Fact-checked by Grok 2 weeks ago

Tumor mutational burden

Tumor mutational burden (TMB) is defined as the total number of per megabase of interrogated genomic in a tumor's DNA, reflecting the extent of genomic instability and potential for generating neoantigens that can elicit an . This quantitative measure serves as a key predictive for the efficacy of inhibitors (ICIs), such as anti-PD-1/ therapies, particularly in solid tumors where higher TMB levels correlate with improved response rates and survival outcomes. For instance, the U.S. (FDA) approved in 2020 for adult and pediatric patients with unresectable or metastatic solid tumors exhibiting a TMB of ≥10 per megabase, based on data from clinical trials like KEYNOTE-158. TMB is typically assessed through next-generation sequencing (NGS) of tumor tissue, with whole exome sequencing (WES) serving as the gold standard by covering approximately 30 megabases of coding regions, though targeted gene panels (e.g., 0.8–2.4 Mb in size, such as MSK-IMPACT or FoundationOne CDx) are more commonly used in clinical settings for their efficiency and cost-effectiveness. These panels focus on hundreds of cancer-related genes and calculate TMB by counting non-synonymous somatic variants, often excluding germline mutations, driver alterations, and sometimes synonymous changes to emphasize passenger mutations that drive immunogenicity. Blood-based TMB (bTMB), derived from circulating tumor DNA, offers a non-invasive alternative, showing concordance with tissue TMB and utility in monitoring treatment response, though it requires validation for broader adoption. Clinically, TMB varies significantly across tumor types, with higher burdens observed in cancers like melanoma (median ~71% high TMB cases), non-small cell lung cancer (44–50%), and microsatellite instability-high (MSI-H) tumors, while lower levels predominate in pediatric cancers, which have notably low TMB (typically <1 mut/Mb, with an average of ~9.6 total nonsynonymous mutations per tumor), and breast cancer. Approximately 13.2% of patients in the KEYNOTE-158 trial qualified as TMB-high, highlighting its relevance in stratifying patients for ICI therapy across pan-cancer settings. Despite these advances, challenges persist, including the lack of universal cutoffs (typically 10–20 mut/Mb but tumor-type dependent), variability in assay performance due to panel size and bioinformatics pipelines, and issues with tumor purity, intratumor heterogeneity, and sample quality that can lead to inconsistent reporting. As of 2025, emerging research continues to refine TMB cutoffs, questioning the one-size-fits-all approach of 10 mut/Mb for optimal immunotherapy prediction. Ongoing standardization efforts, such as those by Friends of Cancer Research and the Quality Assurance Initiative Pathology, aim to harmonize TMB assessment by recommending panels of at least 1 Mb and alignment with WES benchmarks to ensure reliable clinical decision-making.

Definition and Background

Core Concept of TMB

Tumor mutational burden (TMB) quantifies the density of within a tumor's , specifically the total number of non-synonymous —such as missense, , and frameshift variants—per megabase of sequenced or , while excluding variants. This metric focuses on coding regions where can alter protein sequences, providing a measure of the tumor's genomic and potential for immune recognition. The biological foundation of TMB lies in its role as a for neoantigen load, where mutations generate novel peptides that are processed and presented on the tumor cell surface via (MHC) molecules. These neoantigens can be recognized by T-cells, thereby enhancing the tumor's and facilitating immune-mediated destruction. High TMB thus correlates with increased likelihood of effective antitumor immune responses, as more mutations diversify the neoantigen repertoire available for T-cell targeting. TMB is typically expressed in units of mutations per megabase (mut/Mb), allowing for standardized comparisons across samples. Measurements can differ based on the sequencing approach: whole exome sequencing (WES) provides a comprehensive assessment of the ~30-60 Mb coding genome, yielding a direct mut/Mb value, whereas targeted gene panels interrogate smaller subsets of genes and often require algorithmic to estimate equivalent TMB scores. For instance, in non-small cell (NSCLC), median TMB values range from approximately 5-10 mut/Mb among smokers, reflecting tobacco-induced , compared to lower levels around 4 mut/Mb in never-smokers. High TMB has been linked to enhanced responses in , underscoring its relevance in precision oncology.

Historical Development

The concept of tumor mutational burden (TMB) emerged in the early 2010s through large-scale genomic initiatives like (TCGA), which began sequencing tumor exomes across multiple cancer types starting in 2006 and published pan-cancer analyses by 2013. These efforts revealed wide variations in loads among cancers, with initial observations linking higher mutation burdens to increased immune cell infiltration, such as CD8+ T cells, suggesting a potential role in antitumor immunity. By cataloging thousands of tumors, TCGA data laid the groundwork for understanding TMB as a quantitative measure of genomic instability, influencing subsequent research on neoantigen generation and immune recognition. Pivotal clinical studies in the mid-2010s established TMB's association with immunotherapy responses. In 2014, Snyder et al. analyzed exomes from melanoma patients treated with ipilimumab, a CTLA-4 inhibitor, finding that responders had significantly higher somatic mutation burdens compared to non-responders, correlating with neoantigen load and clinical benefit. This was followed in 2015 by Rizvi et al., who examined non-small cell lung cancer (NSCLC) tumors from patients receiving pembrolizumab, a PD-1 inhibitor, and reported that higher nonsynonymous mutation burdens (median 12.4 in responders vs. 6.7 in non-responders) predicted durable responses, independent of smoking history. These findings shifted TMB from a descriptive genomic metric to a candidate predictive biomarker for immune checkpoint blockade. Regulatory milestones accelerated TMB's clinical adoption. In June 2020, the U.S. (FDA) granted accelerated approval for as a tumor-agnostic in adult and pediatric patients with unresectable or metastatic solid tumors characterized by TMB-high status (≥10 mutations per megabase), based on data from the KEYNOTE-158 trial showing an objective response rate of 29.3% in this group. Concurrently, efforts by the Friends of TMB Harmonization Project, launched in 2017, addressed variability in TMB measurement across through multi-lab collaborations and reference standards, promoting standardization from a research tool to a reproducible clinical . By 2024, ongoing initiatives expanded TMB's utility in combination , with subgroup analyses in trials like KEYNOTE-942 showing benefits across TMB and statuses in . In 2023, a phase 3 trial (KEYNOTE-B15) was initiated to further evaluate mRNA-4157 plus in high-risk . As of 2025, research explores acquired high TMB post-targeted to enhance sensitivity.

