J Gastric Cancer. 2023 Jul;23(3):476-486. English.
Published online Jul 24, 2023.
Copyright © 2023. Korean Gastric Cancer Association
Original Article

The Optimal Tumor Mutational Burden Cutoff Value as a Novel Marker for Predicting the Efficacy of Programmed Cell Death-1 Checkpoint Inhibitors in Advanced Gastric Cancer

Jae Yeon Jang,1,2 Youngkyung Jeon,2 Sun Young Jeong,2 Sung Hee Lim,2 Won Ki Kang,2 Jeeyun Lee,2 and Seung Tae Kim2
    • 1Division of Hematology-Oncology, Department of Internal Medicine, Wonju Severance Christian Hospital, Wonju, Korea.
    • 2Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
Received April 11, 2023; Revised July 08, 2023; Accepted July 09, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Purpose

The optimal tumor mutational burden (TMB) value for predicting treatment response to programmed cell death-1 (PD-1) checkpoint inhibitors in advanced gastric cancer (AGC) remains unclear. We aimed to investigate the optimal TMB cutoff value that could predict the efficacy of PD-1 checkpoint inhibitors in AGC.

Materials and Methods

Patients with AGC who received pembrolizumab or nivolumab between October 1, 2020, and July 27, 2021, at Samsung Medical Center in Korea were retrospectively analyzed. The TMB levels were measured using a next-generation sequencing assay. Based on receiver operating characteristic curve analysis, the TMB cutoff value was determined.

Results

A total 53 patients were analyzed. The TMB cutoff value for predicting the overall response rate (ORR) to PD-1 checkpoint inhibitors was defined as 13.31 mutations per megabase (mt/Mb) with 56% sensitivity and 95% specificity. Based on this definition, 7 (13.2%) patients were TMB-high (TMB-H). The ORR differed between the TMB-low (TMB-L) and TMB-H (8.7% vs. 71.4%, P=0.001). The progression-free survival and overall survival (OS) for 53 patients were 1.93 (95% confidence interval [CI], 1.600–2.268) and 4.26 months (95% CI, 2.992–5.532). The median OS was longer in the TMB-H (20.8 months; 95% CI, 2.292–39.281) than in the TMB-L (3.31 months; 95% CI, 1.604–5.019; P=0.049).

Conclusions

The TMB cutoff value for predicting treatment response in AGC patients who received PD-1 checkpoint inhibitor monotherapy as salvage treatment was 13.31 mt/Mb. When applying the programmed death ligand-1 status to TMB-H, patients who would benefit from PD-1 checkpoint inhibitors can be selected.

Keywords
Gastric cancer; Immune checkpoint inhibitor; PD-1 inhibitor

INTRODUCTION

Programmed cell death-1 (PD-1) checkpoint inhibitors have proven to be effective in the treatment of locally advanced unresectable or metastatic gastric cancer (advanced gastric cancer [AGC]). Nivolumab combined with capecitabine or 5-fluorouracil plus oxaliplatin (XELOX or FOLFOX) is currently indicated as the first-line treatment for programmed death ligand-1 (PD-L1) combined positive score (CPS) ≥5 and human epidermal growth factor receptor 2-negative AGC [1, 2]. In Korea, pembrolizumab was approved as the second-line or subsequent treatment for microsatellite instability-high (MSI-H) or mismatch repair-deficient (dMMR) AGC based on the KEYNOTE-158 trial [3, 4]. Nivolumab monotherapy is also recommended as a third-line therapy based on a randomized phase 3 ATTRACTION-2 trial [2, 5].

However, the predictive markers for the efficacy of PD-1 checkpoint inhibitor monotherapy in AGC have not yet been sufficiently established [6, 7, 8]. There is no clear standard for identifying patients who are likely to benefit from PD-1 checkpoint inhibitors does not exist. Although PD-L1 and MSI are considered novel markers for PD-1 checkpoint inhibitors, their roles have not yet been determined [9, 10, 11].

