INTRODUCTION
Upper gastrointestinal (UGI) malignancies include gastric, gastroesophageal junction, and esophageal tumors. According to the World Health Organization's global cancer statistics, 1.09 million people are diagnosed with stomach cancer and 600 000 with esophageal cancer every year, making it the fourth most common cancer worldwide. The incidence of gastric cancer (GC) deaths was 770 000 and that of esophageal cancer deaths was 540 000, ranking second in total mortality among all tumors. Despite improvements in survival rates, the 5-year survival rate for advanced UGI tumors remains unsatisfactory, with cytotoxic chemotherapy being the primary treatment option. Immunotherapies have proven to be a promising option, changing the standard of care for some solid tumors. Due to the low overall response rate to immunotherapy in UGI cancers, there is a need for selection of patients who may be eligible for immunotherapy. Several biomarkers have been reported to predict immunotherapy response, including programmed death ligand 1 (PD-L1), microsatellite instability (MSI), tumor mutational load (TML), and tumor-infiltrating lymphocytes, among others. TML refers to the total number of mutations per megabase of tumor tissue which can be detected by either next-generation sequencing (NGS) or whole-exome sequencing (WES), and reflects the overall burden of tumor antigens. The fundamental principle of immunotherapy is to stimulate and enhance host T lymphocytes to eliminate tumors. Theoretically, a higher TML in cancer cells makes them more likely to be recognized by the immune system, thereby improving the effectiveness of immunotherapy. According to previous studies, non-small cell lung cancer and melanoma have higher tumor mutation loads than other tumors, whereas UGI tract tumors do not have very high tumor mutation loads. Although there has been considerable progress in the study of TML as a predictive biomarker in non-small cell lung cancer and melanoma, its application to UGI cancer remains incomplete. Therefore, the aim of this systematic review and meta-analysis was to evaluate the prognostic value of TML in UGI cancers treated with immune checkpoint inhibitors (ICIs) on the basis of the latest data.
MATERIALS AND METHODS
This systematic review and meta-analysis were conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines provided in Supplementary File . This systematic review and analysis were registered in the International Prospective Register of Systematic Reviews (CRD42023405596, ).
Search strategy
A comprehensive search of PubMed, Embase, Cochrane Library, and Web of Science databases was performed for the period of time up until February 13, 2023, using the following search terms: (esophageal neoplasms OR gastroesophageal neoplasms OR stomach neoplasms) AND (mutation burden OR mutation load OR tumor mutational burden OR TML) AND (immunotherapy OR immune checkpoint inhibitor OR ICI OR immune checkpoint blocker OR ICB OR PD-1 OR PD-L1 OR CTLA-4). The detailed search strategies are provided in Supplementary File .
Study selection
Two investigators (CHM and QT) independently searched and screened the studies and resolved disagreements through discussion and consensus. Subsequently, the titles and abstracts of the studies were assessed for eligibility according to the inclusion criteria, after removing duplicate reports.
- Patients were diagnosed with UGI cancer based on pathological results.
- A well-defined TML cutoff was used to stratify the subjects into two groups: those with high TML and those with low TML.
- Studies should evaluate the predictive value of TML for the prognosis of various ICIs, including anti-PD-1, anti-PD-L1, anti-CTLA-4, and their combination therapies.
- For TML-related overall survival (OS) or progression-free survival (PFS), studies should report the hazard ratio (HR) and its 95% confidence interval (CI). Studies should provide Kaplan–Meier curves or raw OS or PFS data to allow the calculation of relevant metrics if these conditions cannot be met.
- Case reports, reviews, conference abstracts, editorials, letters, animal studies, and studies with incomplete data were excluded from the analysis. Only studies written in English and which met the eligibility criteria were included after a full-text review.
Data extraction
Data extracted from eligible studies included author, publication year, region, tumor type, patient number, ICI type, sample source, TML sequencing method, TML cutoff value, median TML and range, survival outcome, HRs, and 95% CIs.
Quality assessment
The studies included in this review were cohort studies and randomized controlled trials. The quality of the cohort studies was assessed using the Newcastle-Ottawa Scale (NOS), which is divided into three dimensions with a score range of 0–9. Studies were categorized into three levels based on their cumulative scores: high quality, moderate quality, and low quality (0–4). In particular, the quality of only the randomized controlled trials that included patients treated with pembrolizumab in this analysis was assessed using the NOS.
