The FDA doesn’t rate biosimilars like products on a scale from 1 to 10. There’s no A, B, or C grade. Instead, it uses a strict, science-driven process to decide if a biosimilar is close enough to its reference biologic to be approved for use. This isn’t like approving a generic pill. Biosimilars are made from living cells - think proteins, antibodies, or complex molecules - and even tiny changes in how they’re made can affect how they work in the body. That’s why the FDA’s evaluation is far more detailed than what’s needed for traditional generics.
How the FDA Determines Biosimilarity
The process starts with science, not stories. Manufacturers must prove their biosimilar is highly similar to the original biologic - known as the reference product - with no clinically meaningful differences in safety, purity, or potency. That means the biosimilar must behave the same way in the body, even if it’s not an exact chemical copy. The FDA looks at over 200 to 300 specific characteristics of the molecule, called critical quality attributes. These include things like protein structure, sugar attachments (glycosylation), and how the molecule folds. Each one is measured using advanced tools like mass spectrometry and capillary electrophoresis. The goal? To show 95% to 99% similarity across these attributes.
It’s not enough to just match the molecule. The biosimilar must also work the same way. That’s where pharmacokinetic (PK) and pharmacodynamic (PD) studies come in. These tests track how the drug moves through the body and how it affects biological systems - like how fast it’s absorbed, how long it lasts, and how strongly it binds to its target. These studies are usually done in healthy volunteers or patients, with 50 to 100 people per group. Immunogenicity is another big piece: does the body react to the drug as if it’s foreign? Even a small immune response can lead to reduced effectiveness or serious side effects. So, the FDA requires long-term monitoring, often up to a year, to catch any unexpected immune reactions.
Why Biosimilars Aren’t Generics
Many people think biosimilars are just like generic versions of brand-name drugs. They’re not. A generic version of, say, ibuprofen is chemically identical to the brand version. You can break it down into the same molecules, and it’s made with the same ingredients. Biosimilars? They’re made by living cells - yeast, bacteria, or mammalian cells - and even small changes in the cell line, fermentation process, or purification steps can alter the final product. That’s why you can’t just copy the formula. You have to rebuild it from the ground up and prove it works the same way.
The FDA’s approval pathway for biosimilars is called 351(k), created by the Biologics Price Competition and Innovation Act (BPCIA) in 2010. It’s abbreviated, meaning manufacturers don’t have to run massive clinical trials from scratch. But that doesn’t mean it’s easier. In fact, the analytical work alone can cost $120 million to $180 million and take 10 to 12 months. Compare that to a generic drug, which might cost $1 million to develop and take less than a year. The complexity is why only 43 biosimilars have been approved by the FDA as of October 2025, even though over 100 have been approved in Europe.
The FDA Purple Book: Your Official Biosimilar List
If you want to know which biosimilars are approved and what they’re approved for, the FDA’s Purple Book is the only official source. It’s updated daily now, since early 2025, and it’s searchable online. Before, it came out monthly. Now, you can see every approved biosimilar, its reference product, the date it was approved, and whether it’s designated as “interchangeable.”
Interchangeable is the highest level. It means the FDA has determined you can switch between the biosimilar and the reference product as many times as you want, and it won’t change how safe or effective the treatment is. As of October 2025, only 17 of the 43 approved biosimilars have this status. To get it, manufacturers must prove that switching back and forth doesn’t increase risk or reduce effectiveness - often requiring additional clinical studies beyond what’s needed for basic biosimilarity.
For example, the biosimilar for adalimumab (Humira) approved in 2023 became interchangeable in 2024 after demonstrating that patients switching between the original and the biosimilar had no increase in side effects or loss of response over six months. That’s a big deal - it gives pharmacists the legal right to substitute the biosimilar without asking the doctor, just like they do with generics.
What’s Changed in the Last Two Years
The FDA has made major updates to its guidance to speed things up without lowering standards. In September 2024, the agency said manufacturers no longer need to run full comparative efficacy studies if their analytical data is strong enough. That’s a game-changer. Before, you had to run a full clinical trial comparing the biosimilar to the reference product in patients. Now, if the molecule is well understood and the analytical data shows near-perfect similarity, the FDA may accept that as enough. This has cut development time by 12 to 18 months and saved companies $50 million to $100 million per product.
Another big change: the FDA removed the requirement for forced degradation studies - where scientists stress the drug with heat, light, or chemicals to see how it breaks down. These tests were once thought to predict real-world stability, but they didn’t actually correlate with how the drug performed in patients. Removing them saved time and money without affecting safety.
