Skip to main content

ORIGINAL RESEARCH article

Front. Genet., 04 August 2022
Sec. Cancer Genetics and Oncogenomics
This article is part of the Research Topic The Role of Immunophenotype in Tumor Immunotherapy Response View all 33 articles

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

Yixuan Wang&#x;Yixuan Wang1Xin Lai&#x;Xin Lai1Jiayin Wang,,
Jiayin Wang1,2,3*Ying XuYing Xu1Xuanping ZhangXuanping Zhang1Xiaoyan ZhuXiaoyan Zhu1Yuqian LiuYuqian Liu1Yang Shao,Yang Shao4,5Li ZhangLi Zhang6Wenfeng Fang
Wenfeng Fang6*
  • 1School of Computer Science and Technology, Faculty of Electronics and Information Engineering, Xi’an Jiaotong University, Xi’an, China
  • 2School of Management, Hefei University of Technology, Hefei, China
  • 3The Ministry of Education Key Laboratory of Process Optimization and Intelligent Decision-Making, Hefei University of Technology, Hefei, China
  • 4Nanjing Geneseeq Technology Inc., Nanjing, China
  • 5School of Public Health, Nanjing Medical University, Nanjing, China
  • 6State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China

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.

Introduction

Immune checkpoint inhibitor (ICI) therapy has emerged as a promising strategy with confirmed efficacy for advanced or metastatic tumors (Bracarda et al., 2015; Motzer et al., 2015; Chiang et al., 2020; Kuryk et al., 2020; Wołącewicz et al., 2020; Majc et al., 2021). Tumor mutation burden (TMB, defined as the number of somatic mutations per mega-base) is a recognized biomarker of sensitivity to ICIs (Hellmann et al., 2018a; Cristescu et al., 2018; Bai et al., 2020; Wang et al., 2020), according to the underlying connection between the increasing number of somatic mutations and the neo-antigen that the activated T cells can recognize (Van Rooij et al., 2013), enhancing the tumor immunogenicity (Pardoll, 2012; Conway et al., 2018). A high TMB tends to trigger a favorable prognosis (Yarchoan et al., 2017; Legrand et al., 2018), which has been observed in urothelial carcinoma (Rosenberg et al., 2016), small-cell-lung cancer (Hellmann et al., 2018b), non-small-cell lung cancer (NSCLC) (Lim et al., 2015; Rizvi et al., 2015; Carbone et al., 2017; Hellmann et al., 2018c; Singal et al., 2019), and melanoma (Johnson et al., 2016; Goodman et al., 2017). TMB is a suggested test for patients undergoing immunotherapy by both NCCN and FDA (Lemery et al., 2017; Boyiadzis et al., 2018; Subbiah et al., 2020).

In clinical practices, the TMB threshold is a baseline for identifying patients with potential ICI benefits (Samstein et al., 2019). TMB thresholds are typically determined in two ways: either grouped by quartiles, which is obviously imprecise (Campesato et al., 2015; Colli et al., 2016; Riaz et al., 2017; Miao et al., 2018; Wood et al., 2020), or numerical thresholds generated from statistical tests of significance based on efficacy endpoints (Goodman et al., 2017). Notably, among these previous statistical studies, retrospective evaluations of efficacy are limited to a single dimension, most regularly the response. The primary endpoints for immuno-oncology include objective tumor response and time-to-event (TTE), where the TMB biomarker has been observed to be associated with both (Cao et al., 2019). Such diverse efficacy evaluation metrics have sparked controversy in the threshold standardization (Goodman et al., 2017). When assessments base on different endpoints over the same cohort, inconsistent thresholds arise, and clinicians are left inconclusive about which one to choose. Furthermore, clinical decisions need a comprehensive review of the diseased multifaceted efficacy rather than a single endpoint that exhibits a partial treatment effect. Therefore, there is an urgent clinical need for inference on multiple endpoints to derive a comprehensive TMB threshold. However, it is computationally challenging for two reasons. First, if several individual endpoints are to be inferred simultaneously, the intersection cannot be taken directly. Instead, some adjustment for multiple testing is required to control the familywise type I error rate (FWER) (Ristl et al., 2019). Constructing the joint distribution of different endpoints is preferable to the straightforward application of Bonferroni inequalities in terms of maximizing the utilization of available information, providing unbiased results, and allowing for statistical alpha levels closer to nominal levels while boosting the statistical power (Phillips et al., 2003; Asar et al., 2015; Guidance 2017). Secondly, the existing joint modeling studies have mostly taken a perspective on analyzing longitudinal biochemical markers within the survival analysis framework. Whereas the volatility of tumor genomic traits in immunotherapy trials is quite limited, we are more concerned with the within-subject dependence between different endpoints. Binary tumor responses conforming to the Bernoulli distribution do not satisfy the premise of a normal distribution in linear regression. The existing models have limited capacity to comprehend possible shared biologic processes on endpoints of tumor remission with survival and are not applicable to scenarios of immune efficacy investigation.

Moreover, the imprecision of TMB values is another source of threshold controversies (Wood et al., 2020). Despite the different calculation methods of TMB, the accuracy of variant callings can never reach 100% due to technical limitations (Xu et al., 2014; Alioto et al., 2015), and TMB always harbors measurement errors. Existing models neglect the difference between the actual and observed values of TMB, which lead to significant bias in statistical inference (Campesato et al., 2015; Colli et al., 2016; Goodman et al., 2017; Riaz et al., 2017; Miao et al., 2018; Wood et al., 2020). Parameter inference for statistical models is conventionally obtained by maximum likelihood estimation (MLE), and unbiasedness of the score function for likelihood (i.e., expectation equal to zero) is a critical criterion for ensuring estimate consistency. With the accurate TMB values being unascertainable, the observations TMB (TMB=TMB+e) have to be used for surrogates. The presence of its inherent random error term e undermines the unbiased nature of the score expectation, yielding inconsistent regression coefficient estimates. The biasing effect caused by error term confounds the proper relationship between TMB and ICI. Furthermore, naïve statistical inference assesses patient prognosis inaccurately. Thus, the final determination of TMB thresholds must be flawed, hindering accurate screening of applicable patients and closely related to the risk of adverse events to immunotherapy. Although the corrected-score methodology is associated with a measurement error (Nakamura, 1990; Novick & Stefanski, 2002; Augustin, 2004), a new algorithm should be re-inferred due to the complexity of the specific joint model. The challenge lies in the fact that the complete joint probability is essentially a complex integration without an exact analytical solution. Patients’ responses couple with the survival process, based on the random effects governing both, so that the joint score function is usually impossible to strip. It is incapable of eliminating mistakes from this joint likelihood directly. A new iterative numerical estimation procedure is required by considering the biasing impacts induced by the mis-specified TMB covariate.

Therefore, we report a generalized method for optimizing the identification of TMB-positive thresholds. Our method integrates binary response and continuous TTE endpoints to provide a comprehensive efficacy assessment, while, to our best knowledge, it is among the first statistical approaches accounting for TMB measurement errors. To verify the viability of the multi-endpoint joint model, we conducted a series of simulation experiments, and the results confirmed our superiority in the accuracy of parameter estimation and fault tolerance of threshold delineation compared with the standard separate regression model. Meanwhile, we gathered a cohort of 73 non-small-cell lung cancer (NSCLC) patients and 64 nasopharyngeal carcinoma (NPC) patients and validation cohorts of 943 patients who underwent ICI treatment to illustrate the applicability of the model across carcinomas. It is known that different cancer types and TMB calculations often yield different thresholds, but we provide here a generalized statistical method applicable for any known scenarios. The data results show that the proposed model can obtain a more comprehensive and robust TMB threshold to support therapeutic refinement for cancer patients. The source code reproduces the figures, and results can be downloaded from https://github.com/YixuanWang1120/TMB_JM.

Materials and Methods

To comprehensively determine TMB-positivity thresholds from multifaceted efficacy analyses while considering inevitable measurement errors, we present a general approach for the simultaneous joint modeling of multiple endpoints, yielding approximately consistent statistical inference for mis-specified covariates by developing an iterative numerical estimation procedure using the corrected-score method. The observed sample information contains the patient’s clinically recorded objective response rate (ORR) and TTE endpoints, other clinical indicators (correctly specified), and the corresponding TMB observations with measurement errors. The data consist of n independent observations of R, T, Δ, Z, and TMB, denoting the binary tumor response outcome, continuous survival time, event indicator, vector of accurately measured covariates, and mismeasured TMB, respectively. To simplify, the additive measurement error model relates the true unobserved TMB index to the observed TMB: TMB = TMB + e, where eN(0,e).