Statistical analysis
By comparing the PFS and OS between the high and low TML groups, we verified the predictive value of TML in the prognosis of immunotherapy in UGI cancers using HR. Cox proportional hazards regression models were used to calculate HRs and corresponding 95% CIs for PFS and OS using IBM SPSS Statistics 25 for studies that provided raw data. We used the Engauge Digitizer to extract survival data and the Tierney et al. program file to calculate HRs and 95% CIs for studies that presented Kaplan–Meier curves without HRs for PFS and OS. RevMan 5.4.1 was used to estimate the summary HR, 95% CI, and p-values.
STATA 13 was used to assess the heterogeneity using the I2 statistic and p-values. Fixed- and random-effects models were applied to the meta-analyses with low and high heterogeneity, respectively. Heterogeneity levels were classified as low (25% < I2 < 50%), moderate (50% < I2 < 75%), or significant (I2 > 75%). If there was significant heterogeneity, the source was further investigated by assessing publication bias, conducting sensitivity analyses, and performing subgroup analyses. To ensure that a sufficient number of eligible studies were included, we conducted a funnel plot and an Egger's test to assess publication bias, with p > .05 indicating the absence of publication bias. To assess the effect of excluding individual studies on summary HR, sensitivity analyses were performed. The region, tumor type, ICI type, sample source, TML sequencing method, and TML cutoff value were set as subgroups for subgroup analysis.
RESULTS
Literature search
After removing 679 duplicate studies, a total of 860 studies were retrieved from the Web of Science, Embase, PubMed, and Cochrane Library databases in accordance with the search strategy. Title and abstract screening were used to exclude case reports, reviews, conference abstracts, editorials, letters, animal studies, and studies with irrelevant topics. Subsequently, 268 eligible studies were subjected to a full-text review for further screening. Finally, 11 publications between 2018 and 2022 were included in the analysis. The flowchart depicting the process of search and identification of studies is shown in Figure .
[IMAGE OMITTED. SEE PDF]
Study characteristics and quality assessment
The study characteristics are summarized in Table . This meta-analysis included 12 cohorts from 11 studies, totaling 932 patient samples ranging from 20 to 202 patients. Of these, four studies were conducted in Western countries, seven in Asian countries, and one across multiple regions. Regarding the type of ICI treatment, anti-PD-1 monotherapy was used in seven cohorts, whereas anti-PD-1/L1 or anti-CTLA-4 was used as monotherapy or in combination in five cohorts. Patients with esophageal cancer (EC), GC, and esophagogastric cancer were included in the study. WES and NGS were used for the analysis of blood and tumor tissue specimens. Regarding the sample source among the cohorts, eight used tumor tissue samples exclusively, two employed both tumor tissue and matched peripheral blood samples, and two relied solely on blood samples. Different studies have defined varying cutoff values for TML. PFS was used to express the survival data for 10 cohorts, whereas OS was used for eight cohorts. Based on the NOS score, five studies scoring 8–9 were considered high quality, and seven cohorts scoring 6–7 were considered moderate quality (Supplementary File ).
TABLE 1 Characters of included studies in the meta-analysis.
