Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2015 2023-12-21 18:03:55 |
2 "Bioinformatics advancements" changed to "Advancements in bioinformatics" + 1 word(s) 2016 2023-12-21 18:05:49 | |
3 Reference format revised. Meta information modification 2016 2023-12-22 03:44:29 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ahmed, J.; Das, B.; Shin, S.; Chen, A. Management of Tumor Mutational Burden-High Advanced Solid Malignancies. Encyclopedia. Available online: https://encyclopedia.pub/entry/53045 (accessed on 09 July 2024).
Ahmed J, Das B, Shin S, Chen A. Management of Tumor Mutational Burden-High Advanced Solid Malignancies. Encyclopedia. Available at: https://encyclopedia.pub/entry/53045. Accessed July 09, 2024.
Ahmed, Jibran, Biswajit Das, Sarah Shin, Alice Chen. "Management of Tumor Mutational Burden-High Advanced Solid Malignancies" Encyclopedia, https://encyclopedia.pub/entry/53045 (accessed July 09, 2024).
Ahmed, J., Das, B., Shin, S., & Chen, A. (2023, December 21). Management of Tumor Mutational Burden-High Advanced Solid Malignancies. In Encyclopedia. https://encyclopedia.pub/entry/53045
Ahmed, Jibran, et al. "Management of Tumor Mutational Burden-High Advanced Solid Malignancies." Encyclopedia. Web. 21 December, 2023.
Management of Tumor Mutational Burden-High Advanced Solid Malignancies
Edit

A standardized assessment of Tumor Mutational Burden (TMB) poses challenges across diverse tumor histologies, treatment modalities, and testing platforms, requiring careful consideration to ensure consistency and reproducibility. Despite clinical trials demonstrating favorable responses to immune checkpoint inhibitors (ICIs), not all patients with elevated TMB exhibit benefits, and certain tumors with a normal TMB may respond to ICIs. Therefore, a comprehensive understanding of the intricate interplay between TMB and the tumor microenvironment, as well as genomic features, is crucial to refine its predictive value. Advancements in bioinformatics hold potential to improve the precision and cost-effectiveness of TMB assessments, addressing existing challenges. 

TMB tumor mutational burden ICI immune checkpoint inhibitor

1. Background

Tumor Mutational Burden (TMB) has emerged as a promising genomic biomarker of response to immune checkpoint inhibitors that was pioneered by Rizvi and Chan [1]. TMB is defined as the total number of mutations within a tumor genome (mutations per megabase of genome or mut/Mb) and serves as a measure of the potential neoantigen load that may elicit an anti-tumor immune response. This is based upon the type of mutations, a frequency of threshold in the context of clinical activity, the type of cancer cohort, and the sequencing methods used (whole-exome sequencing (WES), whole-genome sequencing (WGS), targeted panel-based next-generation sequencing (NGS)) [2].
TMB molecular signatures are varied, and they arise from a combination of both exogenous and endogenous factors, which represent environmental factors that affect mutagenesis rates and mutations due to random errors in deoxyribonucleic acid (DNA) replication, respectively [3]. This molecular signature has been shown to be predictive of response to immune checkpoint inhibitors (ICIs) in several clinical trial settings. Similarly, hypermutated tumors vary in their causative factors, for example, UV (ultraviolet) light for skin cancer and smoking in non-small cell lung cancer (NSCLC), as well as somatic mutations. In addition, there is a huge variability in mutational load across cancers, with melanoma and NSCLC frequently showing a high TMB. Thus, defining a median range across several cancer types is not practical and other approaches might serve as useful predictive biomarkers of response [4]. Despite these adjustments, cancers such as renal cell cancer (RCC) may not have a high TMB but respond well to ICI.
High TMB is common among several cancers, with an incidence of TMB ≥ 10 mut/Mb of 14.7% based on the TSO500 (TruSight Oncology 500, Illumina, San Diego, CA, USA) panel assay in one study and a pooled overall prevalence of 14% on panel-based or WES assays in solid tumors [5][6]. Among less common tumors, including but not limited to biliary tract cancers, small cell lung cancer, and mesothelioma, the prevalence of TMB-H (≥10 mut/Mb on FoundationOne®CDx (F1CDx) panels) was noted to be 12.8% [7].