Recent advances in whole-exome sequencing (WES) and next-generation sequencing (NGS) of multiple genes have defined the tumor biology of AGC [12, 13, 14]. Furthermore, molecular targeted therapies and immunotherapies, including PD-1 checkpoint inhibitors, have been developed based on the NGS findings [14, 15, 16, 17]. In addition, a high level of tumor mutational burden (TMB), the total number of non-synonymous mutations in the tumor-coding regions, is a novel predictive marker for immunotherapy or a prognostic marker in various tumor types [18, 19, 20]. Some somatic mutations in tumor DNA are associated with tumor neoantigens, which are targets of the immune system [21, 22, 23]. Therefore, tumors with a high TMB are likely to have more neoantigens and respond better to immunotherapies that activate T cell immunity. Although TMB is regarded as a biomarker for the response to immunotherapy in several solid tumors [24, 25, 26], the optimal value for defining TMB-high (TMB-H) is insufficient for AGC.

Therefore, in the present study, the optimal TMB cutoff value that could predict the efficacy of PD-1 checkpoint inhibitor monotherapy as a salvage treatment for AGC was determined.

MATERIALS AND METHODS

Patients

This retrospective study included patients with AGC who received pembrolizumab or nivolumab between October 1, 2020, and July 27, 2021, at Samsung Medical Center in Korea. All patients underwent a tissue biopsy, and tissues were used for NGS diagnostic platform, including TMB, with the Illumina’s TruSight™ Oncology 500 (TSO 500) assay (Illumina Inc., San Diego, CA, USA). Clinicopathological characteristics were collected from electronic medical records. PD-L1 was detected using the immunohistochemical assay PD-L1 antibody 22C3 pharmDx. Tumors with a CPS of ≥1 were defined as positive PD-L1 expression. Epstein-Barr virus was detected using in situ hybridization. The Institutional Review Board (IRB) of Samsung Seoul Medical Center approved this study (IRB No. 2022-12-068), which was conducted in accordance with the Declaration of Helsinki.

NGS and TMB calculation

The tumor samples were obtained at the time of diagnosis of advanced or metastatic AGC, and tissues were formalin-fixed paraffin-embedded. The TSO 500 Kit was used for DNA library preparation and enrichment following the manufacturer’s instructions. Post-enrichment libraries were quantified, pooled, and sequenced using TSO 500 (Illumina Inc.). The quality of TSO 500 sequencing runs was assessed using an Illumina Sequencing Analysis Viewer. Sequencing data were analyzed using TSO 500 Local App version 1.3.0.39. The TSO 500 is a comprehensive tumor profiling assay that measures biomarkers, including single nucleotide variants, copy number variants, indels, fusions, splice variants, TMB, and MSI. The TMB scores are reported as mutations per megabase (mt/Mb) sequenced and TMB levels.

Defining the TMB cutoff value

The TMB was measured using an NGS-based assay and reported as mt/Mb sequenced. Receiver operating characteristic (ROC) curve analysis was used to measure the diagnostic ability of TMB to predict the overall response rate (ORR) of anti-PD-1 therapy. The optimal TMB cutoff value was determined as the point at which Youden’s index was the maximum, defined as follows [27, 28, 29].

Youden’s Index J = Sensitivity + Specificity – 1.

Statistical analysis

Treatment response to anti-PD-1 therapy was evaluated radiologically based on the Response Evaluation Criteria in Solid Tumors guidelines, version 1.1.

Descriptive statistics were used to summarize the patient and tumor characteristics and treatment histories. Associations between TMB status and tumor response, including the ORR and disease control rate (DCR), were analyzed using t-test, and categorical variables were analyzed using the χ2 test and Fisher’s exact test. The progression-free survival (PFS) and overall survival (OS) were analyzed using the Kaplan-Meier method, with medians and corresponding 95% confidence intervals (CIs). The log-rank test was used to compare the differences in PFS and OS between the TMB-H and TMB-low (TMB-L) groups. The P-values of <0.05 were considered to indicate statistical significance. All statistical analyses were performed using SPSS version 25 (IBM Corp., Armonk, NY, USA).

RESULTS

Patient characteristics

A total of 53 patients with AGC with quantifiable TMB were analyzed in this study. The median age at diagnosis was 56 years, and 60.4% of the patients were female. Overall, patients were treated with pembrolizumab (n=21, 39.6%) or nivolumab (n=32, 60.4%). The median TMB of patients was 6.3 mt/Mb and ranged from 0.8–107.1 mt/Mb. Four (7.5%) patients harbored MSI-H tumors, and PD-L1 expression was detected in 28 of 39 (71.8%) patients (Table 1).