Reference | Region | Number of patients (high/low TMB) | Type of ICIs | Tumor type | Sample source | Sequencing method | TMB cutoff value | Median TMB value (range) | Survival outcome |
Bai et al. | Asian (China) | 20 (13/7) | ICI | GC | Tumor | NGS | 8.82 mut/Mb | NA | PFS |
Greally et al. | Western (USA) | 89 (22/67) | ICI | EGC | Tumor | NGS | 8.78 mut/Mb | 5.6 mut/Mb | PFS, OS |
Huang et al. | Asian (China) | 23 (11/12) | Anti-PD-1 | EC | Tumor, blood | WES | 60 mut/tumor | 60 mut/tumor (15–219) | PFS |
Kim et al. | Asian (Korea) | 105 (11/94) | Anti-PD-1 | GC | Tumor | NGS | 18.03 mut/Mb | NA | PFS, OS |
Kim et al. | Asian (Korea) | 63 (8/55) | Anti-PD-1 | GC | Tumor | NGS | 14.31 mut/Mb | 4.23 mut/Mb | PFS |
Lu et al. | Asian (China) | 63 (11/52) | Anti-PD-1 | GC (GEJC) | Tumor | NGS | 8 mut/Mb | NA | PFS, OS |
Mishima et al. | Asian (Japan) | 58 (34/24) | Anti-PD-1 | GC | Tumor | NGS | 10 mut/Mb | NA | PFS |
Samstein et al. | Western (USA) | 126 (30/96) | ICI | EGC | Tumor | NGS | 7.9 mut/Mb | 7.4 mut/Mb | OS |
Shitara et al. | Multiple areas | 202 (36/166) | Anti-PD-1 | GC (GEJC) | Tumor | WES | 175 mut/exome | NA | OS |
Valero et al. | Western (USA) | 49 (12/37) | ICI | EC | Blood | NGS | 8 mut/Mb | 5.3 mut/Mb | PFS, OS |
Valero et al. | Western (USA) | 71 (15/56) | ICI | GC | Blood | NGS | 7 mut/Mb | 4.4 mut/Mb | PFS, OS |
Wang et al. | Asian (China) | 54 (12/42) | Anti-PD-1 | GC (GEJC) | Tumor, blood | WES | 12 mut/Mb | NA | PFS, OS |
Key findings
We performed a meta-analysis of 10 cohorts for PFS and eight cohorts for OS, comparing groups of patients with high and low TML. A fixed-effects model was used to calculate summary HRs with the corresponding 95% CIs. Our results showed that, as indicated by the summary HRs for PFS (HR = 0.74; 95% CI: 0.58, 0.95; p = .020) and OS (HR = 0.68; 95% CI: 0.53, 0.86; p = .001), the high TML group had a better survival benefit than the low TML group (Figure ).
[IMAGE OMITTED. SEE PDF]
Publication bias
Funnel plots for publication bias analysis are shown in Figure . Publication bias was quantitatively assessed using Egger's test for PFS (p = .045). We reanalyzed the correlation after excluding one study with a sample size of 20, as publication bias appeared to be primarily driven by studies with small sample sizes. The results (pooled HR = 0.76; 95% CI: 0.59–0.97; p = .030) showed that PFS remained improved in the high TML group. Notably, publication bias disappeared (p Egger = .067). The absence of publication bias was demonstrated using Egger's test for OS (p = .062).
[IMAGE OMITTED. SEE PDF]
Assessment of heterogeneity and sensitivity analysis
There was little heterogeneity in the summary HR for PFS among the combined studies (I2 = 32%, p = .150) and no heterogeneity in OS (I2 = 0, p = .840) between patients in groups with and without high TML. As shown in Figure , the sensitivity analysis showed that when the study reported by Kim et al. is excluded, the summary 95% CI of the remaining nine studies exceeded 1 (95% CI: 0.61, 1.01), suggesting no association of TML with PFS, contrary to our previous results. This suggests that this study significantly influenced the overall results and could be considered less robust. After exclusion, I2 decreased to 4%. Sensitivity analysis of OS showed a minimal change in the pooled effect when each article was excluded individually. According to GRADE standards, the quality of evidence was low for both OS and PFS.
[IMAGE OMITTED. SEE PDF]
Subgroup analysis of
Table presents the results of PFS subgroup analyses. Regarding the subgroups based on patient recruitment area, the high TML group showed superior PFS (HR = 0.70; 95% CI: 0.51, 0.97; p = .030) in the Asian country subgroup, whereas there is no significant difference in the Western country subgroup (HR = 0.82; 95% CI: 0.55, 1.22; p = .320). The anti-PD-1 subgroup (HR = 0.66; 95% CI: 0.41, 1.06; p = .090) and the ICI subgroup (HR = 0.78; 95% CI: 0.52, 1.15; p = .480) were not statistically different. The correlation between TML level and PFS benefit was independent of the cancer type. PFS did not differ significantly between the groups with and without high TMLs, regardless of whether the specimens were taken from tumors or blood. With regard to the TML sequencing approach, we found that in the WES subgroup, PFS was superior in patients with high TML than in those with low TML, whereas no difference was observed in the NGS subgroup. In subgroup analysis based on TML cutoff, no significant PFS advantage was observed between patients with high TML and those with low TML, regardless of whether their TML cutoff was ≥10 mutations per megabase (mut/Mb). Meanwhile, subgroup analyses significantly reduced the heterogeneity among the subgroups of Western countries, ICI treatment, blood source, WES method, and a TML cutoff value of <10 mut/Mb. No significant heterogeneity was observed among the subgroups.