2. Challenges of Using TMB as Biomarker of Response to ICI

Whole-exome sequencing (WES) encompasses 32 Mb of the coding region, the entire set of 22,000 genes, which constitutes roughly 1% of the genome. WES, the gold standard for TMB calculation, measures the number of somatic alterations, whereas the FDA-authorized surrogate panel assays including the Memorial Sloan-Kettering-Integrated Mutation Profiling of Actionable Cancer Targets or MSK-IMPACT (468 genes) and F1CDx assay (324 genes) measure the density of somatic alterations, including target footprints of approximately 1.14 Mb and 0.8 Mb of the coding regions, respectively [8][9].
Keynote-158, the pivotal trial leading to the tissue-agnostic approval of pembrolizumab, evaluated tissue TMB (tTMB) in FFPE (Formalin-Fixed Paraffin-Embedded) tumor samples through the utilization of the F1CDx assay with a predefined criterion for classifying high tTMB as the presence of a minimum of 10 mut/Mb. The trial showed an objective response rate (ORR) of 29% in the tTMB-high versus 6% in the non-tTMB-high groups when treated with pembrolizumab at 200 mg IV (intravenously) every 3 weeks until unacceptable toxicity or disease progression [10].
The overall survival (OS) and progression-free survival (PFS) were secondary outcomes in the trial, and a mOS (median OS) benefit was not seen (11.5 months in t-TMB-high versus 12.8 months in non-tTMB-high group). The trial was certainly limited in the types of cancers (it included nine different cancer types) and patient numbers, including cancers with relative resistance to immunotherapy. Fourteen percent of patients assessed for efficacy in the TMB-high group also had Microsatellite Instability-High (MSI-H); however, after excluding patients with missing or MSI-H status, the ORR was 28%.
A retrospective analysis of several keynote trials (Keynote-001, Keynote-002, Keynote-010, Keynote-012, Keynote-028, Keynote-045, Keynote-055, Keynote-059, Keynote-061, Keynote-086, Keynote-100) demonstrated improvement in ORR among patients treated with ICI, with tTMB ≥ 175 mutations/exome compared to TMB < 175 mutations/exome (31.4% vs. 9.5%, respectively). Additionally, an analysis of patients in Keynote-010, Keynote-045, and Keynote-061 showed improvements in PFS and OS compared to chemotherapy [11]. A more recent retrospective analysis also showed that patients in Keynote-042 derived PFS and OS benefits if their tTMB was ≥ 175 mutations/exome compared to tTMB < 175 mutations/exome [12]. Multiple other systematic reviews and metanalyses have indicated a strong association between high TMB and enhanced efficacy, with several studies also highlighting improved survival outcomes across cancers and in specific cancer types [13][14][15]. When it comes to evaluating TMB, inherent differences exist in testing workflow methods (Figure 1).
Figure 1. TMB workflow from tissue specimens to clinical report. The boxes with red outlines are steps that are substantially different in labs and workflows. Abbreviations: NGS, next-generation sequencing; PBMC, peripheral blood mononuclear cells; TMB, Tumor Mutational Burden.
