Excellent 4.6 out of 5
Lung Cancer

Tumor Mutational Burden Test - Lung Cancer Biomarker

Tumor mutational burden (TMB) testing measures how many genetic mutations are present in a tumor to help predict whether immunotherapy is likely to be effective. By guiding more informed treatment choices, it can help avoid ineffective therapies, unnecessary side effects, and delays in getting the right cancer treatment.

Start testing
Cancel anytime
HSA/FSA eligible
Results in a week
Physician reviewed

Every result is checked

·
CLIA-certified labs

Federal standard for testing

·
HIPAA compliant

Your data is 100% secure

Key Insights

  • Understand how this test reveals your tumor’s DNA “mutation load,” which can signal how your lung cancer may behave and respond to certain treatments.
  • Identify a quantitative biomarker (mutations per megabase) that helps explain why some lung cancers respond to immune-based therapies while others do not.
  • Learn how tobacco exposure, tumor biology, and DNA repair capacity can shape your tumor mutational burden and influence treatment discussions.
  • Use insights to guide personalized choices with your oncology team, including whether immunotherapy is likely to help and how it fits with targeted therapy or chemotherapy.
  • Track how your results change over time to monitor disease evolution or response when repeat testing is appropriate.
  • When appropriate, integrate this test with PD-L1, microsatellite instability, and driver mutation testing (e.g., EGFR, ALK) for a more complete view of lung cancer biology.

What Is a Tumor Mutational Burden Test?

The Tumor Mutational Burden test measures how many DNA changes are present in a cancer’s genome. It is reported as the number of somatic mutations per megabase of DNA (mut/Mb). In lung cancer, TMB is typically assessed on tumor tissue from a biopsy or surgical sample, and in some cases on blood using circulating tumor DNA (ctDNA). Most labs use next-generation sequencing panels to count mutations across hundreds of cancer-related genes and then extrapolate to a per-megabase figure. Results are compared to laboratory-validated reference ranges and may be categorized as low, intermediate, or high. Different assays can yield slightly different numbers, so the report usually specifies the method and any threshold used for interpretation.

Why this matters: TMB captures how “mutated” a tumor is, which can influence how visible it is to the immune system. A higher TMB often means more abnormal proteins, or neoantigens, that immune cells can recognize. In non-small cell lung cancer, this information provides objective data about tumor immunogenicity, complementing other biomarkers like PD-L1, driver mutations, and microsatellite instability. Understanding TMB can help uncover therapeutic opportunities, refine risk discussions, and illuminate how your cancer may respond as it evolves over time.

Why Is It Important to Test Your Tumor Mutational Burden?

Lung cancers accumulate DNA damage through tobacco smoke, air pollutants, and normal cell turnover. Some tumors carry a heavy mutational load, while others remain relatively quiet. Measuring TMB connects this genetic “noise level” to practical questions: Is the tumor likely to present enough neoantigens for immune checkpoint therapy to recognize? Does the biology suggest a stronger role for immunotherapy or a combination approach with chemotherapy? TMB can also flag when other biomarkers might dominate the treatment plan. For instance, tumors driven by certain actionable alterations often rely on targeted therapy, and they may show limited benefit from immunotherapy despite measurable TMB. In short, TMB helps place your cancer on the map of immune-oncology—where visibility to the immune system can matter.

Zooming out, routine use of TMB is about precision, not labels. It offers a measurable way to assess risk, match therapies more thoughtfully, and monitor how a tumor changes under pressure from treatment. Over time, your oncology team can compare TMB with imaging, symptoms, and other lab results to understand what is working, what is not, and what to adjust next. The goal is not to “pass” a test but to translate a DNA signal into smarter, more personalized decisions that support durability of response and long-term outcomes, though ongoing research continues to refine how best to apply TMB across clinical scenarios.

What Insights Will I Get From a Tumor Mutational Burden Test?

Your results are typically displayed as a numeric value in mutations per megabase, sometimes with a category such as low, intermediate, or high based on the lab’s validated cutoffs. “Normal” does not apply here because we are describing tumor DNA, not healthy tissue. Instead, interpretation focuses on how your number compares to thresholds linked to clinical outcomes in lung cancer. An “optimal” zone is not universal; it depends on the treatment question at hand and the assay used. A value slightly above or below a lab’s threshold may be meaningful only when viewed with your tumor type, PD-L1 status, driver mutations, prior therapy, and overall clinical picture.