Clinical Significance

Biomarker Role in Immunotherapy

Tumor mutational burden (TMB) serves as a predictive for response to inhibitors (ICIs), with the U.S. (FDA) granting accelerated approval in June 2020 for in adult and pediatric patients with unresectable or metastatic tumors exhibiting high TMB (≥10 mutations per megabase, mut/Mb), as determined by an FDA-approved companion diagnostic test. This tumor-agnostic approval highlights TMB's role independent of programmed death-ligand 1 () expression, enabling broader application across cancer types where ICI efficacy was previously limited by status. Mechanistically, elevated TMB correlates with a higher neoantigen burden, as mutations generate novel peptides presented on molecules, thereby enhancing T-cell recognition and infiltration into the . This increased promotes (TILs), particularly cytotoxic T cells, which synergize with ICIs such as anti-PD-1 and anti-PD-L1 therapies to reinvigorate exhausted immune responses and improve antitumor activity. Consequently, tumors with high TMB exhibit greater sensitivity to checkpoint blockade compared to those with low TMB, where neoantigen scarcity limits immune activation. Meta-analyses of patients treated with ICIs substantiate TMB's predictive value, demonstrating that high TMB cohorts achieve significantly better (PFS) than low TMB groups, with pooled hazard ratios (s) ranging from 0.45 to 0.54. For instance, one analysis of over 4,500 patients across multiple studies reported an of 0.45 (95% : 0.36–0.56) for PFS in high versus low TMB, underscoring TMB's association with prolonged ICI benefit. Another in non-small cell lung cancer confirmed an of 0.54 (95% : 0.46–0.63), reinforcing TMB's utility in stratifying ICI responders. TMB's predictive strength varies by cancer type, proving more robust in non-smoker-associated malignancies such as instability-high (MSI-H) colorectal cancer, where high TMB aligns closely with enhanced ICI responses due to shared defect-driven hypermutation. In contrast, its predictive value is attenuated in smoker-linked cancers like lung adenocarcinoma, where environmental mutagens confound TMB's direct correlation with neoantigen and ICI . This specificity highlights TMB's contextual limitations as a pan-cancer , emphasizing the need for integrated assessments in diverse tumor etiologies.

Predictive Value for Treatment Response

Tumor mutational burden (TMB) serves as a predictive for response to , particularly inhibitors, with high TMB levels associated with improved objective response rates (ORR), (PFS), and overall survival (OS) in various clinical trials. In the phase 2 KEYNOTE-158 trial evaluating in patients with advanced tumors, those with high TMB (≥10 mutations per megabase) demonstrated an ORR of 29% compared to 10% in patients with low TMB, highlighting TMB's utility across tumor types independent of status. This trial's results supported the FDA approval of for TMB-high tumors, underscoring the biomarker’s role in identifying responders to PD-1 inhibition. Recent updates from trials in non-small cell lung cancer (NSCLC) further affirm TMB's predictive value. In the phase 3 227 trial, patients with high TMB treated with nivolumab plus showed durable OS benefits, with 5-year OS rates exceeding those in arms, particularly in TMB-high subgroups. Analyses from the CA209-7AL trial in unresectable stage III NSCLC revealed that high TMB patients receiving consolidative nivolumab after achieved significantly longer PFS (not reached versus 15.2 months in low TMB; p=0.042). These findings extend TMB's relevance to and consolidative settings, where high TMB predicted enhanced treatment efficacy. High TMB correlates with higher rates of complete and partial responses, as well as extended duration of response (DOR). For instance, in patients treated with PD-1 inhibitors, high TMB subgroups exhibited ORRs up to 45%, reflecting increased neoantigen load and immune activation. Across trials like KEYNOTE-158, DOR in TMB-high patients was notably prolonged, often not reached at median follow-up, compared to 12-18 months in low TMB cohorts, representing an extension of 6-12 months or more in responsive cases. This benefit is most pronounced with PD-1 inhibitors such as and nivolumab, where high TMB independently predicts superior outcomes, whereas responses to CTLA-4 inhibitors like alone are weaker and less TMB-dependent. Subgroup analyses emphasize TMB's value beyond microsatellite instability-high tumors. In microsatellite-stable (MSS) tumors with high TMB, yields significant benefits, with ORRs comparable to MSI-high cases and improved PFS in PD-1-treated patients. For rare cancers like small cell (SCLC), a 2025 real-world analysis across multiple cancers including SCLC showed a non-significant trend toward better overall survival with high TMB (HR 0.89, 95% CI 0.44-1.09) on inhibition, supporting further evaluation of TMB in metastatic SCLC . These insights support TMB-high/MSS identification for expanded access in underrepresented tumor types.

Prognostic Implications

Tumor mutational burden (TMB) serves as an intrinsic prognostic in cancer patients, particularly in cohorts not receiving , where high TMB frequently correlates with poorer overall survival due to underlying tumor aggressiveness. In a systematic pan-cancer evaluation of (TCGA) data encompassing 6,035 patients across 20 cancer types, high TMB was significantly associated with worse overall survival in 8 cancers, including (hazard ratio [HR] 2.47, 95% CI 1.02–5.98, P=0.045), (HR 6.10, 95% CI 1.91–19.46, P=0.002), colorectal adenocarcinoma, esophageal carcinoma, kidney renal clear cell carcinoma, liver , , and pancreatic adenocarcinoma. Conversely, high TMB predicted better survival in 6 cancer types, such as bladder urothelial carcinoma and kidney renal papillary cell carcinoma, while showing no significant impact in the remaining 6. This divergent pattern underscores TMB's context-specific prognostic utility for risk stratification independent of treatment. In immunotherapy-naïve patients with advanced malignancies, TMB exhibits a nonlinear relationship with , where intermediate levels (>5 and <20 mutations/Mb) confer the worst outcomes compared to low (≤5 mutations/Mb) or high (≥20 mutations/Mb) burdens. Among 1,415 such patients, those with intermediate TMB had a median overall of 174 weeks (HR 1.29, 95% CI 1.07–1.54, P<0.01) versus 238 weeks for low TMB, while high TMB yielded 195 weeks (HR 0.98, P=0.90), suggesting that moderate mutational loads may reflect suboptimal immune equilibrium without therapeutic intervention. Although high TMB often signals favorable responses in immunotherapy contexts, its prognostic implications in chemotherapy-only or untreated settings are generally adverse, as evidenced by mixed signals in pan-cancer analyses where elevated TMB aligns with reduced in over half of evaluated cohorts. The adverse prognostic role of high TMB in non-immunotherapy scenarios stems from its reflection of genomic instability, driven by defects in DNA repair pathways such as and , which foster tumor proliferation, chromosomal aberrations, and heightened metastasis risk. This instability correlates with aggressive tumor biology, including increased cell cycle activity and structural variants that promote invasive growth, thereby worsening patient outcomes independent of immune checkpoint modulation. For instance, high TMB also enables neoantigen generation that could enhance immune surveillance, but in untreated settings, the dominant effect is unchecked genomic chaos leading to rapid disease progression. Specific examples highlight TMB's variable prognostic weight across histologies. In gliomas, elevated TMB is linked to shorter overall survival, with higher burdens associating with advanced grade, older age, and structural mutations that exacerbate poor outcomes in this immunologically "cold" tumor type. In contrast, breast cancer demonstrates largely neutral TMB-prognosis associations in non-immunotherapy cohorts, where high TMB does not independently influence survival unless integrated with factors like immune infiltration or specific gene signatures, reflecting the hormone-driven stability of many breast tumors.

Variation Across Cancer Types

Cancers with High TMB

Tumor mutational burden (TMB) is considered high when exceeding 10 mutations per megabase (mut/Mb), a threshold commonly used for classification in clinical and research settings. Approximately 13% of solid tumors across various cohorts meet this criterion, highlighting a subset of malignancies with elevated genomic instability that may influence therapeutic strategies. Among cancers frequently exhibiting high TMB, cutaneous melanoma stands out with median values of 10-14 mut/Mb, primarily driven by ultraviolet (UV) radiation exposure that induces characteristic C>T transitions. Non-small cell lung cancer (NSCLC) in smokers also shows elevated TMB, with medians around 10-12 mut/Mb attributable to mutagens causing G>T transversions. instability-high (MSI-high) colorectal cancers, resulting from deficiency, often display markedly higher TMB levels exceeding 50 mut/Mb, reflecting hypermutation due to unrepaired replication errors. Endometrial cancers harboring proofreading domain mutations represent another example, where these hereditary alterations lead to ultrahigh TMB often surpassing 100 mut/Mb through polymerase infidelity. Elevated TMB in these cancers arises from diverse etiologies, including environmental factors like UV light in and in NSCLC, endogenous processes such as cytidine deaminase hyperactivity contributing to clustered mutations in multiple tumor types, and hereditary defects like POLE mutations in endometrial carcinoma. Virus-negative , distinct from polyomavirus-positive cases, exhibits high TMB (often >20 mut/Mb) linked to UV signatures, with 2025 analyses confirming its association with aggressive disease and potential responsiveness. Clinically, high-TMB cancers demonstrate improved outcomes with inhibitors (ICIs), with objective response rates of approximately 20-30% in pan-solid tumor cohorts treated with anti-PD-1/ therapies, underscoring TMB's role as a predictive .