And now, for certain well-characterized proteins, the FDA allows “extrapolation.” That means if a biosimilar is approved for one condition - say, rheumatoid arthritis - and the science shows it behaves the same way in the body, it can be approved for other conditions the reference product treats, like Crohn’s disease or psoriasis - without running new trials in those patient groups. This is already being used for biosimilars of rituximab and trastuzumab in oncology.
Why Adoption Is Still Slow
Even with 43 approved biosimilars, only 29 have actually hit the market. Why? Patent lawsuits. Biologic makers often file lawsuits to delay biosimilar entry, and these can drag on for years. On average, it takes 11.3 months from FDA approval to market launch - and sometimes much longer.
Payers - insurance companies and pharmacy benefit managers - also play a role. Even when a biosimilar is cheaper, some insurers still favor the original drug because of long-standing contracts or because prescribers are hesitant to switch. In autoimmune diseases like rheumatoid arthritis, biosimilars for adalimumab only captured 28% of the market by mid-2025, far below the 50% the FDA expected. But in oncology, biosimilars for drugs like rituximab and trastuzumab have taken over 65% to 75% of the market within 18 months of launch. Why? Because cancer treatments are expensive, and hospitals and clinics are more motivated to cut costs.
Real-world data backs up the safety. The FDA’s Sentinel Initiative tracks adverse events across millions of patients. As of Q3 2025, biosimilars had 0.8 adverse events per 10,000 patients. The reference products? 0.7. That’s not a meaningful difference. No biosimilar has shown a unique safety signal in over nine years of post-market monitoring.
What’s Next for Biosimilars
The FDA’s 2025-2027 roadmap is focused on three things: complex molecules, AI, and interchangeability. Right now, only three applications for biosimilars of antibody-drug conjugates (a type of targeted cancer therapy) have been submitted - none approved. These are far more complex than standard monoclonal antibodies. The FDA plans to release specific guidance for them by Q3 2026.
They’re also testing AI tools to analyze the massive amounts of data from analytical studies. Instead of human reviewers looking at thousands of data points, machine learning models will flag anomalies faster and more accurately. A pilot program starts in Q1 2026.
And for combination products - like a biosimilar that includes a delivery device - the FDA is working on a formal interchangeability framework by Q2 2027. This is critical because many biologics are now delivered through auto-injectors or pumps. If the biosimilar doesn’t work the same way with the device, it’s not interchangeable.
The goal? To get more biosimilars to market faster and save patients and the system billions. The FDA now projects $250 billion in cumulative savings by 2030, up from $150 billion in their 2018 estimate. But the real win? More patients getting access to life-saving treatments at a lower cost - without sacrificing safety.
Are biosimilars the same as generic drugs?
No. Generic drugs are chemically identical copies of small-molecule drugs, like aspirin or metformin. Biosimilars are copies of large, complex biologic drugs made from living cells. Even tiny changes in the manufacturing process can alter their structure and function, so biosimilars must be proven to be "highly similar" with no clinically meaningful differences - not identical.
What does "interchangeable" mean for a biosimilar?
An interchangeable biosimilar has been proven to produce the same clinical result as the reference product in any patient, and switching between them won’t increase safety risks. Pharmacists can substitute it for the original drug without needing the prescriber’s permission - just like with generics. Only 17 of the 43 FDA-approved biosimilars have this status as of October 2025.
How long does it take to get a biosimilar approved by the FDA?
On average, it takes 3.2 years from the first FDA meeting to approval - longer than in Europe, where it’s about 2.1 years. The U.S. process is more rigorous, especially in analytical testing. But recent guidance changes have cut development time by 12 to 18 months for many products.
Why are so few biosimilars on the market if 43 are approved?
Patent litigation is the biggest barrier. Biologic manufacturers often sue to delay biosimilar entry, adding years to the timeline. On average, it takes 11.3 months from FDA approval to market launch, and some take much longer. Payer restrictions and prescriber hesitation also slow adoption, especially in autoimmune diseases.
Are biosimilars safe?
Yes. Since the first FDA-approved biosimilar in 2015, there have been no biosimilar-specific safety signals identified in over nine years of monitoring. Adverse event rates are statistically equivalent to the reference products, according to the FDA’s Sentinel Initiative data as of Q3 2025.
Can a biosimilar be approved for more uses than the reference product?
No. But the FDA allows "extrapolation" - approving a biosimilar for additional indications of the reference product without new clinical trials, if the science supports it. For example, if a biosimilar is proven effective for rheumatoid arthritis and the mechanism is the same for psoriasis, it can be approved for both without testing in psoriasis patients. This is common for well-characterized proteins.