If your TMB is relatively higher, that often suggests a more immunogenic tumor environment. Clinically, this can align with greater likelihood of benefit from immune checkpoint therapies in many patients with non-small cell lung cancer, as shown in multiple studies, although the effect size varies and definitions of “high” are not identical across tests. If your TMB is lower, it may point toward strategies where chemotherapy or targeted therapy lead, or where immunotherapy is used in combination rather than alone.

Context matters. TMB can be influenced by sample quality (small biopsies may undercount mutations), tumor purity (too few cancer cells in the sample), prior treatments that reshape the tumor genome, and assay differences between labs. Blood-based TMB offers a noninvasive option but may read lower if the tumor sheds little DNA into the bloodstream, and clonal hematopoiesis can complicate interpretation. TMB is also only one slice of tumor biology. PD-L1, microsatellite instability status, and actionable driver mutations work alongside TMB to guide the best next step. Some co-mutations (for example, in STK11 or KEAP1) can blunt immunotherapy benefit in lung cancer even when TMB is elevated, underscoring why integrated interpretation is essential.

The real power of a tumor mutational burden test is pattern recognition over time. A rising or falling TMB on repeat testing may reflect tumor evolution or treatment pressure, but those changes need careful clinical interpretation alongside imaging and symptoms. Abnormal results are not a diagnosis by themselves and do not guarantee response or resistance. When combined with your history and the rest of your biomarker profile, TMB can help your care team turn a complex genetic pattern into a clear plan—supporting preventive strategy where possible, early detection of treatment failure, and a personalized path through lung cancer therapy that matches the biology of your disease.

Superpower also tests for

See more diseases

Frequently Asked Questions About

What do Tumor mutational burden tests measure?

Tumor mutational burden (TMB) measures the number of somatic (tumor‑specific) mutations—primarily nonsynonymous single‑nucleotide variants and small insertions/deletions—in the tumor’s coding DNA, reported as mutations per megabase (mut/Mb) of sequenced DNA. It is calculated from next‑generation sequencing (whole‑exome or targeted panels) after filtering out germline variants and technical artefacts.

TMB is used as an indirect estimate of neoantigen load and can correlate with the likelihood of response to immune checkpoint inhibitors, but results depend on assay type, panel size, sequencing depth, tumor purity and bioinformatic filters; therefore TMB is a useful biomarker but not a definitive stand‑alone predictor.

How is your Tumor mutational burden sample collected?

For tissue-based TMB testing we use tumor material obtained during a diagnostic or surgical procedure (core-needle biopsy, excision or resection). The lab typically accepts formalin‑fixed paraffin‑embedded (FFPE) blocks or unstained slides; a pathologist confirms tumor content and the sample’s suitability, DNA is extracted and sequenced (targeted NGS panel or whole‑exome) and the number of somatic mutations is reported as mutations per megabase. Matched normal blood may be requested to help filter inherited variants.

When tissue is unavailable, a liquid biopsy option uses a standard blood draw to isolate plasma cell‑free DNA; circulating tumor DNA is sequenced to estimate blood‑based TMB (bTMB). Blood samples must be collected and handled to preserve cfDNA and contain sufficient tumor-derived material for an accurate measurement.

What can my Tumor mutational burden test results tell me about my cancer risk?

Tumor mutational burden (TMB) measures how many mutations are present in the DNA of the specific tumor sample tested (usually reported as mutations per megabase). A higher TMB can indicate that the tumor has created more abnormal proteins (neoantigens), which in some cancer types is associated with a greater chance that immune checkpoint therapies will work. TMB describes the biology of the tested tumor at the time of biopsy and can help guide treatment choices when combined with other clinical information.

TMB is not a standalone measure of your overall cancer “risk” or an inherited risk for developing cancer; it does not predict whether you will get cancer in the future. Results vary by laboratory method, tumor type, sample quality and prior treatments, and cutoffs for “high” TMB differ between tests. Your TMB result should be interpreted together with other biomarkers (e.g., PD‑L1, MSI), pathology and your oncologist’s clinical judgment to guide care decisions.

How accurate or reliable are Tumor mutational burden tests?

Tumor mutational burden (TMB) can be a useful indicator of how likely a cancer is to respond to immune checkpoint inhibitors in some tumor types, but it is not perfectly accurate or universally predictive. Measured TMB correlates with response in several cancers, yet sensitivity and specificity vary widely because biological factors (tumor heterogeneity, clonality, prior treatment) and technical factors (sample quality, tumor purity, sequencing platform, panel size, depth and bioinformatics pipeline) influence results.