Cancers with Low TMB

Tumor mutational burden (TMB) is generally low in the majority of adult solid tumors, reflecting limited exposure to environmental mutagens and proficient mechanisms across these cancer types. Pediatric tumors characteristically display very low TMB, often below mut/Mb, as evidenced by pan-cancer analyses reporting medians as low as 0.09 mut/Mb, attributed to their embryonal origins and reduced lifetime accumulation of mutations compared to adult cancers. Similarly, IDH-mutant gliomas maintain low TMB levels, typically ranging from to 3 mut/Mb, with the vast majority below 2 mut/Mb, due to intact mismatch repair pathways and limited hypermutator phenotypes in these tumors. Prostate cancers also feature low TMB, averaging 2 to 4 mut/Mb and driven primarily by signaling rather than mutagenic processes, further supported by high-TMB subsets being rare (approximately 2–5% of cases). The inherently low TMB in these cancers stems from multiple factors, including minimal exposure to exogenous mutagens such as UV radiation or , which contrasts with high-TMB tumors like melanomas or lung cancers. Efficient mechanisms, such as wild-type BRCA1/2 status and proficient mismatch repair, prevent mutation accumulation in low-TMB cancers like and IDH-mutant gliomas. Additionally, suppressive tumor microenvironments in these immunologically "cold" tumors, characterized by low T-cell infiltration, further limit neoantigen presentation and mutational evolution. Clinically, low-TMB cancers exhibit poorer responses to inhibitors (ICIs), with objective response rates often below 10%, as demonstrated in pan-tumor cohorts where low-TMB groups showed only 6-9% efficacy compared to higher rates in TMB-elevated subsets. In low-TMB , 2024 analyses have highlighted alternative biomarkers, such as T-cell inflamed signatures, which correlate with improved and potential ICI benefit independent of TMB, emphasizing the need for multifaceted immune profiling in therapy selection. These characteristics underscore the challenges in applying TMB-based immunotherapy to low-TMB cancers, prompting exploration of targeted therapies tailored to their molecular drivers.

Measurement Methods

Sequencing Approaches

Tumor mutational burden (TMB) assessment relies on genomic sequencing to detect mutations in tumor DNA, with various approaches balancing comprehensiveness, cost, and clinical feasibility. The primary methods include whole exome sequencing (WES), targeted next-generation sequencing (NGS) panels, and (WGS), each leveraging high-throughput platforms to quantify mutations per megabase in coding regions. These techniques typically require paired tumor-normal samples to distinguish from variants, though tumor-only approaches are increasingly used with computational adjustments. Whole exome sequencing (WES) serves as the gold standard for TMB measurement due to its unbiased capture of protein-coding variants across approximately 2% of the , focusing on the ~20,000 genes and 30-60 megabases of exonic sequence. To ensure accurate detection of low-frequency subclonal in heterogeneous tumors, WES typically demands high sequencing depth of 100-300x coverage for both tumor and matched normal samples. This method provides a of without , making it for research benchmarks, though its broader genomic increases volume and processing demands compared to targeted alternatives. In clinical settings, targeted NGS panels have become the standard for TMB evaluation, interrogating 300-500 cancer-relevant genes to assess mutations within a focused genomic space of 1-2 megabases, which is then extrapolated to estimate whole-exome equivalents. Prominent examples include the MSK-IMPACT panel, which sequences 505 genes at depths exceeding 500x, and FoundationOne CDx, a 324-gene approved by the FDA for diagnostics, both demonstrating strong concordance (r > 0.9) with WES-derived TMB values in validation studies. These panels prioritize actionable alterations alongside TMB, enabling efficient integration into routine workflows while minimizing off-target noise from non-coding regions. Whole genome sequencing (WGS) offers the most comprehensive TMB assessment by capturing all mutations across the entire 3-billion-base-pair , including non-coding variants that may influence tumor or immune recognition. However, its resource-intensive nature—requiring terabytes of , extended computational runtimes, and costs 10-100 times higher than WES—limits routine adoption to specialized research cohorts, such as those exploring pan-cancer mutational landscapes. WGS excels in detecting indels, copy number variations, and structural rearrangements that contribute to overall mutational load but is less practical for high-throughput clinical TMB screening. Most TMB sequencing employs short-read platforms like Illumina's NovaSeq systems, which generate 100-300 base-pair reads with >99.9% accuracy, supporting the high-depth requirements of WES and targeted panels through sequencing. Emerging long-read technologies, such as PacBio's HiFi sequencing, are gaining traction for resolving complex structural variants and repetitive regions that short-read methods struggle with, potentially enhancing TMB accuracy in structurally heterogeneous tumors by 2025. Additionally, liquid approaches using (ctDNA) are seeing increased adoption by late 2025, with NGS-based assays like those from Guardant Health enabling non-invasive TMB monitoring from blood samples, though remains lower (detecting down to approximately 0.1–0.5% frequencies) compared to tissue-based methods.

Calculation Formulas and Pipelines

Tumor mutational burden (TMB) is fundamentally calculated as the number of non-synonymous per megabase (Mb) of the , excluding synonymous , intronic variants, and alterations unless otherwise specified in the . For whole-exome sequencing (WES), the size is typically standardized at 38 Mb to enable consistent across samples. The core formula can be expressed as: \text{TMB} = \frac{\sum \text{somatic non-synonymous mutations}}{38} where the denominator represents the size in , yielding mutations per Mb (mut/Mb). The computational pipeline for deriving TMB from sequencing data involves several sequential steps to ensure accuracy and reproducibility. Initial read alignment is performed using tools like the Genome Analysis Toolkit (GATK) to map tumor and matched normal sequences to a , followed by preprocessing to mark duplicates and perform base quality score recalibration. Variant calling then identifies single-nucleotide variants (SNVs) and insertions/deletions (indels), commonly employing MuTect2, which models tumor-specific noise and compares against the matched normal to distinguish from events. filtering is applied using resources like the Genome Aggregation Database (gnomAD) to remove common polymorphisms, retaining only high-confidence calls. of variants occurs next, often with the Variant Effect Predictor (VEP) tool, to classify mutations as non-synonymous (e.g., missense, ) and filter out non-coding or synonymous changes. Finally, burden normalization computes TMB by dividing the count of qualifying mutations by the effectively covered size in , accounting for sequencing depth and panel-specific regions if not using WES. Adjustments to the standard TMB calculation may incorporate distinctions between and mutations, as well as clonal versus subclonal variants, to refine estimates for clinical relevance. While TMB traditionally includes both (neutral) and (oncogenic) mutations to reflect overall neoantigen potential, some pipelines exclude known drivers or apply weights to emphasize passengers, which constitute the majority of events. For clonality, subclonal mutations (present in tumor subpopulations) are often downweighted or excluded relative to clonal ones, as they may dilute predictive signals for response. Tumor purity adjustments are critical, particularly in heterogeneous samples, using an extended formula such as: \text{TMB}_{\text{adjusted}} = \frac{\sum \text{somatic mutations}}{\text{covered Mb} \times \text{purity estimate}} where purity is derived from tools like FACETS or ABSOLUTE to correct for stromal contamination. Recent standards emphasize pipeline reproducibility, with the 2024 Association for Molecular Pathology (AMP), American Society of Clinical Oncology (ASCO), College of American Pathologists (CAP), and Society for Immunotherapy of Cancer (SITC) consensus recommendations advocating detailed documentation of variant calling thresholds, filtering criteria, and normalization methods to minimize inter-assay variability. Custom scripts, often integrated with GATK workflows, handle Mb normalization, while open-source tools like TMBcalc provide end-to-end pipelines for pan-cancer TMB computation, ensuring alignment with these guidelines.