Definition of the TMB cutoff value

Based on the ROC curve and Youden’s index analyses, the area under the ROC curve was 0.7285, indicating excellent discrimination. As shown in Fig. 1, the optimal TMB cutoff value that showed the maximum sensitivity and specificity for predicting the ORR of immunotherapy in AGC was ≥13.31 mt/Mb. Based on this cutoff value, the sensitivity and specificity were 55.6% and 95.5%, respectively. Based on this definition of the TMB cutoff value, 46 (86.8%) patients were TMB-L, and 7 (13.2%) were TMB-H. The incidence rates of MSI and TMB are shown in Table 2.

Fig. 1
Receiver operating characteristic curve for predicting the overall response rate of immunotherapy in advanced gastric cancer. The tumor mutational burden cut-off value with maximum sensitivity and specificity is 13.31 mt/Mb, representing sensitivity of 55.6% and specificity of 95.5%.

Comparison of the efficacy of ICIs based on the TMB status

The ORR for all enrolled patients was 17% (95% CI, 8.1%–29.8%). Patients with TMB-H showed a better tumor response than those with TMB-L. The ORR to PD-1 checkpoint inhibitors in patients with TMB-H was 71.4% (5 of 7 with partial response; 95% CI, 29.0–96.3), and the ORR in patients with TMB-L was 8.7% (4 of 46 with partial response; 95% CI, 2.4–20.8; P=0.001; Table 3, Fig. 2). The DCR was not statistically significantly different between patients with TMB-L and TMB-H (30.4% [95% CI, 29.0–96.3] vs. 71.4% [95% CI, 17.7–45.8]; P=0.084; Table 3). In the PD-L1 CPS ≥1 group, TMB-H showed a higher ORR of 66.7% (4 of 6; 95% CI, 22.3–95.7) than TMB-L with 13.6% (3 of 22; 95% CI, 2.9–34.9; P=0.021; Table 4). However, when patients with a PD-L1 CPS of ≥5 are analyzed separately, the ORR was superior in TMB-H numerically, but there was no statistically significant difference (18.2% [95% CI, 2.3–51.8] vs. 66.7% [95% CI, 9.4–99.2]; P=0.538; Table 5).

Table 3
Comparison of programmed cell death-1 checkpoint inhibitor efficacy in patients with advanced gastric cancer based on TMB

Fig. 2
Objective response rate in relation to TMB, MSI status, PD-L1 expression, and EBV status. TMB-high 71.9% (95% CI, 29.0–96.3) versus TMB-low 8.7% (95% CI, 2.4–20.8; P=0.001), MSI-H 100% (95% CI, 39.8–100) versus MSS 10.2% (95% CI, 3.4–22.2; P=0.000), PD-L1-positive 25% (95% CI, 10.7–44.9) versus PD-L1-negative 0% (95% CI, 0.0–28.5; P=0.159), and EBV-positive 16.7% (95% CI, 0.4–64.1) versus EBV-negative 16.2% (95% CI, 6.2–32; P=1.000).
TMB = tumor mutational burden; MSI = microsatellite instability; PD-L1 = programmed death ligand-1; EBV = Epstein-Barr virus; CI = confidence interval; MSI-H = microsatellite instability-high; MSS = microsatellite stable; pos = positive; neg = negative.

Table 4
Comparison of programmed cell death-1 checkpoint inhibitors efficacy based on TMB in patients with advanced gastric cancer whose PD-L1 CPS score was ≥1

Table 5
Comparison of programmed cell death-1 checkpoint inhibitors efficacy based on TMB in patients with advanced gastric cancer whose PD-L1 CPS score was ≥5

The PFS and OS in 53 patients were 1.93 (95% CI, 1.600–2.268) and 4.26 (95% CI, 2.992–5.532) months, respectively. The median PFS in patients with TMB-H was 18.6 months (95% CI, 4.636–32.479), and that in patients with TMB-L was 1.9 months (95% CI, 1.499–2.370; P=0.040; Fig. 3A). The OS was longer in patients with TMB-H (median, 20.8 months; 95% CI, 2.292–39.281) than in patients with TMB-L (median, 3.3 months; 95% CI, 1.604–5.019; P=0.049; Fig. 3B).