Reliability improves when using validated, clinically certified assays and when TMB is interpreted together with other biomarkers and the clinical context, but standardized cutoffs and methods are still imperfect across laboratories. A low TMB does not entirely rule out benefit and a high TMB does not guarantee response, so results should guide—rather than dictate—treatment decisions and are best reviewed by oncology specialists.

How often should I test my Tumor mutational burden levels?

There’s no universal schedule for TMB testing — it’s most commonly measured on tumor tissue (or plasma/ctDNA when tissue isn’t available) at diagnosis or specifically before deciding on immune checkpoint inhibitor therapy. Repeat testing is usually done when clinical management changes: at disease progression or recurrence, before starting a new line of therapy, when a new biopsy is obtained, or if enrollment in a clinical trial requires reassessment.

Routine serial TMB testing without a change in clinical status is generally not recommended because results vary by assay and can be affected by tumor heterogeneity and sampling. Discuss timing, the type of assay (tissue vs. blood), and how results will influence treatment with your oncology team.

Are Tumor mutational burden test results diagnostic?

No — Tumor Mutational Burden (TMB) results are not a medical diagnosis. TMB measures the number and pattern of mutations in tumor DNA and can highlight genomic imbalance or potential tumor resilience, but it does not by itself identify a specific disease or determine prognosis.

TMB must be interpreted in the context of symptoms, medical history, imaging, and other laboratory or biomarker data by a qualified clinician who can integrate all findings to reach a diagnostic and treatment decision.

How can I improve my Tumor mutational burden levels after testing?

Tumor mutational burden (TMB) is a property of the tumor’s DNA and is not something a patient can change directly with lifestyle or supplements. It may change only if the tumor’s biology changes (for example after certain therapies) or if a different tumor sample is tested, so you shouldn’t expect a simple way to “raise” TMB after testing.

Practical next steps are to discuss results with your oncologist or a molecular tumor board: they can confirm whether additional genomic testing (MSI/dMMR, POLE, broader NGS, or repeat biopsy/liquid biopsy) is appropriate, consider therapies or clinical trials that use TMB or other biomarkers to guide treatment, and interpret whether combination approaches could make immunotherapy more effective for your case. Avoid unproven interventions marketed to change biomarkers, and follow individualized medical advice from your cancer care team.

How it works

1

Test your whole body

Get a comprehensive blood draw at one of our 3,000+ partner labs or from the comfort of your own home.

2

An Actionable Plan

Easy to understand results & a clear action plan with tailored recommendations on diet, lifestyle changes, supplements and pharmaceuticals.

3

A Connected Ecosystem

You can book additional diagnostics, buy curated supplements for 20% off & pharmaceuticals within your Superpower dashboard.

Superpower tests more than 
100+ biomarkers & common symptoms

Developed by world-class medical professionals

Supported by the world’s top longevity clinicians and MDs.

Dr Anant Vinjamoori

Superpower Chief Longevity Officer, Harvard MD & MBA

A smiling woman wearing a white coat and stethoscope poses for a portrait.

Dr Leigh Erin Connealy

Clinician & Founder of The Centre for New Medicine

Man in a black medical scrub top smiling at the camera.

Dr Abe Malkin

Founder & Medical Director of Concierge MD

Dr Robert Lufkin

UCLA Medical Professor, NYT Bestselling Author

membership

$17

/month
Billed annually at $199
A smartphone displays health app results, showing biomarker summary, superpower score, and biological age details.
A website displays a list of most ordered products including a ring, vitamin spray, and oil.
A smartphone displays health app results, showing biomarker summary, superpower score, and biological age details.A tablet screen shows a shopping website with three most ordered products: a ring, supplement, and skincare oil.
What could cost you $15,000 is $199

Superpower
Membership

Your membership includes one comprehensive blood draw each year, covering 100+ biomarkers in a single collection
One appointment, one draw for your annual panel.
100+ labs tested per year
A personalized plan that evolves with you
Get your biological age and track your health over a lifetime
$
17
/month
billed annually
Flexible payment options
Four credit card logos: HSA/FSA Eligible, American Express, Visa, and Mastercard.
Start testing
Cancel anytime
HSA/FSA eligible
Results in a week
Pricing may vary for members in New York and New Jersey **

Finally, healthcare that looks at the whole you