TABLE 2 Subgroup analysis of progression-free survival between TMB high and TMB low groups.
Subgroup | Number of study | HR (95% CI) | p-Value | I2 | Heterogeneity between subgroups |
Region | |||||
Asian | 7 | 0.70 (0.51, 0.96) | .030 | 49% | p = .570 |
Western | 3 | 0.82 (0.55, 1.22) | .320 | 0% | |
Type of ICIs | |||||
Anti-PD-1 | 6 | 0.66 (0.41, 1.06) | .090 | 53% | p = .600 |
ICI | 4 | 0.78 (0.52, 1.15) | .480 | 0% | |
Tumor type | |||||
EC | 2 | 0.75 (0.27, 2.07) | .580 | 56% | p = .860 |
GC | 7 | 0.68 (0.45, 1.04) | .080 | 45% | |
Sample source | |||||
Tumor | 6 | 0.79 (0.58, 1.09) | .150 | 48% | p = .690 |
Blood | 2 | 0.90 (0.53, 1.54) | .700 | 0% | |
Sequencing method | |||||
WES | 2 | 0.48 (0.27, 0.86) | .010 | 0% | p = .110 |
NGS | 8 | 0.82 (0.62, 1.08) | .150 | 34% | |
TMB cutoff value | |||||
≥10 mut/Mb | 4 | 0.64 (0.31, 1.30) | .210 | 68% | p = .580 |
<10 mut/Mb | 5 | 0.79 (0.56, 1.12) | .190 | 0% |
Subgroup analysis of
Table shows the results of the OS subgroup analysis. Subgroup analysis by treatment type showed that anti-PD-1 therapy had better OS outcomes in patients in the high TML group (HR = 0.61; 95% CI: 0.44, 0.84; p = .002), whereas there was no significant difference in the ICI subgroup. In the GC subgroup, the OS benefit was greater in patients with high TML (HR = 0.62; 95% CI: 0.47, 0.84; p = .002). A higher TML correlated with longer OS (HR = 0.69; 95% CI: 0.51, 0.92; p = .010) when the specimen was tumor-derived. The difference between the groups with higher and lower TML was statistically significant in the WES subgroup, but not in the NGS subgroup. The OS was longer in the high TML group when the TML cutoff value was ≥10 mut/Mb (HR = 0.57; 95% CI: 0.33, 0.97; p = .040). The association between TML levels and OS was independent of the region from which the patients were recruited. In the subgroups of EC, blood samples, and TML cutoff value <10 mut/Mb, the high TML group did not demonstrate any significant advantages over the low TML group. As shown in Table , no heterogeneity was observed within each subgroup or between corresponding subgroups.
TABLE 3 Subgroup analysis of overall survival between TMB high and TMB low groups.
Subgroup | Number of study | HR (95% CI) | p-Value | I2 | Heterogeneity between subgroups |
Region | |||||
Asian | 3 | 0.66 (0.42, 1.04) | .070 | 0% | p = .600 |
Western | 4 | 0.77 (0.54, 1.10) | .160 | 0% | |
Type of ICIs | |||||
Anti-PD-1 | 4 | 0.61 (0.44, 0.84) | .002 | 0% | p = .330 |
ICI | 4 | 0.77 (0.54, 1.10) | .160 | 0% | |
Tumor type | |||||
EC | 1 | 0.86 (0.37, 2.00) | .730 | 0% | p = .480 |
GC | 5 | 0.62 (0.47, 0.84) | .002 | 0% | |
Sample source | |||||
Tumor | 5 | 0.69 (0.51, 0.92) | .010 | 0% | p = .710 |
Blood | 2 | 0.77 (0.45, 1.33) | .350 | 0% | |
Sequencing method | |||||
WES | 2 | 0.53 (0.36, 0.78) | .001 | 0% | p = .110 |
NGS | 6 | 0.79 (0.58, 1.07) | .130 | 0% | |
TMB cutoff value | |||||
≥10 mut/Mb | 2 | 0.57 (0.33, 0.97) | .040 | 0% | p = .280 |
<10 mut/Mb | 5 | 0.80 (0.57, 1.11) | .180 | 0% |
DISCUSSION
Our findings suggest that, among patients with UGI cancers treated with ICIs, the high mutation load group exhibited better PFS and OS than the low mutation load group. Subgroup analysis further reveals that this survival advantage is particularly evident among Asian patients, those with GC, those treated with anti-PD-1, those whose samples were obtained from tumor tissue, and when a TML cutoff of ≥10 mut/Mb was used. As higher TML did not correlate with improved prognosis in non-ICI-treated patients, the improved OS and PFS in patients with high TML should be attributed to the efficacy of immunotherapy. Numerous other studies have also shown that elevated TML is linked to a favorable prognosis in patients with solid tumors undergoing immunotherapy.