A Joint Model Considers Binary and Continuous Endpoints

For patient i(i=1,2,,n), Ri denotes the tumor response (Ri = 1,0 for complete response (CR) and partial response (PR), stable disease (SD) and progressive disease (PD))and Zi denotes a vector of covariates, e.g., age, gender, treatment indicator, cancer stage. Binary response outcomes are typically modeled by logistic regression whose standard form is quite well established for the immunological effectiveness analysis. Ri depends on Zi and TMBi, then the mixed-effect logistical regression sub-model for the ORR endpoint is formulated as:

logit(Ri|Zi,TMBi,bi;θ)=αzTZi+αmTMBi+bi

where αz and αm denote the corresponding response regression coefficients, θ represents all unknown parameters in the joint model, and bi denotes the random effect. The exponent of the estimated parameter exp(α) for the logit regression of binary outcomes can be interpreted intuitively as the multiples of change in the odds ratio caused by a one-unit increase in the corresponding variable.

Let Ti denote the observed event time (such as tumor relapses, progression, death, etc.), which is taken as the minimum of the true event time Ui and the censoring time Ci, that is, Ti=min(Ui,Ci). Define the event indicator as Δi=I(UiCi), where I() is the indicator function. Here, we adopt the widely accepted Cox PH model because it focuses more on the identifying patients’ survival risk classes compared with alternative accelerated failure (AFT) models, is appropriate to the scenario of screening immunotherapy-beneficial patients in this article, and allows for more flexible baseline risk. Ti also depends on Zi, TMBi, unknown parameters θ, and random effect bi; then, the mixed-effect Cox PH regression sub-model for the TTE endpoint is formulated as:

hi(t|Zi,TMBi,bi;θ)=h0(t)exp(βzTZi+βmTMBi+bi)
Si(t|Zi,TMBi,bi;θ)=exp{0th0(s)exp(βzTZi+βmTMBi+bi)ds}=exp{0(t)exp(βzTZi+βmTMBi+bi)}

where h(t) describes the instantaneous risk for an event in the time interval [t, t + dt) provided survival up to t, while S(t) represents the survival probability. h0(t) is referred to as baseline hazard and follows the Weibull distribution h0(t)=λt(λ1) because the trend in the baseline cumulative hazard distribution for progression-free survival in the cohort receiving immunotherapy is consistent with the Weibull distribution with a scale parameter equal to 1 (see in Figure 1). βz is the corresponding vector of covariate effect and βm quantifies the TMB effect.

FIGURE 1
www.frontiersin.org

FIGURE 1. The distribution of baseline cumulative hazard for patients receiving immunotherapy.

The maximum likelihood estimates are derived as the modes of the log-likelihood function corresponding to the joint distribution of the observed samples Dn={Ri,Ti,Δi,Zi,TMBi,i=1,n}. The vector b=(b1,b1,,bn) is the shared random effect on the respective endpoints, accounting for the intra-subject correlation between event times and individual tumor response and is assumed to follow a normal distribution N(0,σb2). Since the random effect bi accounts for the intra-subject association underlying both response and survival process, thus the two are conditionally independent given the random effect. Formally, for patient i, we have that:

p(Ri,Ti,Δi,bi;θ)=p(Ri|bi;θ)p(Ti,Δi|bi;θ)p(bi;θ)(1)

where the likelihood of the response is:

p(Ri|bi;θ)=F(αzTZi+αmTMBi+bi)Ri{1F(αzTZi+αmTMBi+bi)}(1Ri)F(υ)=(1+eυ)1

while the likelihood of the survival is:

p(Ti,Δi|bi;θ)=hi(Ti|bi;θ)ΔiSi(Ti|bi;θ).

By incorporating random effects (Barbieri et al., 2020), it is feasible to jointly model the multiple endpoints and regulate intricate correlations between response probabilities and event times. Then, the joint logarithmic likelihood can be formulated as:

(θ)=ilogp(Ri,Ti,Δi;θ)=ilogp(Ri|bi;θ)p(Ti,Δi|bi;θ)p(bi;θ)dbi(2)

Inference about parameters θ is typically based on the maximization of Eq. 2, while integrals about random effects apparently have no analytical solution. Here, we approximate (θ) based on the Laplace method, which has the advantage over other numerical integration techniques, including Gaussian Hermite quadrature and Monte Carlo (Lin et al., 2008; Rizopoulos et al., 2014), since the multiplicative form of the series can be easily unfolded by adopting the logarithmic trick, facilitating our correction of the measurement errors of the covariates later. The Laplace approximation is as follows:

abef(x)dx2π|f(x0)|ef(x0)

where the function f(x) has a unique global maximum at x0. So, the first-order Laplace approximation to the observed-data joint log-likelihood is:

˜i(θ,b^i)=12log2π+logp(Ri|b^i;θ)+logp(Ti,Δi|b^i;θ)+logp(b^i;θ)12log|k(b^i;θ)|(3)

where

k(bi;θ)=logp(Ri|bi;θ)+logp(Ti,Δi|bi;θ)+logp(bi;θ)=Ri(αzTZi+αmTMBi+bi)log{1+exp(αzTZi+αmTMBi+bi)}+Δi{logh0(Ti)+βzTZi+βmTMBi+bi}0(Ti)exp(βzTZi+βmTMBi+bi)+log(2π)/2+log(σb)b12/2σb2(4)

and the mode b^i is obtained for each patient by solving k(bi)=0 with a fixed θ,

k(bi;θ)=logp(Ri|bi;θ)bi+logp(Ti,Δi|bi;θ)bi+logp(bi;θ)bi=RiF(αzTZi+αmTMBi+bi)+Δi0(Ti)exp(βzTZi+βmTMBi+bi)biσb2(5)
|k(bi;θ)|=F(αzTZi+αmTMBi+bi){1F(αzTZi+αmTMBi+bi)}+0(Ti)exp(βzTZi+βmTMBi+bi)σb2(6)

The difference of Eq. 3 from the previous independent standard regressions lies in that the joint assessment entails examining the endpoint correlations, where logp(Ri|b^i;θ) represents the likelihood of ORR, while logp(Ti,Δi|b^i;θ) represents the information on survival endpoint, and logp(b^i;θ)12log|k(b^i;θ)| incorporates the within-subject dependence between two endpoints. When b^i=0, i.e., there is no correlation between the two clinical endpoints, the joint model degenerates to standard separate logistic regression and Cox PH regression.

Estimates obtained by maximizing ˜(θ)=i˜i(θ,b^i) are thus approximate maximum likelihood estimates (MLEs). The maximization is accomplished by solving the equation Ψ(θ)=˜(θ)θT=0, Ψ(θ) is score function. According to the negative of the inverse Hessian matrix at MLE θ^, we obtain the standard errors for the parameter estimates va^r(θ^)={H(θ^)}1, with H(θ^)={Ψ(θ)θT|θ^}1, and the asymptotic confidence interval is θ^±1.96se^(θ^). It is typically easier to employ a numerical derivative routine for the calculation of Hessian matrix, such as the forward or the central difference approximation.

Based on θ^, we obtain approximately consistent and unbiased estimates of the fixed effects for TMB and the random effects symbolizing intra-subject correlations between both endpoints. With the mutually moderating random effects, the joint likelihood that a patient has a favorable prognosis can be determined. This joint probability characterizes the positive prognosis of patients with both remission of tumor lesions and prolonged survival time, which can be utilized to analyze the patient’s treatment outcome more completely. The joint probability for patient i is:

p(Ri=1,Ti>T0;θ^)=2π|{2log(p(Ri=1|bi;θ^)p(Ti>T0|bi;θ^)p(bi;θ^))bi2}b^i|p(Ri=1|b^i;θ^)p(Ti>T0|b^i;θ^)p(b^i;θ^)(7)

where T0 is a pre-specified survival time.