Fig. 3
Kaplan-Meier estimates of time to PFS and OS based on TMB 13.31 mt/Mb. The median follow-up duration was 4.26 months (range, 0.3–67.8 months). (A) PFS for TMB-high and TMB-low. The median PFS for TMB-high patients was 18.6 months (95% CI, 4.636–32.479) and TMB-low patients was 1.9 months (95% CI, 1.499–2.370; P=0.040). (B) OS for TMB-high and TMB-low. The median OS for TMB-high patients was 20.8 months (95% CI, 2.292–39.281) and TMB-low patients was 3.3 months (95% CI, 1.604–5.019; P=0.049).
OS = overall survival; TMB = tumor mutational burden; PFS = progression-free survival; CI = confidence interval.

DISCUSSION

The ROC curve analysis showed that the TMB cutoff value for predicting tumor response in patients with AGC who received PD-1 checkpoint inhibitors was 13.31 mt/Mb. Furthermore, when applying TMB-H, in addition to PD-L1 expression, patients who would benefit more from PD-1 checkpoint inhibitors could be selected. These findings may be helpful for the use of PD-1 checkpoint inhibitors in routine clinical practice.

The reported optimal threshold for defining TMB-H differed with various studies, tumor types, pre-analytic variables, bioinformatics, and test methods [18, 20, 24, 25, 26, 30]. In addition, most studies were mainly conducted in patients with lung cancer and melanoma and relatively few in AGC [30]. In the present study, the TMB of 53 patients was evaluated using the same NGS method to decrease heterogeneity. The NGS diagnostic platform, Illumina’s TSO 500 assay, has a diagnostic value similar to that of WES [31].

However, the clinical significance of the TMB in AGC remains unclear. Wang et al. [32] selected a cutoff of the top 20% TMB (12 mt/Mb) assessed using WES and toripalimab, a humanized PD-1 antibody. Patients with TMB-H showed improved clinical outcome. In a subset analysis of the ATTRACTION-2 phase III trial, TMB-H and TMB-L were classified as having a median value of 8.2 mt/Mb using an NGS panel, and the 2 groups showed no difference in the efficacy of nivolumab as third- or later-line therapy for AGC [11]. Mishima et al. [33] defined TMB-H as >10 mt/Mb based on an NGS panel in AGC and reported that TMB-H was not associated with response to nivolumab. In the present study, the cutoff value for TMB-H was determined to be 13.31 mt/Mb. Patients with TMB-H showed better ORR (71.4% vs. 8.7%, P=0.001), PFS (18.6 vs. 1.9 months, P=0.040), and OS (20.8 vs. 3.3 months, P=0.049) than patients with TMB-L. Compared with previous studies, a relatively larger number of patients were included in the present study, and the cutoff value was identified using the ROC curve analysis by calculating sensitivity and specificity.

The patients with TMB-H and positive PD-L1 expression had a higher ORR (66.7%; 95% CI, 22.3–95.7), which was in agreement with a previous study [32]. TMB status and PD-L1 expression are independent biomarkers for predicting the efficacy of PD-1 checkpoint inhibitors [18, 30, 34]. In gastric cancer, a cutoff value of PD-L1 CPS ≥5 is associated with an increased efficacy of PD-1 checkpoint inhibitors in combination with oxaliplatin-based chemotherapies [1, 2, 35]. However, in PD-1 checkpoint inhibitor monotherapy as a salvage treatment, the effect of PD-L1 is not clear. In the present study, defining the PD-L1 positive as a CPS of ≥1, the PD-L1-positive and TMB-H group exhibited better efficacy than the PD-L1-positive and TMB-L group. This means that among the patients for whom PD-1 checkpoint inhibitors were not indicated because the PD-L1 CPS was <5, additional patients who were expected to be effective could be found using TMB. Furthermore, the limitation of low sensitivity when the cutoff value of TMB is set to 13.31 mt/Mb could be corrected when used with PD-L1.

A higher mutational load is associated with higher neoantigens in tumor immunity, resulting in the recognition of the tumor as foreign and causing an immune response. Thus, hyper-mutated tumors, such as MSI-H or dMMR tumors, show a better response to PD-1 checkpoint inhibitors. MSI-H is associated with high TMB, which accounts for the majority of MSI-H tumors [36]. In the present study, all MSI-H tumors were TMB-H; however, only 57% (4 of 7) of the TMB-H tumors were MSI-H. In addition, one of 3 patients with both TMB-H and microsatellite stable showed a partial response as the best response and 11.8 months of PFS. Therefore, TMB should be assessed independently of other biomarkers such as MSI status when considering PD-1 checkpoint inhibitors.