As previously mentioned, superior PFS was observed in Asian and GC subgroups. Recent large solid tumor data from an Asian population revealed that the TML of GC in the Advanced Origin Med cohort was significantly higher than that in the Memorial Sloan Kettering Cancer Center cohort, which may account for the clinical benefits observed in our study. This is because our study included a larger cohort of patients with GC and these patients were predominantly Asian. Our results demonstrated a significant OS benefit in patients with high TML who received anti-PD-1 therapy. In addition, promising results have been obtained with combination therapy with anti-PD-1/PD-L1 and anti-CTLA-4. Furthermore, new immunotherapies—such as therapeutic vaccines and chimeric antigen receptor t-cell therapy—are currently under investigation. TML can serve as an indicator for screening individuals who may benefit from immunotherapies. Several trials, including CheckMate-649, ATTRACTION-04, and KEYNOTE-062, have used immunotherapy in combination with chemotherapy or targeted therapies. In fact, nivolumab plus chemotherapy for the first-line treatment of advanced GC has been approved by the Food and Drug Administration of the US.
Although WES is relatively accurate in measuring TML, NGS has gradually replaced it with the development of technology owing to its better economy and timeliness. Our results demonstrated that patients with high TML levels receiving ICI therapy detected by WES exhibited a survival advantage over those detected by NGS, which differs from a previous study. This may be due to the limited number of studies that used the WES method, resulting in less stable results. Compared to tumor tissue samples, blood samples showed no benefit in the ICI-treated high TML group. This may be due to insufficient circulating tumor DNA in peripheral blood. However, it can be challenging to obtain sufficient tumor tissue for molecular testing in patients with advanced disease; therefore, a blood-based method for the detection of TML is a viable option. The correlation between TML and blood-based TML, as well as the synergistic effects of these predictive biomarkers on clinical outcomes, requires further investigation. While there may not be a universal definition of what constitutes high TML, a multi-tumor clinical trial has shown that a high TML is associated with improved survival in patients receiving ICIs across a broad range of cancer types. Anti-PD-1 therapy was approved by the Food and Drug Administration of the US in 2020 for all solid tumor types with TML ≥10 mut/Mb based on Keynote 158 trial. The results of the cohort study further support the notion that, across cancer types, tumors with a TML ≥10 mut/Mb have a higher response rate to ICI therapy. For this reason, a cutoff of 10 mut/Mb was chosen. Our findings demonstrate that the summary HRs for TML cutoff values ≥10 mut/Mb are statistically significant. However, several challenges remain in integrating TML into standard clinical practice, including inconsistencies in its definition and the need for the standardization of analytical methods across laboratories and platforms.
Although the prognostic impact of TML has been investigated in melanoma, lung, colorectal, and breast cancers, its association with survival outcomes in esophageal and GCs remains unknown. Our meta-analysis explored, for the first time, the relationship between TML and UGI tumors, providing a valuable research reference for the future use of TML as a potential predictive biomarker to guide clinical immunotherapy. However, our study has several limitations which require attention. Only English studies were considered in this review, which may have resulted in the omission of important literature published in other languages. The limited number of studies and small sample size may have impacted the accuracy of our findings. In addition, the correlation between tumor and blood TML has not been conclusively established, and the standardization of TML sequencing remains an issue. Therefore, our conclusions may be less stable and require further investigation. In addition to TML, other biomarkers used to predict immunotherapy efficacy include PD-L1, MSI, and Epstein-Barr virus. As the number of approved ICIs increases and the corresponding landmark trials for PD-L1 antibodies use different immunohistochemical assays, the need for clinical and logistic harmonization remains unmet. MSI status and Epstein-Barr virus positivity may serve as more effective predictive biomarkers for immunotherapy responses; however, the incidence rates of these biomarkers are relatively low. To date, no single biomarker has been able to accurately and universally predict the efficacy of immunotherapy. We believe that identifying the optimal combination of biomarkers for predicting immunotherapeutic responses represents a future direction. The most promising predictive combination of TML and other relevant biomarkers must be validated in large-scale randomized trials.