While WES and MSK-IMPACT use similar filtering strategies of using non-synonymous mutations for calculating TMB, tumor-only sequencing panels (like F1CDx) also include short indels in introns and synonymous mutations in the panel testing for TMB. And, considering that neoantigens are formed by non-synonymous mutations only, the synonymous mutations are included in TMB calculation to reduce sampling noise and increase the robustness of TMB scoring methods [16]. In some cases, the use of synonymous mutations in conjunction with a statistical framework substantially increased the concordance of TMB values with a WES-based reference method [17].
Additional differences in the panel content among the commercial panel assays may give rise to inter-assay variations. For example, in focused panels, there may be higher detection rates of pathogenic driver mutations compared to the background mutation rate in the tumor. This may lead to higher levels of variability at low TMB values [18]. Therefore, it may be important to calibrate the thresholds for each panel in addition to generating an adjusted score (e.g., “mutational load”) by taking into account the variability within tumor types [19]. This may help in better interpretation of the TMB scores. Many of these panel-adjusted scores used in determining TMB also show a good correlation to TMB scores generated from WES and WGS [20], whereas the TMB calculation from WES and WGS seems to be highly concordant, although the data analysis pipeline for WES is less intensive and less costly [20][21].
Several factors impact the TMB calculation among different panel tests. The first of these is the size of the panel variation, such as 0.8 Mb in F1CDx versus 1.94 Mb for the TSO500 panel. Studies have shown that a smaller panel size may produce more misclassifications of TMB calculation compared to a larger panel size. This includes variation in thresholds among smaller compared to larger panel sizes. In particular, a smaller panel size is less precise in distinguishing between hypermutated from non-hypermutated cancers [22]. Smaller panel sizes also tend to overestimate TMB values whereas panel tests may overestimate TMB compared to WES [23]
Inter- and intra-tumor heterogeneity can produce imprecise TMB measurement [24]. TMB from metastatic sites could be higher than that from primary sites due to varying clonal heterogeneity, and yet, this disparity may not necessarily influence the survival benefit derived from ICI therapy [25]
Tumor-targeting cytotoxic T lymphocyte variation in the cellular neighborhoods (CNs) is dynamic and impacts response to ICI therapy, with HLA-1 expression downregulation predicting poor outcomes with ICI in colorectal cancer patients [26].
Another challenge lies in the influence of tumor purity, such as that related to tissue sampling, where the low tumor cellularity could result in falsely low TMB measurement [27]. The exclusion of germline alterations also varies across different panels.
The composition of the panel test, with the variable selection of genomic alterations, can also produce variability in TMB calculation [28]. After accounting for artefacts and germline variation, a panel test comparison shows a good correlation with the inclusion of synonymous and coding non-synonymous alterations [29].
Overestimation in TMB can also occur by including mutations with VAF (variant allele frequency) only above a certain threshold [30]. Hence, the specific context of the underlying mutation (synonymous, non-synonymous, or Indels) and whether it occurs in the coding or non-coding regions might contribute to minor variation in TMB calculation across different panels.