Based on the joint probabilities that characterized the positive prognosis of the patients, we rank them and then label the populations to be analyzed according to the proportion of patients who would potentially benefit for ICI. Ultimately, the proposed joint model can stratify patients into two subgroups according to their TMB levels and the positive prognosis labels using the receiver operating characteristic curve (ROC) to balance the classification performance. Thresholds for the low- and high-TMB groups are selected from the local optima across a range of clinically meaningful values by Yoden Index.

The complete TMB threshold identification procedure based on the aforementioned joint model solved by Laplace approximation is given in Algorithm 1.

Algorithm 1. Identifying TMB threshold without measurement errors

www.frontiersin.org

Bias Arising From Measurement Error

Here, we further investigate the negative impact of measurement errors in TMB. The score function in Section 2.1 is unbiased. Base on Eqs. 3, 6, θT=[θRT,θTT,θb], θR=[αzT,αm]T, θT=[λ,βzT,βm]T, θb=σb, we have:

Ψi(Ri,Ti,Δi,Zi,TMBi;Θ)=˜i(θ,b^i)θT=ΨR,i(θ)+ΨT,i(θ)+Ψb,i(θ)Ψi(θR)=ΨR,i(θR)+Ψb,i(θR)={RiF(αzTZi+αmTMBi+b^i)12exp(αzTZi+αmTMBi+b^i){1exp(αzTZi+αmTMBi+b^i)}|k(b^i;θ)|{1+exp(αzTZi+αmTMBi+b^i)}3}(ZiTMBi)Ψi(λ)=ΨT,i(λ)+Ψb,i(λ)=Δi(λ1+logTi)TiλlogTiexp(βzTZi+βmTMBi+b^i)+12TiλlogTiexp(βzTZi+βmTMBi+b^i)|k(b^i;θ)|Ψi(βzT,βm)=ΨT,i(βzT,βm)+Ψb,i(βzT,βm)={ΔiTiλexp(βzTZi+βmTMBi+b^i)+12Tiλexp(βzTZi+βmTMBi+b^i)|k(b^i;θ)|}(ZiTMBi)Ψi(σb)=Ψb,i(σb)=σb1+b^12σb3+(σb|k(b^i;θ)|)1(8)

where ΨRi(θ) represents the score of ORR, ΨTi(θ) represents the score on the survival endpoint, and Ψbi(θ) represents the score of random effect.

The parameter θ^ relating R, T, Δ, Z, and TMB is approximately consistent by satisfying i=1nΨ(Ri,Ti,Δi,Zi,TMBi;θ^)=0, where the score function Ψ is conditionally unbiased for the approximate likelihood:

E{Ψ(Ri,Ti,Δi,Zi,TMBi;Θ)}=0i=1,,n.(9)

What will happen when measurement error exists? We assume the observed TMB is subject to the measurement error model: TMBi=TMBi+ei,i=1,,n. The error term ei is independent and identically normal distributed with mean zero and known variance σe2, and is independent of Ri, Ti, Δi, and Zi. Because true TMB is not observed and hence the true-data score function cannot be used for parameter estimation from the perspective of inconsistency E{Ψ(TMB;Θ)}=E{Ψ(TMB+e;Θ)}0.

As a more specific illustration, we consult the part of survival function:

E{ΔiZiTiλexp(βzTZi+βmTMBi+b^i)Zi}=ΔiZiTiλE{exp(βzTZi+βmTMBi+βmei+b^i)}Zi=ΔiZiTiλexp(βzTZi+βmTMBi+b^i)ZiE{exp(βmei)}0(10)

The additional term E{exp(βmei)} on the scoring function is generated by the measurement error, leading the naïve estimator to be biased apparently. As for the response score and distribution of random effects, F(αzTZi+αmTMBi+αmei+b^i) and 12log|(k(b^i,TMBi,ei;θ))| are also subject to the negative impact of the error term with non-zero expectations:

E{F(αzTZi+αmTMBi+αmei+b^i)}=+F(αzTZi+αmTMBi+αmei+b^i)p(ei)dei

due to the function F() is not axisymmetric about the origin. The presence of the inevitable random error term e undermines the unbiased nature of the score expectation.

Correction of TMB Measurement Error for Threshold Optimization

To reduce the biasing effect caused by measurement errors and obtain a more robust TMB threshold, we integrated the widely applicable corrected score with the joint model, resulting in approximately consistent estimators based on the observed data. A corrected score is a function Ψc of the observed data having the important property that

E{Ψc(Ri,Ti,Δi,Zi,TMBi;Θ)|Ri,Ti,Δi,Zi,TMBi}=Ψ(Ri,Ti,Δi,Zi,TMBi;Θ)(11)

which is conditionally unbiased for the true-data score function according to the property of conditional expectation, E{Ψc(Ri,Ti,Δi,Zi,TMBi;Θ)}=0. The corrected scores provide an approach to reducing bias incurred by a covariate measurement error. Thus, Ψc possesses a consistent, asymptotically normally sequence of solutions for i=1nΨc(Ri,Ti,Δi,Zi,TMBi;Θ)=0 (Nakamura, 1990; Carroll et al., 2006).

Based on Eqs. 4 and 8 and the property of corrected score, we derive a correct kc(bi) for the random effect estimator, and a corrected score Ψc for the ideal likelihood score Ψ. The corrected scores are defined as follows.

Let

kc(bi)=RiJ1j1JRe{F(αzTZi+αm,TMB˜j,i+bi)}+ΔiTiλm(βm)1exp(βzTZi+βmTMBi+bi)biσb2(12)

where the complex variate TMB˜j,i=TMBi+1ξj,i, and ξj,i is a normal random vector with zero mean and variance σe. Then, k(bi) is the corrected-score function for k(bi). The proof can be found in the Supplementary Material.

Furthermore, we obtain the joint corrected-score Ψc(R,T,Δ,Z,TMB;Θ)=[Ψc(θRT),Ψc(θTT),Ψc(θb)]T, where Ψc(θR)=ΨR_c(θR)+Ψb_c(θR), Ψc(θT)=ΨT_c(θT)+Ψb_c(θT), Ψc(θb)=Ψb_c(θb),

ΨR_c,i(θR)=Ri(ZiTMBi)J1j=1JRe{F(αzTZi+αmTMBj,i˜+b^i)(ZiTMBi˜)}ΨT_c,i(λ)=Δi(λ1+logTi)TiλlogTiexp(βzTZi+βmTMBi+b^i)m(βm)1ΨT_c,i(βzT)=[ΔiTiλexp(βzTZi+βmTMBi+b^i)m(βm)1]ZiΨT_c,i(βm)=ΔiTMBiTiλexp(βzTZi+βmTMBi+b^i)m(βm)1[TMBim(βm)1{m(βm)βm}](13)
Ψb_c,i(θR)=12|kc(b^i;θ)|J1j=1JRe{exp(αzTZi+αmTMBj,i˜+b^i){1exp(αzTZi+αmTMBj,i˜+b^i)}{1+exp(αzTZi+αmTMBj,i˜+b^i)}3}(ZiTMB˜j,i)Ψb_c,i(λ)=TiλlogTiexp(βzTZi+βmTMBi+b^i)m(βm)12|kc(b^i;θ)|Ψb_c,i(βzT)=Tiλexp(βzTZi+βmTMBi+b^i)m(βm)12|kc(b^i;θ)|ZiΨb_c,i(βm)=Tiλexp(βzTZi+βmTMBi+b^i)m(βm)12|kc(b^i;θ)|[TMBim(βm)1{m(βm)βm}]Ψb_c,i(σb)=σb1+b^i2σb3+σb1|kc(b^i;θ)|1(14)

We present the joint corrected scores based on the complex variable simulation extrapolation and the property of Eq. 11. Eq. 13 contains ΨR_c(θ) representing the corrected score for ORR, which follows the complex variable simulation extrapolation for logistic regression (see Lemma 3 in the Supplementary Material), while ΨT_c(θ) represents the corrected score for TTE satisfying the property of Eq. 11 (see Lemma 2 in the Supplementary Material). Then, based on the specificity of joint modeling, additional Ψb_c(θ) needs to be considered, which represents the difference between the standard correction and the joint model correction. Then, Ψc is the corrected-score function with the proof in the Supplementary Material. Consistency is achieved by virtue of the fact that the estimators are M-estimators whose score functions are unbiased in the presence of measurement error. The critical challenges of inferring the joint model are the random effects that characterize within-subject correlations. In the presence of measurement error, we need to correct the score functions of the random effects k(b^i) to ensure the unbiasedness of their estimates before dealing with complex joint score functions without exact solutions by kc(b^i) as well as Monte Carlo extrapolation, which is the gap in the existing literature addressed in this article. Solving the equations kc(b^i)=0 and i=1nΨc(Ri,Ti,Δi,Zi,TMBi;Θ)=0 by the Newton–Raphson iteration, it is ultimately possible to yield the approximately consistent estimators θ˜ for mis-specified covariates and b˜ for random effects.