Influencing Factors and Standardization

Biological and Technical Variables

Tumor purity, defined as the proportion of cancer cells within a tumor sample relative to non-cancerous components such as stromal or inflammatory cells, significantly influences tumor mutational burden (TMB) estimation. Low tumor purity dilutes the signal from somatic mutations, leading to underestimation of TMB, as non-tumor cells contribute fewer mutations and reduce the variant allele frequency (VAF) of true tumor variants. Guidelines recommend a minimum tumor purity of 20% for reliable tissue-based panel sequencing assays to ensure sufficient sensitivity in detecting mutations above a typical 5% VAF threshold. Computational tools like ABSOLUTE can estimate tumor purity from sequencing data by integrating copy number alterations and allele frequencies, enabling purity-adjusted TMB calculations that improve accuracy in samples with 15-40% purity. Infiltration by immune or tumor microenvironment (TME) cells further complicates estimates, as lower purity correlates with reduced TMB sensitivity and potential false negatives in immunotherapy response prediction. Tumor heterogeneity, both spatial and temporal, introduces variability in TMB measurements by causing differences in mutation profiles across tumor regions or over time. Spatial heterogeneity arises from subclonal evolution, where distinct tumor areas harbor varying mutation burdens, leading to discordant TMB values between biopsy sites; for instance, in pulmonary adenocarcinoma, intratumor regions can show marked differences in somatic mutations that affect panel-based TMB assessments. Temporal heterogeneity, driven by treatment effects or disease progression, can alter TMB as new mutations emerge or subclones expand, as observed in gastroesophageal adenocarcinoma where baseline and post-treatment samples exhibited heterogeneous TMB and PD-L1 expression. This variability underscores the need for multi-region sampling to capture representative TMB, though single biopsies may underestimate overall burden due to sampling bias. Sequencing coverage, or depth, is a critical variable affecting TMB accuracy, as insufficient depth fails to detect low-frequency while uneven coverage biases detection toward mutation hotspots. A minimum coverage of 100x is generally required for reliable variant calling in TMB assays, with 250x or higher recommended for subclonal or low-VAF to minimize false negatives; however, TMB estimates remain sensitive to depth variations, with lower depths leading to inconsistent counts above typical thresholds like 5% VAF. High-depth sequencing, such as >750x in targeted panels, enhances stability but increases costs, particularly for heterogeneous tumors where uniform coverage across the is challenging. Preprocessing factors, including sample fixation and DNA quality, can introduce artifacts that skew TMB results. Formalin-fixed paraffin-embedded (FFPE) tissues, commonly used in clinical settings, cause DNA damage via deamination, resulting in artifactual C>T transitions that inflate TMB by mimicking mutations. Fresh frozen samples are preferred to avoid these artifacts, as they yield higher-quality DNA with fewer false positives; FFPE-derived DNA requires quality metrics like fragment lengths exceeding 150 to ensure reliable library preparation and sequencing. Microbial in sequencing reads, often from reagents or environmental sources, can further bias TMB by introducing non-human variants or diluting tumor signal if not filtered, necessitating computational pipelines to maintain estimate integrity.

Cutoff Determination and Harmonization

Determining appropriate cutoffs for tumor mutational burden (TMB) is essential for identifying patients likely to benefit from , with the U.S. (FDA) establishing a threshold of ≥10 mutations per megabase (mut/Mb) for high TMB in solid tumors based on the KEYNOTE-158 trial, which supported approval of for this group. Cancer-specific cutoffs often vary to account for inherent differences in mutational landscapes; for instance, studies frequently employ a higher threshold of 20 mut/Mb to classify high TMB, reflecting its elevated baseline mutation rates compared to other cancers. Additionally, percentile-based approaches, such as designating the top 20% of TMB values within a specific cancer type as high, provide a relative framework that adapts to population-level data and enhances prognostic relevance across diverse cohorts. Efforts to harmonize TMB measurements address discrepancies arising from differences, with the Friends of Cancer Research (FOCR) TMB Harmonization Project, culminating in key findings by 2024, demonstrating that inter-laboratory comparisons exhibit 20-30% variability due to panel size, gene coverage, and bioinformatics pipelines. Phase II of this initiative developed calibration models to align targeted next-generation sequencing panels with whole-exome sequencing (WES) references, promoting consistent TMB estimation and reducing classification discordance for high versus low TMB. Validation of TMB assays relies on proficiency testing programs, such as those offered by the (), which evaluate laboratory performance using standardized samples to ensure reliable quantification across clinical settings. Reference materials, including those developed through FOCR collaborations, facilitate calibration by providing benchmarks for detection, minimizing technical biases in TMB reporting. Recent guidelines from the Association for Molecular Pathology (AMP), CAP, and Society for Immunotherapy of Cancer integrate WES as a gold standard with panel-based assays, demonstrating high correlation (often >0.85) between WES and panel-based TMB measurements when appropriate calibration is applied, thereby supporting broader clinical adoption and comparability.

Challenges and Limitations

Assay and Interpretation Issues

One major challenge in TMB assays is , particularly the discordance between targeted next-generation sequencing (NGS) panels and whole-exome sequencing (WES), which can reach up to 40% due to differences in coverage and variant detection . Batch effects in NGS further compromise , as variations in library preparation, sequencing depth, and bioinformatics pipelines can lead to inconsistent TMB estimates across runs or laboratories. Recent 2024 studies have highlighted that formalin-fixed paraffin-embedded (FFPE) artifacts, such as deamination-induced C>T transitions, contribute to false-high TMB calls, necessitating stringent variant (VAF) thresholds to mitigate these errors. Interpretation of TMB results is prone to that can distort clinical . Overcounting mutations in hypermutated regions, often driven by positive selection in cancer genes captured by targeted panels, leads to systematic overestimation of TMB compared to genome-wide assessments. Ignoring tumor exacerbates this, as spatial and temporal heterogeneity—resulting from clonal and neoantigen loss—causes TMB variability across tumor sites or over time, potentially misrepresenting . Liquid biopsy approaches, reliant on (ctDNA), frequently underestimate TMB due to variable shedding rates, with high TMB rarely detectable below 1% ctDNA fraction, limiting their utility in low-shedding tumors. Validation gaps persist in TMB assays, underscored by the scarcity of prospective trials demonstrating consistent predictive value across diverse cohorts. Inter-assay variability compounds this, arising from variations in size, germline filtering, and synonymous mutation inclusion, as seen between platforms like FoundationOne CDx and MSK-IMPACT. Emerging issues involve integration in variant calling, where biases toward high-confidence calls can amplify errors in heterogeneous samples. 2025 reports indicate models are susceptible to in low-coverage data, reducing accuracy in TMB quantification for suboptimal sequencing inputs like those from liquid biopsies. These challenges highlight the need for harmonized protocols to enhance assay reliability, as explored in ongoing efforts.