3. Factors in Tumor Microenvironment (TME), TMB, and Response to ICI

The tumor microenvironment (TME) constitutes a diverse ecosystem, and to harness the entire TME for improved immunotherapies, it is crucial to recognize that multiple immune subsets play a role in shaping the variability in immune response [31][32]. Thus, these anti-tumor immune responses are complex and involve several factors driving the cancer immunity cycle that promote or suppress anti-tumor immunity [33]. Simply relying on a single biomarker such as TMB to explain the response to ICIs may not capture the intricate interplay of sensitivity and resistance mechanisms underlying the use of these therapies [34].
It is also known that beyond tumor histology, there are several other mechanisms that can impact response to ICIs, such as cellular signaling, checkpoint signaling pathways, immune cell activity, variability in HLA expression and TCR repertoire, the gut microbiome, and oncogenic signaling pathways indirectly associated with response to ICIs [35][36].
Exploration in this field is an active area of clinical and translational research. This involves combining PD-1/PD-L1 (anti-programmed death receptor-1/anti-programmed death ligand-1) inhibitors with other immune-modulating or targeted agents, depending on the stromal environment of the tumor, for example in hot versus cold tumor microenvironments.
A higher HLA class II expression has been shown to be associated with positive tumor responses through correlative analysis in CheckMate 064 and CheckMate 069 [37]. Similarly, heterozygosity in HLA-1 is associated with better response to ICIs [38].
Immunologically cold tumors have lower response rates to ICIs. However, TMB-H is not always correlated with CD8+ tumor-infiltrating T-cells, since a portion of CD8+ T-cells are bystanders and recognize antigens unrelated to tumors [39][40].
A post hoc pan-cancer analysis using MSK-IMPACT for TMB scores showed that the OS among patients treated with ICIs was associated with sets of genomic alterations in TMB-low versus TMB-high tumors. In particular, hypermutation (TMB ≥ 100 mut/mb) is associated with certain genomic signatures including POLE/POLD1, dMMR (deficient mismatch repair), the activation of AID/APOBEC (activation-induced cytidine deaminase/apolipoprotein B mRNA (messenger ribonucleic acid) enzyme catalytic polypeptide-like), and the three clock-like mutational processes (SBS1, SBS5) [18].
Somatic mutations have the potential to generate neoantigens, and the resulting cancer-specific genomic signatures can vary across cancer types [41].
The impact of driver genes on TMB is noteworthy in NSCLC, wherein high TMB may be linked to reduced survival in EGFR mutated cancers [42]. Specific mutations, including CDH1 (cadherin-1), RAD50, and MSH2 (muts Homolog 2), have been associated with high TMB in head and neck squamous cell cancers [43]. Certain mutations linked to responses to ICIs have been observed [44]

4. Efficacy and Real-World Data on TMB Testing

Several clinical trials have demonstrated the clinical utility of TMB as a predictive biomarker of response to single-agent and combination immunotherapy [45]. In general, there is substantial real-world evidence indicating a response and enhanced survival in cases of TMB-H tumors, with varying definitions, but primarily in TMB thresholds ≥ 10 mut/Mb. The analysis of large clinico-genomic databases to assess real-world OS analyses of TMB has shown the benefit of high TMB across 24 cancer types compared to a low TMB [46]. Another retrospective analysis of patients with MSI-H and/or TMB-H (≥20 mut/Mb on F1CDx) among 27 different cancer types showed better PFS outcomes with immunotherapy in patients who had previously received chemotherapy [47]. A study in NSCLC demonstrated an increase in real-world OS as TMB scores increased from <10 to 10–19 and ≥20 mut/Mb (10.1, 11.8, and 26.9 months, respectively) [48].

5. Conclusions

In conclusion, while not a perfect biomarker, with advancements in TMB measurement, standardization initiatives, enhanced testing protocols, TMB characterization in diverse cancer types, its amalgamation with other biomarkers of response to ICIs, dynamic monitoring through bTMB, and the rigorous validation of improved testing methodologies, among various other factors, TMB has the potential for enhanced practical utility in the real-world clinical setting.