The complete TMB threshold identification procedure based on the aforementioned Laplace approximation and corrected score is given in Algorithm 2.

Algorithm 2. Identifying TMB threshold with measurement errors

www.frontiersin.org

Experiments and Results

Simulation Study

In order to assess the performance of the proposed joint model with the corrected-score function, we conducted a series of simulation studies whose primary objective was to assess the fixed effect coefficient estimates and the variance of the random effects. Data are simulated in an oncology trial context, with random effects correlated among patients’ multiple endpoints. In the simulations, we assume 200 patients, i.e., i=1,,200. For each patient i, we generate the random effects bi from a normal distribution with zero mean, variance σb. We consider three distinct tumor response states CR&PR (Ri = 1), SD & PD (Ri = 0). The response data are generated based on the logistic probability, π1=F(αzTZi+αmTMBi+bi), π0=1π1. The event time for the patient is generated from the probability density function h0(t)exp(βzTZi+βmTMBi+bi)S0ɛ(t)exp(βzTZi+βmTMBi+bi), where the baseline hazard is assumed to follow the Weibull distribution with the shape parameter equal to 1.0. Censoring time C is generated from the uniform distribution U (0, 8).

Furthermore, we set αz=1.8,αm=0.3, λ=1.0,βz=2.2, and βm=0.4, σb=1.0. The variance for the error term is set to be 0.5, 0.75, and 1.0, respectively, in order to evaluate the performance of the estimators with different measurement error levels. With the specified parameters, for each dataset, based on the joint model, the true-data estimator, the naive estimator and the correct-score estimator with Monte Carlo approximation J = 10 were computed 1,000 replications. As a comparison, we also based the standard regression models; the true-data estimator and the naive estimator were computed. Results of the simulations are presented in Table 1. We report the fitted value, average bias, SD, and SE for each parameter, where SD and SE are defined as the standard error of the estimates over 1,000 simulations and the average of the standard error of the estimates, respectively.

TABLE 1
www.frontiersin.org

TABLE 1. Comparisons of bias and standard errors of estimators between joint model with standard model with varying measurement errors.

According to Table 1, the regression parameter estimates for the two function components perform reasonably well for a variety of measurement error conditions. In the absence of measurement errors, the joint model outperforms ordinary regression models in calculating regression coefficients because it more precisely reflects the potential connections between several endpoints. When considering different levels of measurement errors, the performance of the estimator based on corrected score was significantly superior to that of the naive estimator and only marginally poorer than that of the true-data estimator. Clearly, the performance of the naive estimator deteriorates with increasing error magnitude, which further suggests that the measurement error introduces a more significant bias effect on the parameter estimates. Overall, the results of the simulation experiments support the proposed joint multi-endpoint model and the iterative numerical estimation procedure, as well as the applicability of the associated random effects. Additionally, comparing SE and SD, the precision of the stated standard errors is generally satisfactory. The biases of the joint assessments compared to the standard separate regressions emphasize that the dependence among clinical endpoints could be an important and non-negligible confounder in analyzing the factors determining the treatment effect.

To further exhibit the disturbance of measurement errors on TMB thresholds and the stability of our proposed joint model, we additionally simulated the comparison of efficacy grouped by different TMB thresholds. We simulated the prognosis of a cohort of patients based on the assumption that there is a positive correlation between actual TMB levels and a favorable immunotherapy prognosis, with coefficients set exactly as above. The variance fluctuation of TMB measurement error was set to 1.0. We derive the different thresholds for classifying patients and comparing their efficacy based on the joint statistical inference with the TMB actual values, the quantile method with TMB observations, and the joint-correction statistical inference with TMB observations. The outcomes of the comparison are depicted in Figure 2. We can clearly observe that the discrepancies between the efficacies of different groups are minimized or even reversed (Figure 2C,D) when the patients were classified directly using quartiles in the presence of measurement errors. Contrary to the clinical theory that the higher TMB, the more antitumor immunogenic the patient, patients in the TMB-low subgroup demonstrated greater therapeutic benefit in terms of tumor remission and progression-free survival than those in the TMB-high subgroup. The confounding effect of the measurement errors would dilute the actual link between TMB levels and immunotherapy clinical efficacy (Figures 2A,B), preventing appropriate screening for superior patient populations. In contrast, the bias effect due to measurement errors is reduced when we use the joint model as well as the correction estimation procedure. As shown in Figure 2E,F, the TMB threshold determination based on our proposed method ensures both the validity and a certain degree of error tolerance.

FIGURE 2
www.frontiersin.org

FIGURE 2. Efficacy comparison of patients grouped based on different TMB thresholds. (A) (B) Comparison of ORR and survival curves based on the threshold derived from the joint statistical inference with TMB actual values. (C) (D) Comparison of ORR and survival curves based on median TMB observations. (E) (F) Comparison of ORR and survival curves based on the threshold derived from the joint-correction statistical inference with TMB observed values.

Patient Cohort Characteristics

Sun Yat-sen University Cancer Center recruited 95 NSCLC patients who received anti-PD-(L)1 monotherapy between December 2015 and August 2017, with data collected until January 2019. The study design has already been published (Fang et al., 2019). Between March 2016 and January 2018, R/M NPC patients have enrolled in two single-arm phase I trials (NCT02721589 and NCT02593786), where 128 patients were screened for eligibility. The dose escalation and expansion phases of the study were previously reported (Fang et al., 2018; Ma et al., 2019). Eligible patients aged from 18 to 70 years had histologically or cytologically confirmed locally advanced or metastatic NSCLC or NPC, had an Eastern Cooperative Oncology Group (ECOG) performance-status score of 0 or 1 (on a 5-point scale, with higher numbers indicating greater disability), had at least one measurable lesion according to the Response Evaluation Criteria in Solid Tumors (RECIST version 1.1 (Eisenhauer et al., 2009)), and had failed at least one prior line of systemic therapy. Figure 3 and Supplementary Table S1 depict the distribution of patients’ treatments. Radiographic tumor assessments were taken at the start of the study and every 6 weeks thereafter. The proportion of patients with complete response (CR) and partial response (PR) was known as the ORR. The time from the initial dose until PD or any-cause death was referred to as progression-free survival (PFS). Censored data documented the last radiographic assessment before cut-off, follow-up loss, or treatment change. Overall survival (OS) was defined as the time from the first dosage to death, and patients who remained alive were censored at the date of their last follow-up. The Sun Yat-sen University Cancer Center’s Ethical Review Committee approved this study, which was carried out in conformity with the Declaration of Helsinki. Each patient signed the written informed consent.

FIGURE 3
www.frontiersin.org

FIGURE 3. Patient samples included in the final analysis. (A) Flowchart for NSCLC sample inclusions. Among the 95 patients who underwent anti-PD-(L) 1 therapies and had available FFPE and/or biopsy tumor samples, we performed WES on samples from 73 patients. (B) Flowchart for NPC sample inclusions. Among the 128 patients who underwent anti-PD-(L) 1 or anti-CTLA-4 therapies, we performed targeted NGS on samples from 64 patients.

At Sun Yat-sen University Cancer Center, 95 Chinese patients with NSCLC were treated with anti-PD-(L)1 monotherapies, with 73 patients being included in the final analysis with evaluable radiological results. Concurrently, 128 patients with R/M NPC who had received anti-PD-(L)1 monotherapies were retrospectively investigated, of whom 64 patients were being screened for the final analysis based on sequencing quality and follow-up completeness. When both FFPE and biopsy samples were available for the patient, the FFPE sample was used in the analysis, given the limited intra-tumoral heterogeneity represented by a single biopsy sample. The study design and clinical characteristics of this cohort are summarized in Figure 3 and Table 2 with details in Supplementary Table S1. For lung cancer, 60% of the patients had adenocarcinoma, followed by squamous carcinoma (32%). Almost all patients (99%) were stage IV at diagnosis; the median age of patients with NSCLC and NPC at the treatment initiation was 55 and 46 years, respectively. 49% of the NSCLC patients and 25% NPC patients had a smoking history and more males in both cohorts (70% vs. 30% for NSCLC, 80% vs. 20% for NPC). ORR of the study cohorts was 19% and 12%, and the median progression-free survival (mPFS) was 91 days for lung cancer and 67.5 days for NPC. No difference in PFS was observed among the different immune agents.