The present study has several limitations. First, this was a retrospective study conducted at a single center. There were more female than male patients, which is different from previous reports, suggesting a potential for selection bias. Second, the type (i.e., pembrolizumab or nivolumab) and lines of PD-1 checkpoint inhibitors, as well as sequential treatments, were heterogeneous. Third, only patients who received PD-1 checkpoint inhibitors as monotherapy were included. Finally, because the study cohort was small, additional analyses, such as ROC curve analysis using a combination of PD-L1 and TMB, were not conducted.

In conclusion, in the present study, the TMB cutoff value for predicting the tumor response in patients with AGC who received PD-1 checkpoint inhibitor monotherapy as a salvage treatment was 13.31 mt/Mb based on the ROC curve analysis. Furthermore, when PD-L1 status is combined with TMB-H, patients who could benefit from PD-1 checkpoint inhibitors can be selected. These results may be helpful in the use of PD-1 checkpoint inhibitors in routine clinical practice.

Notes

Funding:This research was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant number, HR20C0025).

Conflict of Interest:No potential conflict of interest relevant to this article was reported.

Author Contributions:

  • Conceptualization: K.S.T.

  • Data curation: J.Y., J.S.Y.

  • Formal analysis: J.J.Y.

  • Funding acquisition: K.S.T.

  • Investigation: K.S.T.

  • Methodology: L.S.H.

  • Project administration: J.J.Y.

  • Resources: L.J.

  • Software: J.J.Y.

  • Supervision: K.W.K.

  • Validation: K.W.K.

  • Visualization: J.J.Y.

  • Writing - original draft: J.J.Y.

  • Writing - review & editing: J.Y., J.S.Y.

