Every year, over 1,000 generic drugs hit U.S. shelves - and nearly all of them go through the same tightly controlled process: the Abbreviated New Drug Application, or ANDA. This isn’t just paperwork. It’s the backbone of affordable medicine in America. If you’ve ever picked up a generic version of a brand-name pill and wondered how it got approved, here’s exactly how it works - from start to finish.
What Is the ANDA Process?
The ANDA is the FDA’s official route for approving generic drugs. It was created by the Hatch-Waxman Act of 1984, which struck a balance: protect innovation for brand-name drug makers while letting generics enter the market faster and cheaper. Unlike brand-name drugs, which require full clinical trials to prove safety and effectiveness, generics don’t need to repeat those studies. Instead, they prove they’re the same as the original - in active ingredient, strength, dosage form, and how the body absorbs it.
The FDA’s Office of Generic Drugs (OGD), part of the Center for Drug Evaluation and Research (CDER), handles every ANDA. They don’t test the drug themselves. They review the data submitted by the manufacturer. The goal? Make sure the generic works just like the brand-name version - no more, no less.
Step 1: Choose the Reference Listed Drug (RLD)
Before anything else, the generic manufacturer picks an approved brand-name drug as their target. This is called the Reference Listed Drug, or RLD. It’s the gold standard. Every claim about equivalence ties back to it.
For example, if you’re making a generic version of Lipitor (atorvastatin), Lipitor is your RLD. The FDA’s Orange Book lists all approved drugs and their RLDs. You can’t pick just any drug - it has to be one that’s still on the market and not withdrawn for safety reasons.
This step also determines patent and exclusivity timelines. If the RLD is still under patent protection, the generic maker must either wait until it expires or file a Paragraph IV certification - a legal challenge saying the patent is invalid or won’t be infringed. That’s where things get complicated.
Step 2: Build the Drug and Prove It’s Equivalent
Once the RLD is chosen, the manufacturer develops the generic version. This isn’t as simple as copying the formula. The active ingredient must match exactly - same chemical structure, same salt form, same purity. But inactive ingredients? Those can be different. Fillers, dyes, coatings - they just can’t affect how the drug works.
Then comes the most critical part: bioequivalence. The generic must deliver the same amount of active ingredient into the bloodstream at the same rate as the RLD. This is proven through a clinical study - usually with 24 to 36 healthy volunteers. They take the generic and the brand-name drug on separate days, and blood samples are taken over hours to measure absorption.
The FDA requires the average concentration of the generic to fall within 80% to 125% of the brand-name drug’s levels. That’s the bioequivalence window. If it’s outside that range, the application gets rejected. For complex drugs like inhalers or topical creams, this gets even trickier - and the FDA has special guidance for those.
Step 3: Submit the ANDA Dossier in eCTD Format
The ANDA isn’t a single form. It’s a 5-module electronic dossier called eCTD - the electronic Common Technical Document. This format is required for all submissions since 2017. Each module has a specific purpose:
- Module 1: Administrative info - company details, application type, fees paid.
- Module 2: Summaries - overviews of quality, nonclinical, and clinical data.
- Module 3: Chemistry, Manufacturing, and Controls (CMC) - the most detailed section. Includes raw material specs, manufacturing steps, stability testing, and packaging details.
- Module 4: Nonclinical study reports - rarely needed for generics unless there’s a safety concern.
- Module 5: Clinical study reports - bioequivalence data and analysis.
The dossier must be flawless. One missing signature, one typo in a batch record, one unexplained deviation in stability data - and the FDA can refuse to file it. That’s not approval denial. It’s a “refusal to file,” meaning they won’t even start reviewing it until you fix it.
Step 4: FDA Filing Review (60 Days)
Within 60 days of submission, the FDA checks if the ANDA is complete. They don’t evaluate science yet. They check:
- Are all modules included?
- Is the fee paid?
- Is the format correct?
- Are there any obvious gaps?
If it passes, they issue a “filing notice” and the clock starts on the 10-month review period under GDUFA. If it fails, they send a Refusal to File letter. You get one shot to fix it and resubmit - but the clock doesn’t restart until they accept the new version.
Step 5: The 10-Month Review Clock
Once filed, the FDA has 10 months to review the ANDA. That’s the GDUFA goal. In 2023, they met it for 84% of applications. But it’s not a straight line. During review, the FDA may send:
- Information Requests (IRs): Requests for more data or clarification. You have 15 days to respond.
- Discipline Review Letters (DRLs): Issues flagged by specific teams - chemistry, bioequivalence, labeling, or manufacturing.
- Complete Response Letter (CRL): The big one. If the FDA finds major flaws, they issue a CRL. You’ll need to fix the problem and resubmit, restarting the clock.
According to FDA data, about 75% of ANDAs get approved on the first try. The other 25% get a CRL. Common reasons? Poor CMC documentation (32%), flawed bioequivalence studies (28%), manufacturing site issues (22%), and labeling mismatches (18%).
Step 6: Facility Inspection
Before approval, the FDA inspects every facility involved - where the drug is made, packaged, tested, or stored. This isn’t a formality. It’s mandatory.
Inspections follow cGMP (Current Good Manufacturing Practices) standards - the same rules brand-name drugs must follow. The FDA looks for cleanliness, equipment calibration, data integrity, and quality control systems. If they find serious violations - like falsified test results or uncontrolled contamination - the application is denied.
Some companies get lucky and avoid inspection if their facility has been inspected recently and passed. Others, especially new or foreign plants, get flagged every time. Foreign sites make up over 70% of generic manufacturing - and the FDA inspects them just as rigorously as U.S. plants.