TABLE 2
www.frontiersin.org

TABLE 2. Baseline clinical characteristics for NSCLC patients and NPC patients.

In addition to the SYSUCC NSCLC cohort and NPC cohort described above, external cohorts of 943 patients from the public literatures treated with ICI are compiled in Supplementary Table S2, encompassing 453 melanoma patients (Snyder et al., 2014; Van Allen et al., 2015; Hugo et al., 2016; Goodman et al., 2017), 407 NSCLC patients (Goodman et al., 2017; Hellmann et al., 2018a; Miao et al., 2018; Rizvi et al., 2018), 56 RCC (Wood et al., 2020), and 27 bladder (Miao et al., 2018), along with treatment modality and outcome analyzed. The mutation callings are derived from three sequencing platforms (WES, F1CDx, and MSK-IMPACT). F1CDx and MSK-IMPACT are NGS targeted panel being authorized by the FDA as practical diagnostic assays. Table 3 summarizes the sequencing methodology and varied TMB thresholds employed in the gathered research.

TABLE 3
www.frontiersin.org

TABLE 3. Patient cohorts from the published literatures.

Joint Model Prompts a Comprehensive and Robust TMB Subgrouping

The multi-endpoint joint analysis used to locate TMB thresholds is superior to the previous studies as it provides a more comprehensive analysis of patient clinical information. Based on the co-analyzed labels, it can give an overall picture of disease efficacy. Based on these compound indices to establish ROC curves to handle true- and false-positive rates in the classification, we selected a TMB threshold from clinically meaningful values to group patients in the experiment and validation sets.

As shown in Figure 4 and Table 4, we can discern that the ROC curves based on the mixed-endpoint joint labels generally had higher AUCs in either the experiment or validation groups, with an average improvement of about 0.2 over those based on ORR labels alone, and the range of confidence intervals likewise supports this conclusion. More importantly, all the AUCs established on the proposed indices exceeded 0.6, ranges from 0.663 to 0.972, reflecting our model’s more robust discrimination capabilities. For comparison, as for the ROCs based on original ORR labels, despite the classification ability varying among cancer types, the ROCs in most cases showed more inferiority, with half of the cases only marginally exceeding 0.5 not reaching 0.6, even equivalent to random chance. The results in Figure 4 and Table 4 fully demonstrate that the subgrouping of TMB under the joint modeling of multiple endpoints is significantly improved compared to the existing subgrouping based on the ORR single label. We attribute this phenomenon to a proportion of the patients with opposing effects on the two rubrics present in these cases. Although high TMB was reported associated with ICI treatment improvement in terms of overall trends, the status of a single indicator alone is not fully representative of the patient’s actual matter. This is why the ROC curves established based on only a single endpoint have such poor performance. Integrating patients’ multi-dimensional information and joint modeling mixed-endpoints can prompt a more comprehensive stratification of TMB. Our approach could provide clinicians with a full assessment of efficacy, resulting in a comprehensive determination of the TMB screening threshold for superior patients.

FIGURE 4
www.frontiersin.org

FIGURE 4. Receiver operating characteristic curves of the predictive capacity of prognosis label for two experiment cohorts and validation cohorts, depicting the true-positive rate (sensitivity, y-axis) and false-positive rate (1-specificity, x-axis) for the metric across all possible TMB thresholds. The corresponding area under the curve (AUC) is illustrated in the figure legends. (A) ROC curves for NPC (panel) experiment cohort, bladder cohort, and RCC cohort based on ORR labels alone. (B) ROC curves for NSCLC (WES) experiment cohort, NSCLC_35 cohort, NSCLC_57 cohort, and NSCLC_240 cohort based on ORR labels alone. (C) ROC curves for Mel_37 cohort, Mel_52 cohort, Mel_64 cohort, and Mel_105 cohort based on ORR labels alone. (D) ROC curves for NPC (panel) experiment cohort, bladder cohort, and RCC cohort based on the mixed-endpoint labels. (E) ROC curves for NSCLC (WES) experiment cohort, NSCLC_35 cohort, NSCLC_57 cohort, and NSCLC_240 cohort based on the mixed-endpoint labels. (F) ROC curves for Mel_37 cohort, Mel_52 cohort, Mel_64 cohort, and Mel_105 cohort based on the mixed-endpoint labels.

TABLE 4
www.frontiersin.org

TABLE 4. AUC comparison. The table reports the area under the curve (AUC), as well as the corresponding 0.95 confidence interval, for each metric (columns) applied to a different cancer cohort (rows). Bold-faced values indicate the best value for each cancer cohort.

To verify that our proposed threshold delineation method for TMB remains valid and robust under the perturbation of measurement errors, we added 10%–20% artificial noise according to the actual TMB level. Given the small number of patients in some cases, which are over-sensitive to data noise, we selected several groups of cases with more patients for analysis. The results are shown in Figure 5 and Table 5. Under the perturbation of artificial noise, the AUC of each group showed mostly a slight decrease compared to the error-free cases. However, the ROC curves based on our proposed joint labels still maintain a high AUC, which is about 0.3 higher on average than the ROC curves based on ORR labels only. These results demonstrate the high error tolerance of our proposed joint model.

FIGURE 5
www.frontiersin.org

FIGURE 5. Receiver operating characteristic curves of the predictive capacity of prognosis label for two experiment cohorts and validation cohorts, depicting the true-positive rate (sensitivity, y-axis) and false-positive rate (1-specificity, x-axis) for the metric across all possible TMB thresholds considering measurement errors. The corresponding area under the curve (AUC) is illustrated in the figure legends. (A) ROC curves for NPC (Panel), NSCLC (WES) experiment cohort, Mel_64 cohort, Mel_105 cohort, and NSCLC_240 cohort based on ORR labels considering TMB measurement errors. (B) ROC curves for NPC (panel), NSCLC (WES) experiment cohort, Mel_64 cohort, Mel_105 cohort, and NSCLC_240 cohort based on the mixed-endpoint labels considering TMB measurement errors.

TABLE 5
www.frontiersin.org

TABLE 5. AUC comparison. The table reports the area under the curve (AUC), as well as the corresponding 0.95 confidence interval, for each metric (columns) applied to a different cancer cohort (rows). Bold-faced values indicate the best value for each cancer cohort.

Joint Analysis Prompts a Significant and Error-Tolerant Patient Subgrouping

In addition to the strengths shown in the ROC curves, based on the derived TMB thresholds, we can classify experimental NSCLC patients into two groups with apparently stratified efficacy. The effect of the dichotomy is shown in Figure 6, where we can notice a significant difference between patients in TMB_Low and TMB_High in terms of immunotherapy benefit (p-values = 0.017 and 0.089). The grouping results on the other cohorts can be seen in Supplementary Figures.

FIGURE 6
www.frontiersin.org

FIGURE 6. Survival curves and ORR comparison between experimental NSCLC patients (n = 73) with low and high TMB. Improved progression-free survival (PFS) and a trend toward increased objective response rate (ORR) are observed in patients with high TMB.

To demonstrate that the TMB thresholds derived from our proposed joint model can significantly separate the treatment effects of patients receiving immunotherapy, we statistically compared the patient outcomes obtained based on our thresholds with those obtained from different quartiles (median, upper tertile, upper quartile) using the log-rank test and the two-sided Mann–Whitney U test. As shown by the results in Table 6, our model-derived TMB thresholds performed satisfactorily and consistently for cohort patient segmentation. This predominance is mainly reflected in the p-values of the statistical tests, which are essentially the lowest among all division scenarios under the threshold division based on the joint model, indicating that the proposed model is predominate. NSCLC_35 and NSCLC_240 were the only two situations in which the p-values performed marginally worse than the quantile divisions. Similarly, five groups of patients were selected to validate the stability of the proposed model in the face of the TMB measurement error. As shown by the results in Table 7, our proposed threshold delineation method for TMB remained efficient and robust under perturbation of measurement error.

