical genomic features, including complex structural variants, are not captured by the conventional TMB metric. Given the diversity of cancer subtypes and the complexity of treatment regimens, this paper proposes that tumor mutations capable of causing various degrees of immunogenicity should be calculated separately. TMB should therefore, be segmented into more exact, higher dimensional feature vectors to exhaustively measure the foreignness of tumors. We systematically reviewed patients’ multifaceted efficacy based on a refined TMB metric, investigated the association between multidimensional mutations and integrative immunotherapy outcomes, and developed a convergent categorical decision-making framework, TMBserval (Statistical Explainable machine learning with Regression-based VALidation). TMBserval integrates a multiple-instance learning concept with statistics to create a statistically interpretable model that addresses the broad interdependencies between multidimensional mutation burdens and decision endpoints. TMBserval is a pan-cancer-oriented many-to-many nonlinear regression model with discrimination and calibration power. Simulations and experimental analyses using data from 137 actual patients both demonstrated that our method could discriminate between patient groups in a high-dimensional feature space, thereby rationally expanding the beneficiary population of immunotherapy. Frontiers Media S.A. 2023-05-10 /pmc/articles/PMC10208409/ /pubmed/37234148 http://dx.doi.org/10.3389/fimmu.2023.1151755 Text en Copyright © 2023 Wang, Wang, Fang, Xiao, Wang, Zhao, Liu, Yang, Liu, Lai and Song 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 Immunology
Wang, Yixuan
Wang, Jiayin
Fang, Wenfeng
Xiao, Xiao
Wang, Quan
Zhao, Jian
Liu, Jingjing
Yang, Shuanying
Liu, Yuqian
Lai, Xin
Song, Xiaofeng
TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits
title TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits
title_full TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits
title_fullStr TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits
title_full_unstemmed TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits
title_short TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits
title_sort tmbserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10208409/
https://www.ncbi.nlm.nih.gov/pubmed/37234148
http://dx.doi.org/10.3389/fimmu.2023.1151755
work_keys_str_mv AT wangyixuan tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT wangjiayin tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT fangwenfeng tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT xiaoxiao tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT wangquan tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT zhaojian tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT liujingjing tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT yangshuanying tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT liuyuqian tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT laixin tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits
AT songxiaofeng tmbservalastatisticalexplainablelearningmodelrevealsweightedtumormutationburdenbettercategorizingtherapeuticbenefits