CONCLUSION
Our meta-analysis demonstrated that TML is a promising predictive biomarker, as high TML is significantly associated with longer survival in patients with UGI cancer who received ICIs. We suggest that 10 mut/Mb should be used as a cutoff value for the detection of TML to identify potential responders to immunotherapy.
AUTHOR CONTRIBUTIONS
Chenghao Ma: Conceptualization (lead); data curation (lead); formal analysis (lead); investigation (lead); methodology (lead); project administration (lead); validation (lead); visualization (lead); writing – original draft (lead); writing – review and editing (lead). Qiong Teng: Data curation (equal); formal analysis (equal); investigation (equal); writing – original draft (equal). Liang Shang: Funding acquisition (equal); project administration (equal); resources (equal); supervision (lead); writing – original draft (equal). Fengying Du: Formal analysis (equal); software (equal); visualization (equal); writing – original draft (equal); writing – review and editing (equal). Leping Li: Funding acquisition (lead); project administration (equal); resources (lead); supervision (equal); writing – original draft (equal).
ACKNOWLEDGMENTS
This study was supported by the Key Research and Development Program of Shandong Province (No. 2021CXGC011104); Special Foundation for Taishan Scholars Program of Shandong Province (No.ts20190978); National Natural Science Foundation of China (No. 82203854).
CONFLICT OF INTEREST STATEMENT
All authors have completed the ICMJE uniform disclosure form. The authors have stated explicitly that there are no conflicts of interest in connection with this article.
DATA AVAILABILITY STATEMENT
Data Availability StatementThe original contributions presented in the study are included in the article. Further inquiries can be directed to the corresponding author.
ETHICS STATEMENT
There were no ethical issues involved in our study.
Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249.
Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17‐48.
Digklia A, Wagner AD. Advanced gastric cancer: current treatment landscape and future perspectives. World J Gastroenterol. 2016;22(8):2403‐2414.
Vrana D, Matzenauer M, Neoral C, et al. From tumor immunology to immunotherapy in gastric and esophageal cancer. Int J Mol Sci. 2018;20(1):13.
Smith A, Roy A, Karapetis CS, Broadbridge V, Price T. Immunotherapy use in oesophagogastric cancers‐a review of the literature. Br J Cancer. 2022;127(1):21‐29.
Xie J, Fu L, Jin L. Immunotherapy of gastric cancer: past, future perspective and challenges. Pathol Res Pract. 2021;218: [eLocator: 153322].
Zhao Q, Cao L, Guan L, et al. Immunotherapy for gastric cancer: dilemmas and prospect. Brief Funct Genomics. 2019;18(2):107‐112.
Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor‐based immunotherapy. Lancet Oncol. 2016;17(12): [eLocator: e542‐e551].
Yi M, Jiao D, Xu H, et al. Biomarkers for predicting efficacy of PD‐1/PD‐L1 inhibitors. Mol Cancer. 2018;17(1):129.
Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer. 2019;19(3):133‐150.
Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response to CTLA‐4 blockade in metastatic melanoma. Science. 2015;350(6257):207‐211.
Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD‐1 blockade in non‐small cell lung cancer. Science. 2015;348(6230):124‐128.
Chan TA, Wolchok JD, Snyder A. Genetic basis for clinical response to CTLA‐4 blockade in melanoma. N Engl J Med. 2015;373(20):1984.
Kim JY, Kronbichler A, Eisenhut M, et al. Tumor mutational burden and efficacy of immune checkpoint inhibitors: a systematic review and meta‐analysis. Cancers (Basel). 2019;11(11): [eLocator: 1798].