TABLE 6
www.frontiersin.org

TABLE 6. Immunotherapy mPFS or mOS and response probability based on different tumor mutation burden (TMB) thresholds for non-small-cell lung cancer (NSCLC), nasopharyngeal carcinoma (NPC), bladder, renal cell carcinoma (RCC), and melanoma. p values are reported by log-rank test and the two-sided Mann–Whitney U test.

TABLE 7
www.frontiersin.org

TABLE 7. Immunotherapy mPFS or mOS and response probability based on different tumor mutation burden (TMB) thresholds with measurement errors for non-small-cell lung cancer (NSCLC), nasopharyngeal carcinoma (NPC), bladder, renal cell carcinoma (RCC), and melanoma. p values are reported by log-rank test and the two-sided Mann–Whitney U test.

Discussion

Tumor mutation burden has recently become an area of interest, as high TMB is associated with improved response to ICI therapies. However, the threshold defining the TMB-high/TMB-positive patients is controversial in clinical, which is exacerbated by the presence of multiple evaluation metrics and TMB calculation errors. The existing TMB threshold-identifying approaches are merely based on a single endpoint, which may suffer from excessive information loss. TMB metric, as a predictive marker, is closely associated with both of the two types of clinical endpoints (ORR and TTE), where the effect in two endpoints may be of different magnitude or even point in different directions. Herein, we report a generalized framework for comprehensively determining the positivity TMB thresholds based on a mixed-endpoint joint model and an iterative numerical estimation procedure considering measurement errors. In our joint model, we choose the Weibull–Cox proportional hazard model for the TTE endpoint. Although the baseline risk h0(t) in standard survival analysis usually be left unspecified, such as the advantageous partial likelihood method. However, within the joint modeling framework, it turns out that following such a route may lead to an underestimation of the standard errors of the parameter estimates (Hsieh et al., 2006). Thus, we recommend choosing an explicit definition of h0(t) based on the dataset characteristics, corresponding to a parametric distribution. The Weibull, the log–normal, and the Gamma distributions are typically employed in the survival analysis context. By analyzing the progression-free survival of patients receiving immunotherapy, we found that the trend of their baseline cumulative hazard distribution was consistent with the Weibull distribution with a scale parameter equal to 1 (see in Figure 1), so the Weibull–Cox proportional hazard model was employed in this article. Our joint model sheds new light on the tumor mutation burden stratification based on a multi-endpoint assessment of immunotherapy benefits, suggesting more comprehensive and robust TMB-positive thresholds for clinical physicians. Attending physicians should make treatment recommendations based on patients’ multi-dimensional information.

Conclusion

The existing statistical methods for determining TMB thresholds are based on a single clinical endpoint while ignoring the difference between the true and observed TMB values. Our study considers TMB measurement error and integrates multifaceted clinical efficacy to optimize TMB thresholds. We report a multi-endpoint joint model as a generalized method for inferring TMB thresholds that facilitates consistent statistical inference using an iterative numerical estimation procedure considering mis-specified TMB. Our simulation results show that the proposed model maintains higher accuracy and stability than standard regressions, in terms of both parameter estimation and threshold determination. To validate the feasibility of the proposed thresholds, we pooled a cohort of 73 patients with non-small-cell lung cancer and 64 patients with nasopharyngeal carcinoma treated with anti-PD-(L)1, as well as a validation cohort of 943 patients for retrospective analysis. From the simulation and experimental results, we reasonably conclude that 1) our proposed joint model with the parameter estimation procedure can more robustly assess patient efficacy even under the interference of measurement error in TMB. 2) Integrating patients’ multi-dimensional information to employ multi-endpoint efficacy analysis can prompt a more comprehensive TMB subgrouping. 3) The TMB-positive threshold derived from multi-endpoint joint analysis can classify patients into two groups with more apparently stratified efficacy. Our model is applicable to clinical multiple endpoint data and can better assist physicians in their clinical decisions.

Data Availability Statement

The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found in the article/Supplementary Material.

Ethics Statement

The studies involving human participants were reviewed and approved by the Sun Yat-Sen University Cancer Center’s Ethical Review Committee. The patients/participants provided their written informed consent to participate in this study.

Author Contributions

YW, XL, JW and WF conceived and designed the study. YW, XL and JW developed the methodology. WF, YS, LZ, YW, XL and YX collected and managed the data. YW wrote the first draft. YW, XL, JW, LZ and WF reviewed, edited, and approved the manuscript. XL, JW, YX, XPZ, XYZ, YL, LZ and WF provided administrative, technical, or material support. JW was primarily responsible for the final manuscript.

Funding

This work was funded by the National Natural Science Foundation of China, grant number 92046009, the Natural Science Basic Research Program of Shaanxi, grant number 2020JC-01, the National Natural Science Foundation of China, grant number 82173101, and the National Natural Science Foundation of China, grant number 81972556.

Conflict of Interest

The author YS was employed by the company Nanjing Geneseeq Technology Inc.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors, and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

The authors thank the patients and their families for participation in the study and the reviewer for the constructive and valuable suggestions.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fgene.2022.915839/full#supplementary-material

References

Alioto, T. S., Buchhalter, I., Derdak, S., Hutter, B., Eldridge, M. D., Hovig, E., et al. (2015). A Comprehensive Assessment of Somatic Mutation Detection in Cancer Using Whole-Genome Sequencing. Nat. Commun. 6, 10001. doi:10.1038/ncomms10001

PubMed Abstract | CrossRef Full Text | Google Scholar

Asar, Ö., Ritchie, J., Kalra, P. A., and Diggle, P. J. (2015). Joint Modelling of Repeated Measurement and Time-To-Event Data: an Introductory Tutorial. Int. J. Epidemiol. 44 (1), 334–344. doi:10.1093/ije/dyu262

PubMed Abstract | CrossRef Full Text | Google Scholar

Augustin, T. (2004). An Exact Corrected Log-Likelihood Function for Cox's Proportional Hazards Model under Measurement Error and Some Extensions. Scand. J. Stat. 31 (1), 43–50. doi:10.1111/j.1467-9469.2004.00371.x

CrossRef Full Text | Google Scholar

Bai, R., Lv, Z., Xu, D., and Cui, J. (2020). Predictive Biomarkers for Cancer Immunotherapy with Immune Checkpoint Inhibitors. Biomark. Res. 8, 34. doi:10.1186/s40364-020-00209-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Barbieri, A., and Legrand, C. (2020). Joint Longitudinal and Time-To-Event Cure Models for the Assessment of Being Cured. Stat. Methods Med. Res. 29 (4), 1256–1270. doi:10.1177/0962280219853599

PubMed Abstract | CrossRef Full Text | Google Scholar

Boyiadzis, M. M., Kirkwood, J. M., Marshall, J. L., Pritchard, C. C., Azad, N. S., and Gulley, J. L. (2018). Significance and Implications of FDA Approval of Pembrolizumab for Biomarker-Defined Disease. J. Immunother. cancer 6 (1), 35. doi:10.1186/s40425-018-0342-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Bracarda, S., Altavilla, A., Hamzaj, A., Sisani, M., Marrocolo, F., Del Buono, S., et al. (2015). Immunologic Checkpoints Blockade in Renal Cell, Prostate, and Urothelial Malignancies. Seminars Oncol. 42 (3), 495–505. doi:10.1053/j.seminoncol.2015.02.004

CrossRef Full Text | Google Scholar

Campesato, L. F., Barroso-Sousa, R., Jimenez, L., Correa, B. R., Sabbaga, J., Hoff, P. M., et al. (2015). Comprehensive Cancer-Gene Panels Can Be Used to Estimate Mutational Load and Predict Clinical Benefit to PD-1 Blockade in Clinical Practice. Oncotarget 6 (33), 34221–34227. doi:10.18632/oncotarget.5950

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, D., Xu, H., Xu, X., Guo, T., and Ge, W. (2019). High Tumor Mutation Burden Predicts Better Efficacy of Immunotherapy: a Pooled Analysis of 103078 Cancer Patients. Oncoimmunology 8 (9), e1629258. doi:10.1080/2162402X.2019.1629258

PubMed Abstract | CrossRef Full Text | Google Scholar

Carbone, D. P., Reck, M., Paz-Ares, L., Creelan, B., Horn, L., Steins, M., et al. (2017). First-Line Nivolumab in Stage IV or Recurrent Non-small-cell Lung Cancer. N. Engl. J. Med. 376 (25), 2415–2426. doi:10.1056/NEJMoa1613493

PubMed Abstract | CrossRef Full Text | Google Scholar

Carroll, R. J., Ruppert, D., Stefanski, L. A., and Crainiceanu, C. M. (2006). Measurement Error in Nonlinear Models: A Modern Perspective. London, UK: Chapman and Hall/CRC.