References

  1. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 2015, 348, 124–128.
  2. Yuza, K.; Nagahashi, M.; Watanabe, S.; Takabe, K.; Wakai, T. Hypermutation and microsatellite instability in gastrointestinal cancers. Oncotarget 2017, 8, 112103.
  3. Wu, S.; Powers, S.; Zhu, W.; Hannun, Y.A. Substantial contribution of extrinsic risk factors to cancer development. Nature 2016, 529, 43–47.
  4. Samstein, R.M.; Lee, C.-H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019, 51, 202–206.
  5. Jung, J.; Heo, Y.J.; Park, S. High tumor mutational burden predicts favorable response to anti-PD-(L) 1 therapy in patients with solid tumor: A real-world pan-tumor analysis. J. Immunother. Cancer 2023, 11, e006454.
  6. Kang, Y.-J.; O’Haire, S.; Franchini, F.; IJzerman, M.; Zalcberg, J.; Macrae, F.; Canfell, K.; Steinberg, J. A scoping review and meta-analysis on the prevalence of pan-tumour biomarkers (dMMR, MSI, high TMB) in different solid tumours. Sci. Rep. 2022, 12, 20495.
  7. Shao, C.; Li, G.; Huang, L.; Pruitt, S.; Castellanos, E.; Frampton, G.; Carson, K.R.; Snow, T.; Singal, G.; Fabrizio, D. Prevalence of high tumor mutational burden and association with survival in patients with less common solid tumors. JAMA Netw. Open 2020, 3, e2025109.
  8. Milbury, C.A.; Creeden, J.; Yip, W.-K.; Smith, D.L.; Pattani, V.; Maxwell, K.; Sawchyn, B.; Gjoerup, O.; Meng, W.; Skoletsky, J. Clinical and analytical validation of FoundationOne® CDx, a comprehensive genomic profiling assay for solid tumors. PLoS ONE 2022, 17, e0264138.
  9. Cheng, D.T.; Mitchell, T.N.; Zehir, A.; Shah, R.H.; Benayed, R.; Syed, A.; Chandramohan, R.; Liu, Z.Y.; Won, H.H.; Scott, S.N. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 2015, 17, 251–264.
  10. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H. 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.
  11. Cristescu, R.; Aurora-Garg, D.; Albright, A.; Xu, L.; Liu, X.Q.; Loboda, A.; Lang, L.; Jin, F.; Rubin, E.H.; Snyder, A. Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: A pan-tumor retrospective analysis of participants with advanced solid tumors. J. Immunother. Cancer 2022, 10, e003091.
  12. Mok, T.; Lopes, G.; Cho, B.; Kowalski, D.; Kasahara, K.; Wu, Y.-L.; de Castro Jr, G.; Turna, H.; Cristescu, R.; Aurora-Garg, D. Associations of tissue tumor mutational burden and mutational status with clinical outcomes in KEYNOTE-042: Pembrolizumab versus chemotherapy for advanced PD-L1-positive NSCLC. Ann. Oncol. 2023, 34, 377–388.
  13. Wu, Y.; Xu, J.; Du, C.; Wu, Y.; Xia, D.; Lv, W.; Hu, J. 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.
  14. 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, 748674.
  15. Aggarwal, C.; Ben-Shachar, R.; Gao, Y.; Hyun, S.W.; Rivers, Z.; Epstein, C.; Kaneva, K.; Sangli, C.; Nimeiri, H.; Patel, J. Assessment of Tumor Mutational Burden and Outcomes in Patients with Diverse Advanced Cancers Treated with Immunotherapy. JAMA Netw. Open 2023, 6, e2311181.
  16. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34.
  17. Yao, L.; Fu, Y.; Mohiyuddin, M.; Lam, H.Y. ecTMB: A robust method to estimate and classify tumor mutational burden. Sci. Rep. 2020, 10, 4983.
  18. Sha, D.; Jin, Z.; Budczies, J.; Kluck, K.; Stenzinger, A.; Sinicrope, F.A. Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov. 2020, 10, 1808–1825.
