Clinical Trial Eligibility: How Biomarkers and Inclusion Criteria Shape Cancer Treatment

When you or a loved one is considering a cancer clinical trial, the first question isn’t always about the drug-it’s about whether you qualify. The answer lies in two powerful tools: biomarkers and inclusion criteria. These aren’t just medical jargon. They’re the gatekeepers deciding who gets access to cutting-edge treatments and who doesn’t. And in cancer care, getting past those gates can mean the difference between hope and helplessness.

What Are Biomarkers, Really?

Biomarkers are measurable signs in your body that tell doctors something about your cancer. They’re not guesses. They’re facts-like a fingerprint for your tumor. These can be genes, proteins, or even molecules in your blood or tissue. For example, a mutation in the BRCA1 gene, or high levels of the protein PD-L1, aren’t just data points. They’re signals that tell researchers: this patient might respond to this drug.

The FDA breaks biomarkers into seven types, but for clinical trials, three matter most: predictive, prognostic, and pharmacodynamic. Predictive biomarkers tell you if a drug will work. Prognostic ones tell you how aggressive the cancer is. Pharmacodynamic ones show if the drug is hitting its target inside your body.

In 2022, 92% of new cancer drug approvals by the FDA came with a biomarker requirement. That means if your tumor doesn’t have the right marker, you won’t be offered the drug-even if it’s working wonders for others. It sounds strict, but it’s not arbitrary. Trials using biomarkers to pick patients have a nearly 50% success rate in Phase 2, compared to just 27% for trials that don’t. That’s not luck. That’s precision.

Inclusion Criteria: The Rules of the Game

Inclusion criteria are the rules you must meet to join a trial. They’re not random. Every single one is there for a reason: safety, science, or both.

Common criteria include:

  • Stage of cancer (e.g., metastatic, not early-stage)
  • Previous treatments (e.g., must have tried at least two lines of chemo)
  • Organ function (e.g., kidneys and liver must work well enough to handle the drug)
  • Performance status (e.g., able to walk and care for yourself)
  • Age limits (sometimes, but not always)
  • Biological markers (e.g., HER2-positive, EGFR mutation, MSI-H)
The big shift? Biomarkers are now often the first filter. A trial might start by saying: “Only patients with a KRAS G12C mutation.” That’s it. No other cancer type qualifies. This sounds limiting-but it’s actually fairer. It stops people from taking drugs that won’t work for them, sparing them side effects and false hope.

Take the case of neratinib for HER2-mutated breast cancer. In unselected patients, response rates were around 12%. But when they only enrolled patients with the HER2 mutation, response rates jumped to 32%. That’s not a small win. That’s life-changing.

Why Biomarkers Are Changing Everything

Before biomarkers, trials were like casting a wide net. You’d enroll hundreds of people with the same cancer type-lung, breast, colon-and hope the drug worked for some. Most didn’t. Phase 2 failure rates were over 60%.

Now, trials are like laser-guided missiles. You find the patients whose tumors have the exact target the drug is built to hit. The results? Faster recruitment, fewer dropouts, higher success rates.

In 2022, 73% of new oncology trials used biomarkers for eligibility. That’s up from 41% in 2017. The market for biomarker testing in trials is projected to hit $42 billion by 2027. This isn’t a trend. It’s the new standard.

Why? Because it works. Companies save money. Patients get better outcomes. Regulators approve drugs faster. Everyone wins-except maybe the patients who don’t have the right biomarker. And that’s the catch.

A patient and doctor reviewing a colorful flowchart of clinical trial inclusion criteria in a clinic.

The Hidden Challenges: Testing, Timing, and Access

Biomarkers sound simple. Get a biopsy. Run a test. See if you qualify. But in reality, it’s messy.

First, the test itself takes time. Specialized biomarker tests-like NGS panels or liquid biopsies-can take 7 to 14 days. That’s two weeks you might not have if your cancer is moving fast. And not every hospital can run them. You might need to send your tissue to a lab across the country.

Second, not all biomarkers are created equal. Some are well-validated. Others are still experimental. The European Medicines Agency found that 68% of biomarkers used in early trials don’t have enough validation to be trusted for decisions. That means some patients get excluded-or included-based on shaky science.

Third, access isn’t equal. A biomarker like HLA-A*02:01 shows up in 50% of Europeans but only 20% of North Americans. That means a trial designed in the U.S. might fail to recruit enough patients in Europe-or vice versa. Global trials are getting harder, not easier.

And then there’s cost. Biomarker testing adds thousands of dollars to each patient’s screening. Many insurance plans don’t cover it unless it’s part of an approved treatment. So patients often pay out of pocket-or miss out entirely.