Step 7: Approval or Denial
If everything checks out, the FDA issues an approval letter. The generic drug is now listed in the Orange Book with an “AB” rating - meaning it’s therapeutically equivalent to the brand-name drug. Pharmacists can substitute it without a doctor’s permission.
If denied, you get a CRL. It lists every deficiency. Some can be fixed quickly. Others - like a failed bioequivalence study - mean starting over. Many companies spend millions and years trying to get it right.
Why This Matters: Cost, Access, and Quality
Generics make up 90% of prescriptions in the U.S. but only 23% of drug spending. In 2023, they saved the system $373 billion. Without the ANDA process, most people couldn’t afford their meds.
But it’s not perfect. Some experts warn about quality gaps, especially with narrow therapeutic index drugs - like warfarin or levothyroxine - where tiny differences can cause harm. The FDA acknowledges this and monitors post-market reports closely. So far, the data shows generics are safe for nearly everyone.
And the system is evolving. Under GDUFA IV (2023-2027), the FDA aims to review 90% of ANDAs within 10 months and cut the median review time to 8 months. They’re also using AI to sort through documents faster and tackling complex generics like inhalers and injectables with new guidance.
Who’s Winning in the Generic Market?
It’s a crowded field. Teva leads with 15.2% of the U.S. market, followed by Viatris (12.7%) and Sandoz (10.3%). But over 100 companies compete - from giants to small startups. The real prize? First-to-file exclusivity. If you’re the first to challenge a patent and win, you get 180 days of no competition. That’s how some companies made over $1 billion off a single generic, like the first version of Humira.
What Happens After Approval?
Approval isn’t the end. The FDA keeps watching. They inspect facilities annually. They track adverse events. If a generic starts causing unexpected side effects, they can pull it. Manufacturers must report every batch issue, every complaint, every deviation.
And if a new brand-name drug comes out? The generic maker can update their label or submit a new ANDA. The system is designed to keep moving - keeping prices low and access high.
How long does it take to get a generic drug approved by the FDA?
The FDA aims to review a complete ANDA within 10 months under GDUFA guidelines. But the total timeline from start to approval is usually 3 to 4 years. That includes 6-9 months for bioequivalence studies, 3-6 months for manufacturing development, and 2-4 months to prepare the application. Delays happen if the FDA issues a Complete Response Letter or if facility inspections uncover problems.
Can a generic drug be different from the brand-name version?
Yes - but only in inactive ingredients like fillers, dyes, or coatings. The active ingredient must be identical in strength, amount, and how it’s delivered. The drug must also be bioequivalent - meaning it enters the bloodstream at the same rate and amount as the brand-name drug. The FDA requires this to ensure the generic works the same way.
Why do some generic drugs get rejected by the FDA?
The most common reasons are incomplete or flawed chemistry, manufacturing, and controls (CMC) data (32% of cases), problems with bioequivalence studies (28%), manufacturing site violations (22%), and labeling errors (18%). Even small mistakes - like inconsistent batch records or unvalidated testing methods - can lead to rejection. The FDA doesn’t tolerate data integrity issues.
Are generic drugs as safe as brand-name drugs?
Yes, for the vast majority of patients. The FDA requires generics to meet the same strict standards for quality, strength, and purity as brand-name drugs. Studies show generics perform the same in clinical use. However, for drugs with a narrow therapeutic index - like blood thinners or thyroid meds - small differences in absorption can matter. The FDA monitors these closely and has approved thousands of safe generics in this category.
What’s the difference between ANDA and NDA?
An NDA is for new brand-name drugs and requires full clinical trials to prove safety and effectiveness - often taking 10-15 years and costing over $2 billion. An ANDA is for generics and only requires proof of equivalence to an already-approved drug. It skips the large-scale trials, focusing instead on manufacturing quality and bioequivalence. The ANDA process typically takes 3-4 years and costs $1-5 million.
Can a generic drug be approved before the brand-name patent expires?
Yes - but it can’t be sold until the patent expires or the patent challenge succeeds. A generic company can file an ANDA with a Paragraph IV certification, claiming the patent is invalid or won’t be infringed. This triggers a 45-day window for the brand-name company to sue. If they do, approval is delayed for up to 30 months unless the court rules in favor of the generic. This is the core of the Hatch-Waxman Act’s patent dispute system.
What’s the Orange Book and why does it matter?
The Orange Book, officially called “Approved Drug Products with Therapeutic Equivalence Evaluations,” is the FDA’s official list of all approved drugs and their therapeutic equivalence ratings. It tells pharmacists which generics can be substituted for brand-name drugs. An “AB” rating means the generic is therapeutically equivalent. Without this list, pharmacists wouldn’t know if they could legally swap a generic for a brand.
What’s Next for Generic Drug Approval?
The FDA is pushing to reduce its ANDA backlog from over 1,200 in 2020 to under 300 by 2025. They’re also expanding their Complex Generic Drug Products Initiative - with new guidance for inhalers, nasal sprays, and injectables. By 2026, they expect to approve 10-15 biosimilars a year, using a process similar to ANDA but for biologic drugs.
Artificial intelligence is being tested to speed up document review. By 2025, the FDA hopes to cut administrative time by 25%. That means faster approvals - and more affordable drugs reaching patients sooner.
The system isn’t flawless. But for over 40 years, the ANDA process has delivered on its promise: safe, effective, and affordable medicine for millions. And as long as the FDA keeps refining it, that promise will keep holding.
val kendra
December 4, 2025 AT 07:49Jenny Rogers
December 4, 2025 AT 12:58