Google Scholar

Chalmers, Z. R., Connelly, C. F., Fabrizio, D., Gay, L., Ali, S. M., Ennis, R., et al. (2017). Analysis of 100,000 Human Cancer Genomes Reveals the Landscape of Tumor Mutational Burden. Genome Med. 9 (1), 34. doi:10.1186/s13073-017-0424-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Chiang, A. C., and Herbst, R. S. (2020). Frontline Immunotherapy for NSCLC - the Tale of the Tail. Nat. Rev. Clin. Oncol. 17 (2), 73–74. doi:10.1038/s41571-019-0317-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Colli, L. M., Machiela, M. J., Myers, T. A., Jessop, L., Yu, K., and Chanock, S. J. (2016). Burden of Nonsynonymous Mutations Among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. Cancer Res. 76 (13), 3767–3772. doi:10.1158/0008-5472.CAN-16-0170

PubMed Abstract | CrossRef Full Text | Google Scholar

Conway, J. R., Kofman, E., Mo, S. S., Elmarakeby, H., and Van Allen, E. (2018). Genomics of Response to Immune Checkpoint Therapies for Cancer: Implications for Precision Medicine. Genome Med. 10 (1), 93. doi:10.1186/s13073-018-0605-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Cristescu, R., Mogg, R., Ayers, M., Albright, A., Murphy, E., Yearley, J., et al. (2018). Pan-tumor Genomic Biomarkers for PD-1 Checkpoint Blockade-Based Immunotherapy. Science 362 (6411), eaar3593. doi:10.1126/science.aar3593

PubMed Abstract | CrossRef Full Text | Google Scholar

Davis, P. J., and Rabinowitz, P. (2007). Methods of Numerical Integration. Chelmsford, MA, USA: Courier Corporation.

Google Scholar

Eisenhauer, E. A., Therasse, P., Bogaerts, J., Schwartz, L. H., Sargent, D., Ford, R., et al. (2009). New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline (Version 1.1). Eur. J. Cancer 45 (2), 228–247. doi:10.1016/j.ejca.2008.10.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, W., Ma, Y., Yin, J. C., Hong, S., Zhou, H., Wang, A., et al. (2019). Comprehensive Genomic Profiling Identifies Novel Genetic Predictors of Response to Anti-PD-(L)1 Therapies in Non-small Cell Lung Cancer. Clin. Cancer Res. 25 (16), 5015–5026. doi:10.1158/1078-0432.CCR-19-0585

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, W., Yang, Y., Ma, Y., Hong, S., Lin, L., He, X., et al. (2018). Camrelizumab (SHR-1210) Alone or in Combination with Gemcitabine Plus Cisplatin for Nasopharyngeal Carcinoma: Results from Two Single-Arm, Phase 1 Trials. Lancet Oncol. 19 (10), 1338–1350. doi:10.1016/S1470-2045(18)30495-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Goodman, A. M., Kato, S., Bazhenova, L., Patel, S. P., Frampton, G. M., Miller, V., et al. (2017). Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 16 (11), 2598–2608. doi:10.1158/1535-7163.MCT-17-0386

PubMed Abstract | CrossRef Full Text | Google Scholar

Guidance, D. (2017). Multiple Endpoints in Clinical Trials Guidance for Industry. White Oak: Center for Biologics Evaluation and Research (CBER.

Google Scholar

Hellmann, M. D., Callahan, M. K., Awad, M. M., Calvo, E., Ascierto, P. A., Atmaca, A., et al. (2018a). Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell. 33 (5), 853–861. e4. doi:10.1016/j.ccell.2018.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Hellmann, M. D., Ciuleanu, T.-E., Pluzanski, A., Lee, J. S., Otterson, G. A., Audigier-Valette, C., et al. (2018b). Nivolumab Plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N. Engl. J. Med. 378 (22), 2093–2104. doi:10.1056/NEJMoa1801946

PubMed Abstract | CrossRef Full Text | Google Scholar

Hellmann, M. D., Nathanson, T., Rizvi, H., Creelan, B. C., Sanchez-Vega, F., Ahuja, A., et al. (2018c). Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-small-cell Lung Cancer. Cancer Cell. 33 (5), 843–852. e4. doi:10.1016/j.ccell.2018.03.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Hsieh, F., Tseng, Y.-K., and Wang, J.-L. (2006). Joint Modeling of Survival and Longitudinal Data: Likelihood Approach Revisited. Biometrics 62 (4), 1037–1043. doi:10.1111/j.1541-0420.2006.00570.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Hugo, W., Zaretsky, J. M., Sun, L., Song, C., Moreno, B. H., Hu-Lieskovan, S., et al. (2016). Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell. 165 (1), 35–44. doi:10.1016/j.cell.2016.02.065

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, D. B., Frampton, G. M., Rioth, M. J., Yusko, E., Xu, Y., Guo, X., et al. (2016). Targeted Next Generation Sequencing Identifies Markers of Response to PD-1 Blockade. Cancer Immunol. Res. 4 (11), 959–967. doi:10.1158/2326-6066.CIR-16-0143

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuryk, L., Bertinato, L., Staniszewska, M., Pancer, K., Wieczorek, M., Salmaso, S., et al. (2020). From Conventional Therapies to Immunotherapy: Melanoma Treatment in Review. Cancers 12 (10), 3057. doi:10.3390/cancers12103057

CrossRef Full Text | Google Scholar

Legrand, F. A., Gandara, D. R., Mariathasan, S., Powles, T., He, X., Zhang, W., et al. (2018). Association of High Tissue Tmb and Atezolizumab Efficacy across Multiple Tumor Types. J. Clin. Oncol. 36 (15_Suppl. l), 12000. doi:10.1200/jco.2018.36.15_suppl.12000

CrossRef Full Text | Google Scholar

Lemery, S., Keegan, P., and Pazdur, R. (2017). First FDA Approval Agnostic of Cancer Site - when a Biomarker Defines the Indication. N. Engl. J. Med. 377 (15), 1409–1412. doi:10.1056/NEJMp1709968

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, C., Tsao, M. S., Le, L. W., Shepherd, F. A., Feld, R., Burkes, R. L., et al. (2015). Biomarker Testing and Time to Treatment Decision in Patients with Advanced Nonsmall-Cell Lung Cancer. Ann. Oncol. 26 (7), 1415–1421. doi:10.1093/annonc/mdv208

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, X., Taylor, J. M. G., and Ye, W. (2008). A Penalized Likelihood Approach to Joint Modeling of Longitudinal Measurements and Time-To-Event Data. Statistics its Interface 1 (1), 33–45. doi:10.4310/sii.2008.v1.n1.a4

CrossRef Full Text | Google Scholar

Ma, Y., Fang, W., Zhang, Y., Yang, Y., Hong, S., Zhao, Y., et al. (2019). A Phase I/II Open-Label Study of Nivolumab in Previously Treated Advanced or Recurrent Nasopharyngeal Carcinoma and Other Solid Tumors. Oncologist 24 (7), 891–e431. doi:10.1634/theoncologist.2019-0284

PubMed Abstract | CrossRef Full Text | Google Scholar

Majc, B., Novak, M., Kopitar-Jerala, N., Jewett, A., and Breznik, B. (2021). Immunotherapy of Glioblastoma: Current Strategies and Challenges in Tumor Model Development. Cells 10 (2), 265. doi:10.3390/cells10020265