What You Can Do: Navigating the System

If you’re exploring a clinical trial, here’s how to move forward:

  1. Ask for biomarker testing early. Don’t wait for your oncologist to bring it up. Say: “Can you test my tumor for biomarkers like PD-L1, KRAS, EGFR, or MSI?”
  2. Know your options. Not all tests are the same. A tissue biopsy is the gold standard. Liquid biopsies (blood tests) are faster and less invasive-but not always accurate enough for eligibility. Ask which test will be used and why.
  3. Check trial databases. Use ClinicalTrials.gov. Filter by cancer type and biomarker. You’ll find trials that match your profile.
  4. Ask about centralized testing. Some trials send kits to your local hospital. Others require you to travel. Find out what’s involved before you commit.
  5. Get a second opinion. If you’re told you don’t qualify, ask if another center might. Biomarker testing protocols vary. What one lab calls “negative,” another might call “low positive.”
Many patients assume they’re out of options if they don’t match a trial’s criteria. But that’s not always true. New trials open every week. Biomarker panels are expanding. What’s not eligible today might be eligible in six months.

Patients at a crossroads with biomarker cards, one path glowing toward treatment, another blocked by access barriers.

The Future: More Than One Marker

The next wave isn’t single biomarkers. It’s multi-omic panels-combining DNA, RNA, protein, and even immune cell data into one profile. By 2025, 65% of new trials are expected to use these complex signatures.

AI is helping too. Companies are using machine learning to find hidden patterns in data that humans miss. One trial in 2023 used AI to identify a new biomarker in lung cancer that predicted response to immunotherapy with 89% accuracy-something no single gene had ever done.

And soon, trials may adjust eligibility on the fly. Imagine starting a trial, getting your biomarker results, and then having the protocol change based on what it finds. That’s called “dynamic eligibility.” It’s already being tested in early-phase trials.

The goal? No more one-size-fits-all. Every patient gets a trial built for their cancer-not the other way around.

Final Thought: It’s Not Just Science. It’s Justice.

Biomarkers and inclusion criteria aren’t just technical tools. They’re ethical ones. They promise better outcomes. But they also risk leaving behind people who can’t access testing-because of where they live, how much they earn, or what kind of hospital they go to.

The science is moving fast. The system? Not so much. That’s why patient advocates are pushing for standardized testing, insurance coverage, and global equity. Because no one should be denied a chance at a new treatment just because their hospital doesn’t have the right lab.

If you’re in a trial-or thinking about one-know your biomarkers. Ask questions. Push for answers. Your cancer is unique. Your treatment should be too.

What are the most common biomarkers used in cancer clinical trials?

The most common biomarkers in cancer trials include HER2 (breast cancer), EGFR and ALK (lung cancer), BRCA1/2 (ovarian and breast), KRAS and NRAS (colorectal), PD-L1 (many cancers), MSI-H/dMMR (colorectal and endometrial), and NTRK fusions (rare but targetable across cancer types). These are well-established, FDA-recognized markers used to match patients with targeted therapies or immunotherapies.

Can I still join a trial if my biomarker test is negative?

It depends. Some trials are strictly biomarker-positive only. Others have “basket” or “umbrella” designs that include biomarker-negative patients in control arms or different treatment groups. Always ask the trial team: “Is there a cohort for patients like me, even if my biomarker is negative?” Sometimes, you’re eligible for a different arm of the same trial.

How long does biomarker testing take, and can I speed it up?

Standard tissue testing takes 7-14 days. Liquid biopsies can be faster-sometimes under 7 days-but aren’t always reliable for eligibility. To speed things up, ask if the trial uses a centralized lab with expedited processing. Some trials pre-ship test kits to your doctor. Others partner with commercial labs like Foundation Medicine or Tempus that offer faster turnaround. Don’t wait-ask about timing before you get your biopsy.

Why do some trials reject patients even if they have the right biomarker?

Having the biomarker is just the first step. Trials also require specific cancer stage, prior treatments, organ function, and overall health. For example, a patient with an EGFR mutation might still be excluded if their liver enzymes are too high or if they’ve had a recent heart attack. Biomarkers are necessary-but not always sufficient. Every criterion is there to protect your safety and ensure the trial’s scientific validity.

Is biomarker testing covered by insurance?

In the U.S., Medicare and most private insurers cover FDA-approved biomarker tests when used to guide treatment. But for experimental or exploratory biomarkers used only in trials, coverage is inconsistent. Some trials cover the cost. Others don’t. Always ask the trial coordinator in writing: “Who pays for this test?” and get it in your consent form.