Clinical and Regulatory Hurdles

Despite its promise as a predictive for response, tumor mutational burden (TMB) exhibits inconsistent pan-cancer performance, limiting its clinical utility in routine practice. For instance, while high TMB correlates with improved outcomes in certain immunogenic tumors like non-small cell lung cancer and , it fails to reliably predict responses in others, such as those with low TMB but alternative responsiveness mechanisms, including microsatellite instability-high (MSI-H) colorectal cancers where neoantigen load drives efficacy independently of TMB levels. Moreover, retrospective analyses underpinning key approvals, such as the KEYNOTE-158 trial for , introduce selection biases that overestimate TMB's predictive value, as post-hoc assessments may not capture real-world variability in patient cohorts. These limitations highlight evidence gaps, with response rates in high-TMB cohorts (≥20 mutations/Mb) varying across studies, often around 30-60%. Regulatory hurdles further impede TMB's integration into clinical guidelines. The U.S. (FDA) granted accelerated approval in 2020 for in adults and pediatric patients with unresectable or metastatic TMB-high (≥10 mutations/Mb) solid tumors that have progressed following prior treatments and lack satisfactory alternatives, based on the KEYNOTE-158 basket trial demonstrating a 29% response rate. However, this remains an accelerated approval pending confirmatory trials, and controversies persist regarding the trial's reliance on a single companion diagnostic assay (FoundationOne CDx), raising concerns over generalizability. In contrast, the (EMA) has not approved or any agent specifically for TMB-high indications, leading to regulatory variations across countries where TMB testing lacks harmonized endorsement and reimbursement pathways differ significantly. These discrepancies complicate global adoption, as evidenced by ongoing debates in 2024 about TMB's agnostic status versus the need for tumor-specific validations. Adoption barriers exacerbate these challenges, primarily driven by high testing costs and limited accessibility. Next-generation sequencing-based TMB assessments typically range from $3,000 to $5,000 per test in the U.S. as of 2025, often exceeding $5,000 when bundled with comprehensive genomic profiling, straining healthcare budgets and deterring widespread use. In low-resource settings, inadequate and mechanisms further restrict , with surveys indicating that up to 59% of barriers to advanced sequencing stem from funding issues. Additionally, the requirement for FDA-approved companion diagnostics like FoundationOne CDx mandates specialized labs, creating logistical hurdles in diverse clinical environments. Ethical concerns around over-testing have intensified in , as inconsistent TMB cutoffs and assay variability may lead to unnecessary in non-responders, raising issues of , financial burden, and potential harm without clear prospective evidence. Debates continue on whether TMB should serve as a sole or be incorporated into composite scores with expression or to mitigate these risks.

Future Directions

Integration with Other Biomarkers

Tumor mutational burden (TMB) is increasingly integrated with other biomarkers to enhance its predictive utility for response in , addressing limitations such as variability across tumor types and incomplete correlation with clinical outcomes. Combining TMB with expression has shown improved prognostic accuracy; for instance, a composite score incorporating TMB, PD-L1 on immune cells, and CD39 expression achieved area under the curve (AUC) values of 0.649 for 12-month overall survival and 0.674 for 24-month survival in muscle-invasive patients treated with PD-L1 blockade, outperforming individual markers. Similarly, in advanced urothelial , high TMB (≥175 mutations per ) combined with PD-L1 combined positive score (CPS) ≥10 identified subgroups with superior and overall survival benefits from monotherapy or with compared to chemotherapy alone. TMB also overlaps significantly with (MSI), with approximately 80-100% of MSI-high cases in exhibiting high TMB (≥10 mutations per megabase), allowing MSI status to serve as a complementary indicator for immunotherapy eligibility in TMB-assessed tumors. Neoantigen prediction tools further refine TMB's role by estimating immunogenic peptide loads from mutations; tools like pVAC-Seq and NetMHCpan-4.1 use whole- sequencing data to prioritize neoantigens based on TMB-derived variants, correlating higher predicted neoantigen burdens with improved responses to inhibitors in non-small cell and . Composite scores that incorporate TMB with immune-related metrics provide a more holistic assessment of tumor . The Tumor Immune Dysfunction and Exclusion () score, derived from transcriptomic signatures of T-cell dysfunction and exclusion, is often combined with TMB to predict resistance; for example, in subtypes, TIDE-integrated analyses with TMB and neoantigen load identified immune evasion patterns, with lower TIDE scores in high-TMB groups associating with better efficacy. Recent multi-omics panels, such as those leveraging on genomic and histopathological data, integrate TMB with tumor-infiltrating lymphocyte (TIL) density; in , a 20-gene TMB estimation model correlated with increased + TIL density (R=0.891 for neoantigen burden), enabling prognostic independent of traditional TMB cutoffs. These 2025-era approaches, including lasso-based models from TCGA cohorts, emphasize TMB's synergy with TIL metrics to forecast . The primary benefits of these integrations include reduced false negatives among low-TMB patients who may still respond to due to other favorable immune features. Clonal TMB variants, when combined with neoantigen predictions, enhance specificity (0.8-0.9) for identifying true responders while minimizing misclassification in heterogeneous tumors like urothelial cancer. Clinical examples from 2024 meta-analyses in head and neck demonstrate this, where high TMB predicted superior overall response rates ( 2.62) and survival ( 0.53) to inhibitors across 1,200 patients, with integrations potentially capturing low-TMB subsets via or overlays to broaden treatment access. As of October 2025, studies presented at ESMO highlighted AI-powered (e.g., Lunit AI) predicting outcomes in colorectal, , and cancers, complementing TMB by analyzing H&E slides for immune features. Professional frameworks endorse TMB within multi-biomarker panels rather than as a standalone metric. The European Society for Medical Oncology (ESMO) guidelines recommend reporting TMB alongside other genomic biomarkers like and deficiency in solid tumor assays, specifying validated thresholds (e.g., ≥10 mutations per megabase) and graphical visualizations to guide blockade decisions. This approach ensures TMB contributes to personalized strategies without over-reliance on isolated thresholds.