References

    1. Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet 2021;398:27–40.
    1. Kim TH, Kim IH, Kang SJ, Choi M, Kim BH, Eom BW, et al. Korean practice guidelines for gastric cancer 2022: an evidence-based, multidisciplinary approach. J Gastric Cancer 2023;23:3–106.
    1. Maio M, Ascierto PA, Manzyuk L, Motola-Kuba D, Penel N, Cassier PA, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE-158 study. Ann Oncol 2022;33:929–938.
    1. Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair–deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol 2020;38:1–10.
    1. Kang YK, Boku N, Satoh T, Ryu MH, Chao Y, Kato K, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390:2461–2471.
    1. Chen L, Kang Y, Tanimoto M, Boku N. ATTRACTION-04 (ONO-4538-37): a randomized, multicenter, phase 2/3 study of nivolumab (Nivo) plus chemotherapy in patients (Pts) with previously untreated advanced or recurrent gastric (G) or gastroesophageal junction (GEJ) cancer. Ann Oncol 2017;28:v266.
    1. Moehler M, Dvorkin M, Boku N, Özgüroğlu M, Ryu MH, Muntean AS, et al. Phase III trial of avelumab maintenance after first-line induction chemotherapy versus continuation of chemotherapy in patients with gastric cancers: results from JAVELIN gastric 100. J Clin Oncol 2021;39:966–977.
    1. Shitara K, Özgüroğlu M, Bang YJ, Di Bartolomeo M, Mandalà M, Ryu MH, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet 2018;392:123–133.
    1. Yoon HH, Jin Z, Kour O, Kankeu Fonkoua LA, Shitara K, Gibson MK, et al. Association of PD-L1 expression and other variables with benefit from immune checkpoint inhibition in advanced gastroesophageal cancer: systematic review and meta-analysis of 17 phase 3 randomized clinical trials. JAMA Oncol 2022;8:1456–1465.
    1. Janjigian YY, Bendell J, Calvo E, Kim JW, Ascierto PA, Sharma P, et al. CheckMate-032 study: efficacy and safety of nivolumab and nivolumab plus ipilimumab in patients with metastatic esophagogastric cancer. J Clin Oncol 2018;36:2836–2844.
    1. Kim JH, Ryu MH, Park YS, Ma J, Lee SY, Kim D, et al. Predictive biomarkers for the efficacy of nivolumab as ≥ 3rd-line therapy in patients with advanced gastric cancer: a subset analysis of ATTRACTION-2 phase III trial. BMC Cancer 2022;22:378.
    1. Chia NY, Tan P. Molecular classification of gastric cancer. Ann Oncol 2016;27:763–769.
    1. Wang H, Ding Y, Chen Y, Jiang J, Chen Y, Lu J, et al. A novel genomic classification system of gastric cancer via integrating multidimensional genomic characteristics. Gastric Cancer 2021;24:1227–1241.
    1. Cai H, Jing C, Chang X, Ding D, Han T, Yang J, et al. Mutational landscape of gastric cancer and clinical application of genomic profiling based on target next-generation sequencing. J Transl Med 2019;17:189.
    1. Kwak Y, Seo AN, Lee HE, Lee HS. Tumor immune response and immunotherapy in gastric cancer. J Pathol Transl Med 2020;54:20–33.
    1. Joshi SS, Badgwell BD. Current treatment and recent progress in gastric cancer. CA Cancer J Clin 2021;71:264–279.
    1. Patel TH, Cecchini M. Targeted therapies in advanced gastric cancer. Curr Treat Options Oncol 2020;21:70.
    1. Marabelle A, Fakih M, Lopez J, Shah M, Shapira-Frommer R, Nakagawa K, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–1365.
    1. Jardim DL, Goodman A, de Melo Gagliato D, Kurzrock R. The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell 2021;39:154–173.
    1. Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol 2019;30:44–56.
    1. Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer 2014;14:135–146.
    1. Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012;482:400–404.
    1. Riaz N, Morris L, Havel JJ, Makarov V, Desrichard A, Chan TA. The role of neoantigens in response to immune checkpoint blockade. Int Immunol 2016;28:411–419.
    1. Panda A, Betigeri A, Subramanian K, Ross JS, Pavlick DC, Ali S, et al. Identifying a clinically applicable mutational burden threshold as a potential biomarker of response to immune checkpoint therapy in solid tumors. JCO Precis Oncol 2017;2017:PO.17.00146
    1. Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann Oncol 2019;30:1096–1103.
    1. Sha D, Jin Z, Budczies J, Kluck K, Stenzinger A, Sinicrope FA. Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov 2020;10:1808–1825.
    1. Fluss R, Faraggi D, Reiser B. Estimation of the Youden index and its associated cutoff point. Biom J 2005;47:458–472.
    1. Hajian-Tilaki K. The choice of methods in determining the optimal cut-off value for quantitative diagnostic test evaluation. Stat Methods Med Res 2018;27:2374–2383.
    1. Youden WJ. Index for rating diagnostic tests. Cancer 1950;3:32–35.
    1. Wu Y, Xu J, Du C, Wu Y, Xia D, Lv W, et al. The predictive value of tumor mutation burden on efficacy of immune checkpoint inhibitors in cancers: a systematic review and meta-analysis. Front Oncol 2019;9:1161.
    1. Wei B, Kang J, Kibukawa M, Arreaza G, Maguire M, Chen L, et al. Evaluation of the TruSight oncology 500 assay for routine clinical testing of tumor mutational burden and clinical utility for predicting response to pembrolizumab. J Mol Diagn 2022;24:600–608.
    1. Wang F, Wei XL, Wang FH, Xu N, Shen L, Dai GH, et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann Oncol 2019;30:1479–1486.
    1. Mishima S, Kawazoe A, Nakamura Y, Sasaki A, Kotani D, Kuboki Y, et al. Clinicopathological and molecular features of responders to nivolumab for patients with advanced gastric cancer. J Immunother Cancer 2019;7:24.
    1. Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, et al. 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 2018;36:633–641.
    1. Kang YK, Chen LT, Ryu MH, Oh DY, Oh SC, Chung HC, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:234–247.
    1. Wu CE, Yeh DW, Pan YR, Huang WK, Chen MH, Chang JW, et al. Chromosomal instability may not be a predictor for immune checkpoint inhibitors from a comprehensive bioinformatics analysis. Life (Basel) 2020;10:276.

Metrics
Share
Figures

1 / 3

Tables

1 / 5

Funding Information
PERMALINK