How Bioequivalence Studies Are Conducted: Step-by-Step Process

How Bioequivalence Studies Are Conducted: Step-by-Step Process

Nov, 17 2025

Written by : Zachary Kent

When a generic drug hits the pharmacy shelf, you might think it’s just a cheaper version of the brand-name pill. But behind that simple swap is a rigorous, science-driven process called a bioequivalence study. These studies don’t guess whether the generic works-they prove it. And they’re the reason millions of people safely switch from expensive brand drugs to generics every year.

Why Bioequivalence Studies Matter

Bioequivalence studies answer one critical question: Does the generic drug deliver the same amount of active ingredient into the bloodstream at the same speed as the brand-name version? If the answer is yes, then the generic is considered therapeutically equivalent. That means it works the same way in your body, with the same benefits and risks.

This isn’t just about saving money. The U.S. FDA estimates that generic drugs saved the American healthcare system over $1.68 trillion between 2010 and 2019. Without bioequivalence studies, those savings wouldn’t be possible. Regulators like the FDA, EMA, and Japan’s PMDA require them before approving any generic drug that’s meant to be absorbed into the bloodstream.

But here’s the catch: Not all drugs are the same. Some are easy to match. Others-like those with narrow therapeutic windows or complex delivery systems-need extra scrutiny. That’s why the process isn’t one-size-fits-all.

The Standard Study Design: Crossover Trials

Most bioequivalence studies use a two-period, two-sequence crossover design. That sounds complicated, but it’s simple in practice.

Imagine 24 to 32 healthy volunteers. Half get the generic drug first, then after a break, the brand-name drug. The other half get them in reverse order. This design controls for individual differences-since each person serves as their own control, you’re comparing apples to apples.

The washout period between doses is crucial. It must be long enough for the first drug to completely leave the body-usually at least five half-lives. For a drug that takes 12 hours to clear, that’s 60 hours. For one with a 72-hour half-life? That’s six days. Get this wrong, and leftover drug from the first dose skews the results. One contract research organization reported a $250,000 delay because they underestimated the washout time for a long-acting drug.

How Blood Samples Are Collected

After each dose, participants give blood samples at specific times. These aren’t random. They’re carefully timed to capture how the drug moves through the body.

Typical sampling points include:

  • Before dosing (baseline)
  • Just before the expected peak concentration (Cmax)
  • At and around the peak
  • During the elimination phase

The goal is to capture the full concentration-time curve. Sampling continues until the area under the curve (AUC) reaches at least 80% of the total possible exposure (AUC∞). For most drugs, this means collecting samples for 3 to 5 half-lives-sometimes up to 48 hours or more.

Plasma or serum is used, not whole blood. The samples are analyzed using highly precise methods-usually liquid chromatography with mass spectrometry (LC-MS/MS). These methods must be validated to ensure accuracy within ±15% (±20% at the lowest detectable level). If the lab’s method isn’t good enough, the whole study can be rejected.

Key Metrics: Cmax and AUC

Two numbers tell the whole story: Cmax and AUC.

  • Cmax is the highest concentration of the drug in the blood. It tells you how fast the drug gets absorbed.
  • AUC(0-t) is the total drug exposure over time, from dosing until the last measurable concentration. AUC(0-∞) includes estimated total exposure beyond the last sample.

Both values are log-transformed before analysis. Then, statisticians run an ANOVA model with effects for sequence, period, treatment, and subject. The result? A 90% confidence interval for the geometric mean ratio of the generic to the brand drug.

The rule is simple: That interval must fall between 80.00% and 125.00% for both Cmax and AUC. If it does, the drugs are bioequivalent. For drugs with a narrow therapeutic index-like warfarin or lithium-the window tightens to 90.00%-111.11%.

Scientists analyzing drug concentrations in plasma using LC-MS/MS equipment with dissolution test beaker and data readouts.

What Happens When the Numbers Don’t Fit?

Not every study passes. About 5-10% fail on the first try. Common reasons include:

  • Washout periods too short
  • Sampling schedule missing key time points
  • Statistical errors in analysis
  • Assay validation issues

According to the FDA, 45% of deficient studies fail due to inadequate washout, 30% because of poor sampling, and 25% from statistical mistakes. Bioanalytical method failures alone cost an average of $187,000 per delay, according to BioAgilytix’s 2023 white paper.

One example: Alembic Pharmaceuticals tried to get a generic version of Trulicity (dulaglutide) approved in 2022. Their Cmax values varied too much across studies. The FDA rejected it. The drug’s complex structure made absorption unpredictable-something a pilot study might have caught early.

When the Standard Doesn’t Work

Not all drugs can be studied with a crossover design. Some have half-lives longer than two weeks. Others can’t be safely given multiple times. For those, alternatives exist.