Cao J, Yang X, Chen S, et al. The predictive efficacy of tumor mutation burden in immunotherapy across multiple cancer types: a meta‐analysis and bioinformatics analysis. Transl Oncol. 2022;20: [eLocator: 101375].
Sholl LM, Hirsch FR, Hwang D, et al. The promises and challenges of tumor mutation burden as an immunotherapy biomarker: a perspective from the International Association for the Study of Lung Cancer pathology committee. J Thorac Oncol. 2020;15(9):1409‐1424.
Ning B, Liu Y, Wang M, Li Y, Xu T, Wei Y. The predictive value of tumor mutation burden on clinical efficacy of immune checkpoint inhibitors in melanoma: a systematic review and meta‐analysis. Front Pharmacol. 2022;13: [eLocator: 748674].
Liberati A, Altman DG, Tetzlaff J, et al. The PRISMA statement for reporting systematic reviews and meta‐analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339: [eLocator: b2700].
Stang A. Critical evaluation of the Newcastle‐Ottawa scale for the assessment of the quality of nonrandomized studies in meta‐analyses. Eur J Epidemiol. 2010;25(9):603‐605.
Shitara K, Özgüroğlu M, Bang YJ, et al. Molecular determinants of clinical outcomes with pembrolizumab versus paclitaxel in a randomized, open‐label, phase III trial in patients with gastroesophageal adenocarcinoma. Ann Oncol. 2021;32(9):1127‐1136.
Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time‐to‐event data into meta‐analysis. Trials. 2007;8:16.
Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ. 2003;327(7414):557‐560.
Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. BMJ. 1997;315(7109):629‐634.
Bai Y, Xie T, Wang Z, et al. Efficacy and predictive biomarkers of immunotherapy in Epstein‐Barr virus‐associated gastric cancer. J Immunother Cancer. 2022;10(3): [eLocator: e004080].
Greally M, Chou JF, Chatila WK, et al. Clinical and molecular predictors of response to immune checkpoint inhibitors in patients with advanced Esophagogastric cancer. Clin Cancer Res. 2019;25(20):6160‐6169.
Huang J, Xu B, Mo H, et al. Safety, activity, and biomarkers of SHR‐1210, an anti‐PD‐1 antibody, for patients with advanced esophageal carcinoma. Clin Cancer Res. 2018;24(6):1296‐1304.
Kim HD, Ryu MH, Park YS, Lee SY, Moon M, Kang YK. Insertion‐deletion rate is a qualitative aspect of the tumor mutation burden associated with the clinical outcomes of gastric cancer patients treated with nivolumab. Gastric Cancer. 2022;25(1):226‐234.
Kim J, Kim B, Kang SY, et al. Tumor mutational burden determined by panel sequencing predicts survival after immunotherapy in patients with advanced gastric cancer. Front Oncol. 2020;10: [eLocator: 314].
Lu ZH, Yang SL, Luo XR, et al. The combination of gene hyperamplification and PD‐L1 expression as a biomarker for the clinical benefit of tislelizumab in gastric/gastroesophageal junction adenocarcinoma. Gastric Cancer. 2022;25(5):943‐955.
Mishima S, Kawazoe A, Nakamura Y, et al. Clinicopathological and molecular features of responders to nivolumab for patients with advanced gastric cancer. J Immunother Cancer. 2019;7(1):24.
Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51(2):202‐206.
Valero C, Lee M, Hoen D, et al. Pretreatment neutrophil‐to‐lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat Commun. 2021;12(1): [eLocator: 729].
Wang F, Wei XL, Wang FH, 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(9):1479‐1486.
Valero C, Lee M, Hoen D, et al. The association between tumor mutational burden and prognosis is dependent on treatment context. Nat Genet. 2021;53(1):11‐15.
Wu Y, Xu J, Du C, 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: [eLocator: 1161].
Huang T, Chen X, Zhang H, et al. Prognostic role of tumor mutational burden in cancer patients treated with immune checkpoint inhibitors: a systematic review and meta‐analysis. Front Oncol. 2021;11: [eLocator: 706652].
Wu L, Yao H, Chen H, et al. Landscape of somatic alterations in large‐scale solid tumors from an Asian population. Nat Commun. 2022;13(1):4264.
Antonia S, Goldberg SB, Balmanoukian A, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non‐small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol. 2016;17(3):299‐308.