  19. Mankor, J.M.; Paats, M.S.; Groenendijk, F.H.; Roepman, P.; Dinjens, W.N.; Dubbink, H.J.; Sleijfer, S.; Consortium, C.; Cuppen, E.; Lolkema, M.P. Impact of panel design and cut-off on tumour mutational burden assessment in metastatic solid tumour samples. Br. J. Cancer 2020, 122, 953–956.
  20. Ruel, L.-J.; Li, Z.; Gaudreault, N.; Henry, C.; Saavedra Armero, V.; Boudreau, D.K.; Zhang, T.; Landi, M.T.; Labbé, C.; Couture, C. Tumor mutational burden by whole-genome sequencing in resected NSCLC of never smokers. Cancer Epidemiol. Biomark. Prev. 2022, 31, 2219–2227.
  21. Vilimas, T. Measuring tumor mutational burden using whole-exome sequencing. In Biomarkers for Immunotherapy of Cancer: Methods and Protocols; Springer: New York, NY, USA, 2020; pp. 63–91.
  22. Buchhalter, I.; Rempel, E.; Endris, V.; Allgäuer, M.; Neumann, O.; Volckmar, A.L.; Kirchner, M.; Leichsenring, J.; Lier, A.; von Winterfeld, M. Size matters: Dissecting key parameters for panel-based tumor mutational burden analysis. Int. J. Cancer 2019, 144, 848–858.
  23. Fang, H.; Bertl, J.; Zhu, X.; Lam, T.C.; Wu, S.; Shih, D.J.; Wong, J.W. Tumour mutational burden is overestimated by target cancer gene panels. J. Natl. Cancer Cent. 2023, 3, 56–64.
  24. Conroy, J.M.; Pabla, S.; Glenn, S.T.; Nesline, M.; Burgher, B.; Lenzo, F.L.; Papanicolau-Sengos, A.; Gardner, M.; Morrison, C. Tumor mutational burden (TMB): Assessment of inter-and intra-tumor heterogeneity. J. Clin. Oncol. 2019, 37, 27.
  25. Schnidrig, D.; Turajlic, S.; Litchfield, K. Tumour mutational burden: Primary versus metastatic tissue creates systematic bias. Immuno-Oncol. Technol. 2019, 4, 8–14.
  26. Schürch, C.M.; Bhate, S.S.; Barlow, G.L.; Phillips, D.J.; Noti, L.; Zlobec, I.; Chu, P.; Black, S.; Demeter, J.; McIlwain, D.R. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 2020, 182, 1341–1359.e1319.
  27. Anagnostou, V.; Niknafs, N.; Marrone, K.; Bruhm, D.C.; White, J.R.; Naidoo, J.; Hummelink, K.; Monkhorst, K.; Lalezari, F.; Lanis, M. Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer. Nat. Cancer 2020, 1, 99–111.
  28. Budczies, J.; Allgäuer, M.; Litchfield, K.; Rempel, E.; Christopoulos, P.; Kazdal, D.; Endris, V.; Thomas, M.; Fröhling, S.; Peters, S. Optimizing panel-based tumor mutational burden (TMB) measurement. Ann. Oncol. 2019, 30, 1496–1506.
  29. Heydt, C.; Rehker, J.; Pappesch, R.; Buhl, T.; Ball, M.; Siebolts, U.; Haak, A.; Lohneis, P.; Büttner, R.; Hillmer, A.M. Analysis of tumor mutational burden: Correlation of five large gene panels with whole exome sequencing. Sci. Rep. 2020, 10, 11387.
  30. Makrooni, M.A.; O’Sullivan, B.; Seoighe, C. Bias and inconsistency in the estimation of tumour mutation burden. BMC Cancer 2022, 22, 840.
  31. Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman, W.H. The clinical role of the TME in solid cancer. Br. J. Cancer 2019, 120, 45–53.
  32. Bejarano, L.; Jordāo, M.J.; Joyce, J.A. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021, 11, 933–959.
  33. Mellman, I.; Chen, D.S.; Powles, T.; Turley, S.J. The cancer-immunity cycle: Indication, genotype, and immunotype. Immunity 2023, 56, 2188–2205.
  34. Nowicki, T.S.; Hu-Lieskovan, S.; Ribas, A. Mechanisms of resistance to PD-1 and PD-L1 blockade. Cancer J. 2018, 24, 47.
  35. Yan, X.; Zhang, S.; Deng, Y.; Wang, P.; Hou, Q.; Xu, H. Prognostic factors for checkpoint inhibitor based immunotherapy: An update with new evidences. Front. Pharmacol. 2018, 9, 1050.