PubMed Abstract | CrossRef Full Text | Google Scholar

Miao, D., Margolis, C. A., Vokes, N. I., Liu, D., Taylor-Weiner, A., Wankowicz, S. M., et al. (2018). Genomic Correlates of Response to Immune Checkpoint Blockade in Microsatellite-Stable Solid Tumors. Nat. Genet. 50 (9), 1271–1281. doi:10.1038/s41588-018-0200-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Motzer, R. J., Escudier, B., McDermott, D. F., George, S., Hammers, H. J., Srinivas, S., et al. (2015). Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 373 (19), 1803–1813. doi:10.1056/NEJMoa1510665

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakamura, T. (1990). Corrected Score Function for Errors-In-Variables Models: Methodology and Application to Generalized Linear Models. Biometrika 77 (1), 127–137. doi:10.1093/biomet/77.1.127

CrossRef Full Text | Google Scholar

Novick, S. J., and Stefanski, L. A. (2002). Corrected Score Estimation via Complex Variable Simulation Extrapolation. J. Am. Stat. Assoc. 97 (458), 472–481. doi:10.1198/016214502760047005

CrossRef Full Text | Google Scholar

Pardoll, D. M. (2012). The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat. Rev. Cancer 12 (4), 252–264. doi:10.1038/nrc3239

PubMed Abstract | CrossRef Full Text | Google Scholar

Phillips, A., and Haudiquet, V. (2003). ICH E9 Guideline ?Statistical Principles for Clinical Trials? a Case Study. Stat. Med. 22 (1), 1–11. discussion 13-7. doi:10.1002/sim.1328

PubMed Abstract | CrossRef Full Text | Google Scholar

Riaz, N., Havel, J. J., Makarov, V., Desrichard, A., Urba, W. J., Sims, J. S., et al. (2017). Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell. 171 (4), 934–949. e16. doi:10.1016/j.cell.2017.09.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Ristl, R., Urach, S., Rosenkranz, G., and Posch, M. (2019). Methods for the Analysis of Multiple Endpoints in Small Populations: A Review. J. Biopharm. Statistics 29 (1), 1–29. doi:10.1080/10543406.2018.1489402

CrossRef Full Text | Google Scholar

Rizopoulos, D. (2011). Dynamic Predictions and Prospective Accuracy in Joint Models for Longitudinal and Time-To-Event Data. Biometrics 67 (3), 819–829. doi:10.1111/j.1541-0420.2010.01546.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizopoulos, D., and Lesaffre, E. (2014). Introduction to the Special Issue on Joint Modelling Techniques. Stat. Methods Med. Res. 23 (1), 3–10. doi:10.1177/0962280212445800

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizvi, H., Sanchez-Vega, F., La, K., Chatila, W., Jonsson, P., Halpenny, D., et al. (2018). Molecular Determinants of Response to Anti-programmed Cell Death (PD)-1 and Anti-programmed Death-Ligand 1 (PD-L1) Blockade in Patients with Non-small-cell Lung Cancer Profiled with Targeted Next-Generation Sequencing. J. Clin. Oncol. 36 (7), 633–641. doi:10.1200/JCO.2017.75.3384

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizvi, N. A., Hellmann, M. D., Snyder, A., Kvistborg, P., Makarov, V., Havel, J. J., et al. (2015). Mutational Landscape Determines Sensitivity to PD-1 Blockade in Non-small Cell Lung Cancer. Science 348 (6230), 124–128. doi:10.1126/science.aaa1348

PubMed Abstract | CrossRef Full Text | Google Scholar

Rosenberg, J. E., Hoffman-Censits, J., Powles, T., van der Heijden, M. S., Balar, A. V., Necchi, A., et al. (2016). Atezolizumab in Patients with Locally Advanced and Metastatic Urothelial Carcinoma Who Have Progressed Following Treatment with Platinum-Based Chemotherapy: a Single-Arm, Multicentre, Phase 2 Trial. Lancet 387 (10031), 1909–1920. doi:10.1016/S0140-6736(16)00561-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Samstein, R. M., Lee, C.-H., Shoushtari, A. N., Hellmann, M. D., Shen, R., Janjigian, Y. Y., et al. (2019). Tumor Mutational Load Predicts Survival after Immunotherapy across Multiple Cancer Types. Nat. Genet. 51 (2), 202–206. doi:10.1038/s41588-018-0312-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Singal, G., Miller, P. G., Agarwala, V., Li, G., Kaushik, G., Backenroth, D., et al. (2019). Association of Patient Characteristics and Tumor Genomics with Clinical Outcomes Among Patients with Non-small Cell Lung Cancer Using a Clinicogenomic Database. JAMA 321 (14), 1391–1399. doi:10.1001/jama.2019.3241

PubMed Abstract | CrossRef Full Text | Google Scholar

Snyder, A., Makarov, V., Merghoub, T., Yuan, J., Zaretsky, J. M., Desrichard, A., et al. (2014). Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 371 (23), 2189–2199. doi:10.1056/NEJMoa1406498

PubMed Abstract | CrossRef Full Text | Google Scholar

Subbiah, V., Solit, D. B., Chan, T. A., and Kurzrock, R. (2020). The FDA Approval of Pembrolizumab for Adult and Pediatric Patients with Tumor Mutational Burden (TMB) ≥10: a Decision Centered on Empowering Patients and Their Physicians. Ann. Oncol. 31 (9), 1115–1118. doi:10.1016/j.annonc.2020.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Allen, E. M., Miao, D., Schilling, B., Shukla, S. A., Blank, C., Zimmer, L., et al. (2015). Genomic Correlates of Response to CTLA-4 Blockade in Metastatic Melanoma. Science 350 (6257), 207–211. doi:10.1126/science.aad0095

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Rooij, N., van Buuren, M. M., Philips, D., Velds, A., Toebes, M., Heemskerk, B., et al. (2013). Tumor Exome Analysis Reveals Neoantigen-specific T-Cell Reactivity in an Ipilimumab-Responsive Melanoma. J. Clin. Oncol. 31 (32), e439–e442. doi:10.1200/JCO.2012.47.7521

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Ge, J., Lan, Y., Shi, Y., Luo, Y., Tan, Y., et al. (2020). Tumor Mutational Burden Is Associated with Poor Outcomes in Diffuse Glioma. BMC Cancer 20 (1), 213. doi:10.1186/s12885-020-6658-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Wołącewicz, M., Hrynkiewicz, R., Grywalska, E., Suchojad, T., Leksowski, T., Roliński, J., et al. (2020). Immunotherapy in Bladder Cancer: Current Methods and Future Perspectives. Cancers 12 (5), 1181. doi:10.3390/cancers12051181

CrossRef Full Text | Google Scholar

Wood, M. A., Weeder, B. R., David, J. K., Nellore, A., and Thompson, R. F. (2020). Burden of Tumor Mutations, Neoepitopes, and Other Variants Are Weak Predictors of Cancer Immunotherapy Response and Overall Survival. Genome Med. 12 (1), 33. doi:10.1186/s13073-020-00729-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, H., DiCarlo, J., Satya, R. V., Peng, Q., and Wang, Y. (2014). Comparison of Somatic Mutation Calling Methods in Amplicon and Whole Exome Sequence Data. BMC Genomics 15, 244. doi:10.1186/1471-2164-15-244

PubMed Abstract | CrossRef Full Text | Google Scholar

Yarchoan, M., Hopkins, A., and Jaffee, E. M. (2017). Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 377 (25), 2500–2501. doi:10.1056/NEJMc1713444

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: clinical immunology, stratification biomarker, tumor mutation burden, joint modeling, multiple endpoints, measurement error

Citation: Wang Y, Lai X, Wang J, Xu Y, Zhang X, Zhu X, Liu Y, Shao Y, Zhang L and Fang W (2022) A Joint Model Considering Measurement Errors for Optimally Identifying Tumor Mutation Burden Threshold. Front. Genet. 13:915839. doi: 10.3389/fgene.2022.915839

Received: 08 April 2022; Accepted: 13 June 2022;
Published: 04 August 2022.

Edited by:

Jian Song, University Hospital Münster, Germany

Reviewed by:

Song Cao, Washington University in St. Louis, United States
Hongmin Cai, South China University of Technology, China

Copyright © 2022 Wang, Lai, Wang, Xu, Zhang, Zhu, Liu, Shao, Zhang and Fang. 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.

*Correspondence: Jiayin Wang, wangjiayin@mail.xjtu.edu.cn; Wenfeng Fang, fangwf@sysucc.org.cn

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.