Ongoing Research and Innovations

Recent phase III clinical trials in 2025 are investigating tumor mutational burden (TMB)-guided inhibitors (ICIs) in rare tumors, such as the ongoing evaluation of in advanced miscellaneous rare solid tumors. Additionally, studies like the envafolimab monotherapy trial in high-TMB advanced solid tumors highlight TMB's role in selecting patients across diverse histologies, including rarer subtypes. Longitudinal TMB tracking via has emerged as a key focus for monitoring resistance, with 2025 research demonstrating that early on-treatment TMB dynamics in predict response and clonal evolution in head and neck . Innovations in are advancing the understanding of intratumor TMB heterogeneity, enabling detection of low-frequency somatic variants and resistant subclones that bulk sequencing misses. Complementing this, models are predicting TMB directly from hematoxylin and eosin-stained slides, achieving area under the curve accuracies of 0.910–0.934 in lung and colorectal cancers, which could streamline non-invasive assessments without genomic sequencing. Emerging research explores -tumor TMB interactions, where variants in genes like , FANCL, and MSH6 significantly elevate TMB levels across pan-cancer cohorts, potentially enhancing neoantigen load and ICI efficacy. The gut microbiome's influence on mutational burden is also under investigation, with microbial interactions inducing mismatch repair deficiency signatures that increase rates in colorectal tumors. From 2024 to 2025, efforts to standardize liquid biopsy for TMB have intensified through consortia like Friends of Cancer Research, developing guidelines for validation, bioinformatics pipelines, and orthogonal confirmation to enable reliable dynamic monitoring. Looking ahead, global consortia, such as the and Friends of Cancer Research, are prioritizing diverse population data to address ancestry-driven TMB variations, aiming to recalibrate thresholds for equitable . As of October 2025, ESMO research linked thymic to response, suggesting it as a complementary to TMB for patient selection.