  36. Blank, C.U.; Haanen, J.B.; Ribas, A.; Schumacher, T.N. The “cancer immunogram”. Science 2016, 352, 658–660.
  37. Rodig, S.; Gusenleitner, D.; Jackson, D.; Gjini, E.; Giobbie-Hurder, A.; Jin, C.; Chang, H.; Lovitch, S.; Horak, C.; Weber, J.S. Association of distinct baseline tissue biomarkers with response to nivolumab (NIVO) and ipilimumab (IPI) in melanoma: CheckMate 064. J. Clin. Oncol. 2017, 35, 9515.
  38. Chowell, D.; Morris, L.G.; Grigg, C.M.; Weber, J.K.; Samstein, R.M.; Makarov, V.; Kuo, F.; Kendall, S.M.; Requena, D.; Riaz, N. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018, 359, 582–587.
  39. McGrail, D.; Pilié, P.; Rashid, N.; Voorwerk, L.; Slagter, M.; Kok, M.; Jonasch, E.; Khasraw, M.; Heimberger, A.; Lim, B. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann. Oncol. 2021, 32, 661–672.
  40. Simoni, Y.; Becht, E.; Fehlings, M.; Loh, C.Y.; Koo, S.-L.; Teng, K.W.W.; Yeong, J.P.S.; Nahar, R.; Zhang, T.; Kared, H. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018, 557, 575–579.
  41. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.-L. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421.
  42. Offin, M.; Rizvi, H.; Tenet, M.; Ni, A.; Sanchez-Vega, F.; Li, B.T.; Drilon, A.; Kris, M.G.; Rudin, C.M.; Schultz, N. Tumor mutation burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with EGFR-mutant lung cancers. Clin. Cancer Res. 2019, 25, 1063–1069.
  43. Chen, Y.; Chen, C.-b.; Zheng, X.-b.; Gao, X.; Jun, L.; Xiong, J.-n.; Lin, J.; Xu, Y.; Guan, Y.-F.; Li, Y. Association of driver genes with high-tumor mutation burden and outcome in patients with head and neck cancer: Implications for immunotherapy. J. Clin. Oncol. 2020, 38, e18533.
  44. Sholl, L.M.; Hirsch, F.R.; Hwang, D.; Botling, J.; Lopez-Rios, F.; Bubendorf, L.; Mino-Kenudson, M.; Roden, A.C.; Beasley, M.B.; Borczuk, A. 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, 1409–1424.
  45. Chan, T.A.; Yarchoan, M.; Jaffee, E.; Swanton, C.; Quezada, S.A.; Stenzinger, A.; Peters, S. Development of tumor mutation burden as an immunotherapy biomarker: Utility for the oncology clinic. Ann. Oncol. 2019, 30, 44–56.
  46. Gandara, D.R.; Agarwal, N.; Gupta, S.; Klempner, S.J.; Andrews, M.C.; Mahipal, A.; Subbiah, V.; Eskander, R.N.; Carbone, D.P.; Snider, J. Tumor mutational burden (TMB) measurement from an FDA-approved assay and real-world overall survival (rwOS) on single-agent immune checkpoint inhibitors (ICI) in over 8,000 patients across 24 cancer types. J. Clin. Oncol. 2023, 41, 2503.
  47. Palmeri, M.; Mehnert, J.; Silk, A.; Jabbour, S.; Ganesan, S.; Popli, P.; Riedlinger, G.; Stephenson, R.; de Meritens, A.; Leiser, A. Real-world application of tumor mutational burden-high (TMB-high) and microsatellite instability (MSI) confirms their utility as immunotherapy biomarkers. ESMO Open 2022, 7, 100336.
  48. Huang, R.S.; Carbone, D.P.; Li, G.; Schrock, A.; Graf, R.P.; Zhang, L.; Murugesan, K.; Ross, J.S.; Tolba, K.; Sands, J. Durable responders in advanced NSCLC with elevated TMB and treated with 1L immune checkpoint inhibitor: A real-world outcomes analysis. J. Immunother. Cancer 2023, 11, e005801.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 208
Revisions: 3 times (View History)
Update Date: 22 Dec 2023
1000/1000
Video Production Service