  • Parallel studies: Two separate groups-one gets the generic, the other the brand. Used for very long-acting drugs.
  • Replicate designs: For highly variable drugs (CV >30%), a four-period design is used. This gives more data on within-subject variability. The EMA requires this; the FDA allows it under reference-scaled bioequivalence.
  • Multiple-dose studies: For extended-release formulations, the study runs for several days to mimic real-world use.
  • Pharmacodynamic or clinical endpoint studies: For topical creams, inhalers, or injectables, you measure the drug’s effect-like skin redness or lung function-instead of blood levels.

The FDA’s 2020 guidance requires clinical endpoint data for certain dermatological products because skin absorption can vary wildly based on formulation-even if blood levels look the same.

Dissolution Testing: The First Line of Defense

Before any human study, companies run dissolution tests. They put the generic and brand tablets in fluids that mimic stomach and intestine pH (1.2 to 6.8). Then they measure how quickly the drug dissolves.

The test must show similarity across at least 12 units per condition. The similarity factor (f2) must be above 50. This isn’t just paperwork-it’s a predictor. If two drugs don’t dissolve the same way, they’re unlikely to behave the same in the body.

For some simple drugs (BCS Class I-highly soluble and highly permeable), dissolution testing alone can replace human studies. About 27% of 2022 FDA approvals used this “biowaiver” approach. It saves time, money, and volunteers.

Side-by-side comparison of generic and brand drug formulations with dissolution profiles and bioequivalence confidence interval.

What Makes a Study Successful?

Based on data from 127 contract research organizations, the top success factors are:

  • Running a pilot study first (used by 89% of successful studies)
  • Real-time analysis of blood samples (cuts protocol deviations by 40%)
  • Using the same batch of brand drug across all study arms
  • Ensuring the generic batch is from commercial-scale production (at least 100,000 units or 1/10 of full scale)

Dr. Jennifer Bright, former director of the FDA’s Office of Generic Drugs, said pilot studies reduce failure rates from 35% to under 10%. That’s huge when each study costs $1-3 million.

The Bigger Picture: Regulation and Innovation

The global bioequivalence market was worth $1.87 billion in 2022 and is growing fast. Why? Because patents are expiring on $66 billion worth of branded drugs between 2023 and 2025.

Regulators are adapting. The FDA is exploring real-world evidence and modeling to reduce study needs. The EMA and Japan are refining their rules for complex drugs. But the core hasn’t changed: prove equivalence, prove safety, prove consistency.

And it works. In 2022, the FDA approved 936 generic drugs based on bioequivalence data-98% of all generic approvals that year. No safety signals were found in a 2023 FDA meta-analysis of 1,200 approved generics.

It’s not perfect. Some experts warn that relying only on blood levels might miss differences in how a drug acts locally-like in the lungs or skin. But for systemic drugs, the current system is the most reliable tool we have.

What You Can Trust

Next time you pick up a generic pill, know this: It didn’t just get approved because it looks the same. It went through a scientifically strict, data-heavy process designed to match it exactly to the original. The numbers don’t lie. And the savings? They’re real.

What are the two main pharmacokinetic parameters measured in bioequivalence studies?

The two main parameters are Cmax (maximum plasma concentration) and AUC (area under the concentration-time curve). Cmax tells you how quickly the drug is absorbed, while AUC tells you how much of the drug is absorbed overall. Both must fall within 80-125% of the brand-name drug’s values for the generic to be approved.

Why do bioequivalence studies use healthy volunteers instead of patients?

Healthy volunteers eliminate variables like disease states, other medications, or organ dysfunction that could affect drug absorption or metabolism. This ensures any differences seen are due to the drug product itself-not the patient’s condition. Once bioequivalence is proven, it’s assumed to hold true across patient populations.

Can a generic drug be approved without a human bioequivalence study?

Yes, under certain conditions. For drugs classified as BCS Class I-highly soluble and highly permeable-regulators like the FDA may approve a generic based on dissolution testing alone, known as a biowaiver. This applies to about 27% of approvals. But for complex formulations, injectables, or drugs with narrow therapeutic windows, human studies are mandatory.

What happens if a bioequivalence study fails?

If the 90% confidence interval for Cmax or AUC falls outside 80-125%, the study fails. The sponsor must investigate why-was it the formulation, the sampling, the analysis? They often run a pilot study, tweak the formula, and resubmit. Failure rates are around 5-10% for first attempts. Repeated failures can delay market entry by months or years, costing millions.

How long does a typical bioequivalence study take?

A single-period crossover study usually takes 3-6 weeks per subject, including screening, dosing, and follow-up. With 24-32 subjects, the full study runs 4-8 months. Including protocol development, lab validation, and regulatory submission, the entire process from start to approval typically takes 18-24 months. For complex drugs, it can take longer.

What to Watch For in the Future

The field is moving toward more efficient methods. Modeling and simulation (like PBPK models) are growing fast. Real-world data and artificial intelligence may soon help predict bioequivalence without full trials. But for now, the gold standard remains: carefully designed studies, precise measurements, and strict statistical rules.

It’s a system built on science, not guesswork. And that’s why you can trust your generic prescription.

© 2025. All rights reserved.