References

  1. [1]
    Tumor Mutational Burden (TMB) as a Predictive Biomarker in Solid ...
    Tumor mutational burden (TMB) is broadly defined as the number of somatic mutations per megabase of interrogated genomic sequence.Missing: facts | Show results with:facts
  2. [2]
    REVIEW Tumour mutational burden: an overview for pathologists
    Tumour mutational burden (TMB) is a diagnostic biomarker that can stratify cancer patients for response to immune checkpoint inhibitor therapies.Missing: facts | Show results with:facts
  3. [3]
    Development and validation of blood tumor mutational burden ...
    Dec 7, 2022 · Tumor mutational burden (TMB), measured by exome or panel sequencing of tumor tissue or blood (bTMB), is a potential predictive biomarker ...Missing: facts | Show results with:facts<|control11|><|separator|>
  4. [4]
    Tumor mutational burden standardization initiatives - PMC - NIH
    1. TUMOR MUTATIONAL BURDEN AS A BIOMARKER OF RESPONSE TO IMMUNE CHECKPOINT INHIBITORS. Tumor mutational burden (TMB) is the total number of somatic mutations in ...Missing: facts | Show results with:facts
  5. [5]
    What is tumor mutation load (TML)? - Thermo Fisher Scientific
    Tumor mutation load (TML), also known as tumor mutation burden (TMB), is a way to identify and quantify the number of non-synonymous, somatic mutations in ...
  6. [6]
    Tumor Mutational Burden as a Predictive Biomarker for Response to ...
    Tumor mutational burden (TMB)—the number of somatic mutations per DNA megabase (Mb)—has emerged as a proxy for neoantigen burden that is an independent ...Cancer Immunotherapy · Tmb As A Predictive... · Table 1
  7. [7]
    Tumor Mutation Burden - an overview | ScienceDirect Topics
    Tumor mutation burden is defined as the total number of nonsynonymous mutations present in a tumor. Patients with high TMB tumors are expected to have more ...Missing: facts | Show results with:facts
  8. [8]
    Effect of prior therapy on tumor mutational burden in NSCLC - PMC
    As expected, current and former smokers had statistically higher TMB than never smokers (median 9 vs. 9 vs. 4 mut/Mb, P<0.001). The median TMB was 8 mut/Mb in ...
  9. [9]
    Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma
    Nov 19, 2014 · We conducted a study to determine whether the genetic landscape of a tumor affects the clinical benefit provided by CTLA-4 blocking agents.
  10. [10]
    TMB Harmonization Project - Friends of Cancer Research
    This standardization plays a crucial role in optimizing the use of TMB for identifying patients best suited to benefit from immunotherapy, enhancing ...
  11. [11]
    FDA approves pembrolizumab for adults and children with TMB-H ...
    Jun 17, 2020 · The Food and Drug Administration granted accelerated approval to pembrolizumab (KEYTRUDA, Merck & Co., Inc.) for the treatment of adult and pediatric patients
  12. [12]
    Mutational Landscape and Sensitivity to Immune Checkpoint Blockers
    It is somehow intuitive that the presence of high tumor mutational burden can increase the likelihood of neoantigens formation, and that the most mutated tumors ...Immune Escape And... · Dna Repair Deficiencies In... · Predicted Neoantigen Load...
  13. [13]
  14. [14]
    Tumor mutation burden predicts response and survival to immune ...
    Our study indicates that high TMB is associated with better PFS. Thus, TMB can be considered as a predictive marker of PFS of patients treated with ICIs in the ...
  15. [15]
    Predictive value of tumor mutational burden for immunotherapy in ...
    Feb 3, 2022 · Meta-analyses of high TMB versus low TMB as well as immunotherapy versus chemotherapy in patients with high/low TMB were conducted. Hazard ratio ...
  16. [16]
    Tumor Mutational Burden Predicting the Efficacy of Immune ...
    This meta-analysis revealed that TMB can be used as a potential predictive biomarker of colorectal cancer patients receiving ICI therapy.
  17. [17]
    Consolidative nivolumab versus observation in unresectable stage ...
    Sep 29, 2025 · In the phase III CheckMate 816 trial for resectable stage IB to IIIA NSCLC ... TMB had significantly longer PFS than those with a low TMB (NR vs.
  18. [18]
    Tumor Mutational Burden as an Independent Predictor of Response ...
    High tumor mutational burden (TMB) may be a response biomarker for PD-1/PD-L1 blockade in tumors such as melanoma and non–small cell lung cancer (NSCLC).
  19. [19]
    Microsatellite-Stable Tumors with High Mutational Burden Benefit ...
    Our data demonstrate that MS-stable/TMB-high tumors are more common than MSI-high cancers and may benefit from immunotherapy.
  20. [20]
    Tumor mutational burden and survival on immune checkpoint ...
    Feb 6, 2025 · This study used the TMB algorithm from the FDA-approved test supporting solid tumor CDx and composite mortality variable validated against the ...
  21. [21]
    Tumor mutational and indel burden: a systematic pan-cancer ... - PMC
    Tumor mutational burden (TMB) has been widely studied as a predictive biomarker of response to immune checkpoint inhibitors (ICIs).Missing: facts | Show results with:facts
  22. [22]
    High Tumor Mutational Burden Correlates with Longer Survival in ...
    Here we evaluated the relationship between TMB and overall survival in 1,415 immunotherapy-naïve patients with diverse advanced malignancies. TMB was studied ...Missing: untreated | Show results with:untreated
  23. [23]
    Identification of tumor mutation burden-associated molecular and ...
    Feb 23, 2023 · We explored the ability of gene mutations to predict high- versus low-TMB cancers in TCGA pan-cancer and in 14 individual cancer types. We ...
  24. [24]
    Tumor break load quantitates structural variant-associated genomic ...
    May 14, 2025 · Genomic instability is associated with worse overall survival and a higher chance of metastasis. However, it can also offer therapeutic ...
  25. [25]
    Tumor mutational burden is associated with poor outcomes in ...
    Patients with a higher TMB exhibited shorter overall survival. TMB was associated with grade, age, subtype and mutations affecting genomic structure. Moreover, ...
  26. [26]
    The prognostic role of tumor mutation burden on survival of breast ...
    Nov 17, 2022 · Meta-analysis revealed that the prognostic relevance of TMB in breast cancer is limited in scope. High TMB may be associated with longer survival only in ICIs- ...
  27. [27]
    Universal cutoff for tumor mutational burden in predicting the efficacy ...
    May 24, 2023 · Our study demonstrates that 10 mut/Mb is the optimal, universal cutoff for TMB-high that guides the clinical application of anti-PD-(L) 1 therapy for advanced ...<|control11|><|separator|>
  28. [28]
    Prevalence of High Tumor Mutational Burden and Association With ...
    Oct 1, 2020 · Median (interquartile range) TMB was 2.6 (1.7-6.1) mut/Mb, and 332 patients (12.8%) had TMB-H (≥10 mut/Mb). Prevalence of TMB-H was highest ...
  29. [29]
    Pan-cancer analysis of heterogeneity of tumor mutational burden ...
    Diseases with more mutagen exposure, such as lung cancer and melanoma, were more highly mutated (median TMB 5.33 and 14.08 mutations/Mb, respectively).
  30. [30]
    Tumour mutational burden as a biomarker in patients with mismatch ...
    Mar 31, 2023 · Median TMB was 49 mut/Mb [interquartile range (IQR): 35–69 mut/Mb; range: 8–251 mut/Mb]. CGP was performed on primary tumours in 98 (89.1 ...
  31. [31]
    Interpretation of somatic POLE mutations in endometrial carcinoma
    Tumours with hotspot POLE mutations and MSI had a high TMB (median TMB of 339.0 mut/Mb, > 100 mut/Mb in all four cases) and a high proportion of C>A and T>G ...
  32. [32]
    Molecular Characterization of Polyomavirus-Positive and Negative ...
    We used WES to corroborate that VN tumors exhibit high TMB, harbor COSMIC UV signatures, and have more mutations in TP53, RB1, and NOTCH1. By leveraging WTS, we ...
  33. [33]
    Tumor mutational burden for the prediction of PD-(L)1 blockade ...
    TMB has emerged as a predictor of response to immune checkpoint inhibitors (ICIs) in various cancer types, and several studies have shown that patients with ...
  34. [34]
    Analysis of 100000 human cancer genomes reveals the landscape ...
    Apr 19, 2017 · Tumor mutational burden. TMB was defined as the number of somatic, coding, base substitution, and indel mutations per megabase of genome ...<|control11|><|separator|>
  35. [35]
    Assessment of Tumor Mutational Burden in Pediatric Tumors by ...
    Jan 17, 2020 · Tumor mutational burden (TMB) is a quantitative measure of acquired somatic mutations in the cancer cell genome. Initial exploratory analyses of ...
  36. [36]
    IDH mutant high-grade gliomas - PMC
    TMB for IDH mutant glioma is < 20 mutations/MB with the vast majority have TMB < 2 mutations/MB compared to PMMRDIA TMB of < 30 mutations/MB and MMR-deficient ...
  37. [37]
    Comprehensive analysis of tumour mutational burden and its clinical ...
    Feb 25, 2021 · The tumorigenesis of prostate cancer involves genetic mutations. Tumour mutational burden (TMB) is an emerging biomarker for predicting the ...
  38. [38]
    Identification of tumor mutation burden-associated molecular and ...
    Feb 24, 2023 · The cancer types having less than 1% of high-TMB cancers included THYM, PRAD, KIRC, KIRP, LAML, OV, PCPG, and THCA. Since the high TMB is a ...
  39. [39]
    The landscape of genomic alterations across childhood cancers
    Feb 28, 2018 · Here we present a comprehensive analysis of genetic alterations in a pan-cancer cohort including 961 tumours from children, adolescents, and young adults.
  40. [40]
    High and low mutational burden tumors versus immunologically hot ...
    Dec 27, 2018 · In addition, prostate and pancreatic cancers are both cold tumors with low tumor mutation burden (TMB) and are consequently not responsive to ...
  41. [41]
    Comparative Effectiveness of Immune Checkpoint Inhibitors vs ...
    Mar 31, 2022 · ... cancer objective responses in 1 of 11 patients (9%) in the high TMB and 7 of 115 patients (6%) in the low TMB group. Notably, patients with ...
  42. [42]
    Tumour mutation burden and infiltrating immune cell subtypes ... - NIH
    May 29, 2024 · This study explores their relationship based on gene mutation and transcription data in The Cancer Genome Atlas (TCGA) database, and the effects ...
  43. [43]
    Optimizing the evaluation of gene-targeted panels for tumor ... - Nature
    Oct 26, 2021 · Though whole exome sequencing (WES) is the gold-standard for measuring tumor mutational burden (TMB), the development of gene-targeted ...
  44. [44]
    Implementing TMB measurement in clinical practice - NIH