Sobhani N, Scaggiante B, Morris R, et al. Therapeutic cancer vaccines: from biological mechanisms and engineering to ongoing clinical trials. Cancer Treat Rev. 2022;109: [eLocator: 102429].
Qi C, Gong J, Li J, et al. Claudin18.2‐specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022;28(6):1189‐1198.
Shitara K, Van Cutsem E, Bang YJ, et al. Efficacy and safety of Pembrolizumab or Pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first‐line, advanced gastric cancer: the KEYNOTE‐062 phase 3 randomized clinical trial. JAMA Oncol. 2020;6(10):1571‐1580.
Janjigian YY, Shitara K, Moehler M, 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(10294):27‐40.
Kang YK, Chen LT, Ryu MH, 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(2):234‐247.
Johnson DB, Frampton GM, Rioth MJ, et al. Targeted next generation sequencing identifies markers of response to PD‐1 blockade. Cancer Immunol Res. 2016;4(11):959‐967.
Gandara DR, Paul SM, Kowanetz M, et al. Blood‐based tumor mutational burden as a predictor of clinical benefit in non‐small‐cell lung cancer patients treated with atezolizumab. Nat Med. 2018;24(9):1441‐1448.
Friedlaender A, Nouspikel T, Christinat Y, Ho L, McKee T, Addeo A. Tissue‐plasma TMB comparison and plasma TMB monitoring in patients with metastatic non‐small cell lung cancer receiving immune checkpoint inhibitors. Front Oncol. 2020;10:142.
Marabelle A, Fakih M, Lopez J, 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(10):1353‐1365.
Valero C, Lee M, Hoen D, et al. Response rates to anti‐PD‐1 immunotherapy in microsatellite‐stable solid tumors with 10 or more mutations per Megabase. JAMA Oncol. 2021;7(5):739‐743.
McNamara MG, Jacobs T, Lamarca A, Hubner RA, Valle JW, Amir E. Impact of high tumor mutational burden in solid tumors and challenges for biomarker application. Cancer Treat Rev. 2020;89: [eLocator: 102084].
Ke L, Li S, Cui H. The prognostic role of tumor mutation burden on survival of breast cancer: a systematic review and meta‐analysis. BMC Cancer. 2022;22(1):1185.
Li Y, Ma Y, Wu Z, et al. Tumor mutational burden predicting the efficacy of immune checkpoint inhibitors in colorectal cancer: a systematic review and meta‐analysis. Front Immunol. 2021;12: [eLocator: 751407].
Nan Z, Guoqing W, Xiaoxu Y, et al. The predictive efficacy of tumor mutation burden (TMB) on nonsmall cell lung cancer treated by immune checkpoint inhibitors: a systematic review and meta‐analysis. Biomed Res Int. 2021;2021: [eLocator: 1780860].
Yeong J, Lum HYJ, Teo CB, et al. Choice of PD‐L1 immunohistochemistry assay influences clinical eligibility for gastric cancer immunotherapy. Gastric Cancer. 2022;25(4):741‐750.
Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024. This work is published under http://creativecommons.org/licenses/by/4.0/ (the "License"). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Background
Tumor mutational load (TML) has emerged as a potential biomarker for multiple solid tumors. However, data on its prognostic impact on upper gastrointestinal (UGI) cancer are limited. Therefore, the aim of this systematic review and meta‐analysis was to assess the prognostic value of TML for the survival of patients with UGI cancer.
Method
A comprehensive search of the PubMed, Embase, Cochrane Library, and Web of Science databases was conducted up to February 13, 2023. Eleven studies met our inclusion criteria. Hazard ratios (HRs) for progression‐free survival and overall survival and their 95% confidence intervals (CIs) were calculated. Subsequently, the combined HR and its 95% CI were calculated for UGI tract cancers in the high and low TML groups.
Results
In total, 932 patients with UGI tract cancer from 11 publications were included. The high TML group treated with immunotherapy showed significantly improved overall survival (HR = 0.68; 95% CI: 0.53, 0.86;
Conclusion
Our study demonstrated that patients with UGI tumors and higher TML have a better prognosis with immunotherapy, suggesting that TML is a promising predictive biomarker for immunotherapy.
Registration
The study protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO Registration No: CRD42023405596).
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details

1 Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
2 Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China