    Jan 24, 2019 · Three gene panel tests (FoundationOne, FoundationOne CDx, and MSK-IMPACT) have documented good concordance with TMB assessment by WES using ...
  45. [45]
    Comparison of commonly used solid tumor targeted gene ...
    Mar 26, 2020 · These results suggest that TMB values, as derived from the gene panels examined, are analytically and prognostically equivalent.
  46. [46]
    Tumor Mutational Burden by Whole-Genome Sequencing in ... - NIH
    TMB is a prognostic indicator of survival in resected NSCLC from individuals who never smoked. In this setting of low mutation counts, TMB can be accurately ...
  47. [47]
    Introduction and impact of routine whole genome sequencing in the ...
    Jul 12, 2024 · Paired whole genome sequencing (WGS) in contrast allows comprehensive assessment of small variants, copy number and structural variants along ...
  48. [48]
    Tumor Mutational Burden | TMB NGS testing - Illumina
    Tumor mutational burden (TMB) is the number of somatic mutations within the coding region of a tumor genome. It correlates with response to immunotherapeutic ...Missing: rationale | Show results with:rationale
  49. [49]
    Long-read sequencing of an advanced cancer cohort resolves ...
    Nov 13, 2024 · We show the potential of long-read sequencing for resolving complex cancer-related structural variants, viral integrations, and extrachromosomal circular DNA.<|separator|>
  50. [50]
    Implementation of Circulating Tumor DNA (ctDNA) Testing in ...
    Jul 26, 2025 · Conclusion. Study underscores substantial increase in the adoption of liquid biopsy and the real-world utility of ctDNA-based NGS testing in ...
  51. [51]
    Inflation of tumor mutation burden by tumor-only sequencing in ...
    Mar 19, 2021 · The TMB values were calculated as number of nonsynonymous mutations per Mb of coding regions. Four criteria were applied to identify patient ...
  52. [52]
    About the GATK Best Practices
    Jun 25, 2024 · The GATK Best Practices provide step-by-step recommendations for performing variant discovery analysis in high-throughput sequencing (HTS) data.About The Gatk Best... · 2. Analysis Phases · Lots Of Workflows That...
  53. [53]
    Mutect2 – GATK
    To call mutations on a tumor sample, call in this mode using a PoN and germline resource. After FilterMutectCalls filtering, consider additional filtering by ...Missing: TMB VEP
  54. [54]
    A Bioinformatics Toolkit for Next-Generation Sequencing in Clinical ...
    Specifically, we explore the bioinformatics steps involved in this process, including the calling of genetic alterations, their annotation, and interpretation.
  55. [55]
    Supplementary Methods
    All variants are annotated using the. Ensembl variant effect prediction (VEP) tool (McLaren et al., 2016) which is one of the most widely used and continuously ...
  56. [56]
    TMBcalc: a computational pipeline for identifying pan-cancer Tumor ...
    Apr 4, 2024 · This paper presents a pan-cancer analysis of Tumor Mutational Burden (TMB). We developed a novel computational pipeline, TMBcalc, to calculate the TMB.
  57. [57]
    Review Tumour mutational burden as a biomarker for immunotherapy
    Tumor mutational burden (TMB) is an emerging biomarker for response to PD-1/PD-L1 inhibitors. Various strategies exists to assess it and present limitations ...
  58. [58]
    The impact of mutational clonality in predicting the response to ...
    Sep 15, 2023 · We assessed the influence on the response of non-synonymous mutations (tumor mutational burden or TMB), clonal and subclonal mutations, ...
  59. [59]
    Tumor mutational burden and purity adjustment before and after ...
    May 20, 2021 · In a pure specimen, a clonal mutation (one present in all tumor cells) such as a core driver or early passenger mutation will have a VAF of 0.5 ...
  60. [60]
    [PDF] PR_AMP_TMB_Testing_Guideli...
    The recommendations cover pre-analytical, analytical, and post-analytical factors of TMB analysis, emphasizing comprehensive methodological descriptions for ...Missing: reproducibility | Show results with:reproducibility
  61. [61]
    FDA Approval Summary: Pembrolizumab for the Treatment of Tumor ...
    TMB ≥10 mut/Mb represented the cut-off point identified in the non–KEYNOTE-158 “training” dataset and prespecified in the KEYNOTE-158 statistical analysis plan.
  62. [62]
    Predictive Impact of Tumor Mutational Burden on Real-World ...
    Jun 7, 2024 · ... melanoma, but evidence supports a more proportional effect of TMB in metastatic melanoma and other cancer types treated with PD-1 inhibitors.
  63. [63]
    Harmonization of tumor mutation burden testing with comprehensive ...
    Feb 2, 2024 · In particular, the FOCR TMB Harmonization Project initiated a program that helped highlight the limitations of some TS panels and improved the ...
  64. [64]
    phase II of the Friends of Cancer Research TMB Harmonization ...
    Oct 1, 2021 · Aligning tumor mutational burden (TMB) quantification across diagnostic platforms: phase II of the Friends of Cancer Research TMB Harmonization ...Missing: 2024 | Show results with:2024
  65. [65]
    TUMOR MUTATIONAL BURDEN-TMB - CAP
    This program is intended for laboratories using next-generation sequencing to determine tumor mutational burden. This program is appropriate for laboratories ...Missing: surveys | Show results with:surveys
  66. [66]
    Establishing guidelines to harmonize tumor mutational burden (TMB)
    Mar 26, 2020 · Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation ... 20% or less in lung cancer ...
  67. [67]
    Recommendations for Tumor Mutational Burden Assay Validation ...
    Jun 5, 2024 · Laboratories may use reference materials to supplement but not supplant clinical samples for TMB assay validation. 6, Testing, Laboratories may ...Missing: NIST | Show results with:NIST
  68. [68]
    Enhancing the quality of panel-based tumor mutation burden ...
    Jan 23, 2024 · This multicenter study elucidates the major technical factors as sources of variability in panel-based TMB assays and proposed comprehensive recommendations.
  69. [69]
    Tumor mutational burden assessment and standardized ...
    Feb 20, 2024 · Background. High tumor mutational burden (TMB) was reported to predict the efficacy of immune checkpoint inhibitors (ICIs).
  70. [70]
    Tumour mutational burden is overestimated by target cancer gene ...
    The overestimation is caused by positive selection for mutations in cancer genes and cannot be completely addressed by the removal of mutational hotspots. We ...Missing: pitfalls overcounting
  71. [71]
    The role of neoantigens and tumor mutational burden in cancer ...
    Sep 2, 2025 · Among various biomarkers, neoantigens and tumor mutational burden (TMB) have emerged as critical factors in tailoring personalized treatments.Missing: rationale | Show results with:rationale
  72. [72]
    Validity and utility of blood tumor mutational burden (bTMB) is ... - NIH
    Aug 10, 2023 · Detection of high bTMB was associated with ctDNA shed, and was rarely detected in samples with ctDNA fraction below 1%. Concordance analysis in ...Missing: underestimates | Show results with:underestimates
  73. [73]
    TMBquant: an explainable AI-powered caller advancing tumor ...
    Sep 8, 2025 · Accurate tumor mutation burden (TMB) quantification is critical for immunotherapy stratification, yet remains challenging due to variability ...
  74. [74]
  75. [75]
    Keytruda | European Medicines Agency (EMA)
    The European Medicines Agency decided that Keytruda's benefits are greater than its risks and it can be authorised for use in the EU.
  76. [76]
    Tumor mutational burden: why is it still a controversial agnostic ...
    Dec 23, 2024 · Tumor Mutational Burden (TMB) is one of the latest biomarkers that is being studied and considered as a promising agnostic immunotherapy biomarker.
  77. [77]
    Real-world trends in costs of next generation sequencing (NGS ...
    May 28, 2021 · Average allowed amounts for NGS tests that included TMB testing varied from $438 to $3700 per test. To gain a better perspective on evolution of ...
  78. [78]
    ESMO study on the availability and accessibility of biomolecular ...
    The most important barriers to multiple versus single-gene sequencing techniques are the reimbursement of the test (59% versus 24%), and the availability of a ...
  79. [79]
    Pembrolizumab in Patients with Advanced Miscellaneous Rare ...
    Apr 10, 2025 · Single-agent pembrolizumab showed modest efficacy and was well tolerated in patients with rare solid tumors (ClinicalTrials.gov Identifier: ...Study Design And Patients · Results · Pd-L1 Expression And Til...
  80. [80]
    Monotherapy of envafolimab in patients with high tumor mutational ...
    May 28, 2025 · Background: Tumor mutational burden (TMB) has emerged as a predictive biomarker of immune checkpoint blockade response in cancers.
  81. [81]
    Longitudinal liquid biopsy identifies an early predictive biomarker of ...
    Sep 1, 2025 · Higher TMB is associated with an increased likelihood of response, likely due to the greater availability of neoantigens that can be targeted by ...
  82. [82]
    Single-cell multi-omics in cancer immunotherapy: from tumor ...
    Aug 25, 2025 · Recent studies ... Unravelling intratumoral heterogeneity through High-Sensitivity Single-Cell mutational analysis and parallel RNA sequencing.
  83. [83]
    Artificial intelligence-based digital pathology using H&E-stained ...
    Aug 4, 2025 · AI-based digital pathology uses H&E slides to aid in immune biomarker detection and predict immunotherapy response, including cancer detection ...
  84. [84]
    Germline variants predictive of tumor mutational burden and ... - NIH
    Mar 19, 2021 · Our results suggest that germline variants can influence the molecular phenotypes of tumors and predict the response to immune checkpoint inhibitors.
  85. [85]
    Microbial interactions induce the mutational signature of mismatch ...
    The tumor mutation burden (TMB, 1.075–79.50/Mb) in our cohort was comparable to that observed in colorectal cancers from TCGA (Fig. S1A). We identified the most ...
  86. [86]
  87. [87]
    Bringing next-generation precision oncology to patients - PMC
    Mar 12, 2025 · Worldwide Innovative Network (WIN) Consortium in Personalized Cancer Medicine: Bringing next-generation precision oncology to patients.
  88. [88]
    Ancestry-driven recalibration of tumor mutational burden and ...
    Oct 10, 2022 · TMB-high was significantly associated with improved outcomes only in European ancestries and merits validation in non-European ancestry populations.