Patent protection provides the principal basis for pharmaceutical product lifecycle management. Drug companies typically secure patent protection for a drug candidate and its likely method of use during the research and development phase. These patents should capture all aspects of the product manufacture, analysis, and clinical performance. Although these early patents provide critical protection for innovative products, later-stage patents can protect novel uses or improvements not proposed or described in the original disclosures.
This article will provide insights on enhancing life cycle management through late-stage pharmaceutical patent protection to maximize drug product success. Strategies include maximizing drug patent opportunities, evaluating patent protection in the context of likely approval pathways for competitive products, understanding the limits of patent protection, and coordinating patent protection with the Hatch-Waxman Act and FDA regulatory exclusivities.
Brief Introduction to Pharmaceutical Patents
Patent protection is vital to the pharmaceutical industry due to the significant time and expense invested in pharmaceutical research and development and the ease of copying and reproducing drug products. Legal and regulatory protections that prevent others from manufacturing or selling the invention for several years offer drug companies an incentive to develop new products and a means of recouping their investments.
In general, patents fall into three categories:
- Utility Patents
- Plant Patents
- Design Patents
Pharmaceutical patents are utility patents. After being granted by the U.S. Patent and Trademark Office (USPTO), utility patents have a term that generally expires 20 years from the patent application filing date.
Under the umbrella of utility patents are chemical, electrical, and mechanical patents. Pharmaceutical patents are generally chemical utility patents. These patents can cover four aspects of pharmaceutical products:
- Drug substance: Covers the active ingredient in the drug product, such as fluoxetine hydrochloride, the active ingredient in Prozac.
- Method of use: Covers indications and conditions of use for an approved drug product, such as using *finasteride* to treat male pattern baldness (distinct from its original use to treat enlarged prostate).
- Formulation: Covers the drug product's finished dosage form (e.g., tablet, capsule, or solution) that contains a drug substance and one or more other ingredients, such as a controlled-release tablet formulation of bupropion for once-daily dosing.
- Process: Covers the procedure used to make the active ingredient or the drug product, such as a novel synthetic route to manufacture atorvastatin calcium with higher yield and fewer impurities.
Pharmaceutical Patent Eligibility Criteria
To obtain patent protection, a sponsor must show that the invention is novel, non-obvious, adequately described, and enabled.
Novel. Under 35 U.S.C. §102, to qualify as "novel," the material elements of the invention must not have been disclosed in any prior art – namely, previous technology, publication, or use (as described further below). For example, an attempt to secure a method-of-use patent for an antidepressant to treat insomnia might fail if the treatment of insomnia is a side effect of the use of the drug to treat depression. When the prior art use has the effect of treating insomnia, the proposed new use of the antidepressant for the purpose of treating insomnia would not qualify for a new method of use patent.
Non-Obvious. To evaluate non-obviousness, the U.S. courts and the PTO use a structured set of factors laid out by the Supreme Court in Graham v. John Deere Co., 383 U.S. 1 (1966) and further elaborated by the Federal Circuit. Under 35 U.S.C. § 103, a product is "non-obvious" if there is a significant difference between the prior art and the new invention such that the new invention would not have been obvious to a “Person of Ordinary Skill In The Art” (sometimes referred to as a POSITA).
In addition, to help guard against hindsight bias when evaluating what would have been obvious at the time of invention, the courts and PTO take several secondary factors into account when evaluating non-obviousness. These objective indicia of non-obviousness include:
- Commercial success of the invention;
- Long-felt but unsolved needs;
- Failure of others;
- Unexpected results;
- Copying by others;
- Licensing (as evidence that the industry recognized the patent’s value);
- Industry praise, skepticism overcome, etc.
In 2007, the Supreme Court’s decision in KSR Int’l Co. v. Teleflex Inc., 550 U.S. 398 (2007) held that obviousness is ultimately a question of law based on underlying factual determinations. Today, the Federal Circuit and PTO apply a more flexible, common-sense approach to obviousness. But the Graham factors remain the primary analytic framework, and objective indicia are still required to be considered where present.
Adequately Described. Under 35 U.S.C. § 112(a), for a product to be "adequately described," the inventor must describe the product with enough detail to show that they were in possession of the invention at the time of filing. Applicants cannot seek protection for claims that are broader than the supporting specification. If an applicant is focused on a particular attribute of an invention, the applicant must clearly indicate that attribute in the specification. Some patent claims are determined to cover equivalent compositions or methods to those explicitly described in the claims.
Enabled. Under 35 U.S.C. § 112(a), to be "enabled,” a product must be described with enough detail to enable a POSITA to make and use the claimed invention without undue experimentation. The enablement requirement is in place to ensure that inventions are communicated to any interested parties in a meaningful way.
The adequately described and enabled requirements are separate but related, often overlapping in practice. For example, very broad claims that go beyond what’s described may fail both requirements, but legally they are distinct.
The Critical Role of Prior Art
In general, prior art refers to any public disclosure of the invention. Disclosures to the public that are considered prior art can take many forms, including product brochures, published journal articles, public presentations, sales or public use of the product, published applications, or patents. Once a product is disclosed to the public, the U.S. Patent and Trademark Office (USPTO) can use that prior art to defeat the applicant’s assertions of novelty and non-obviousness that are required for patent eligibility. Under the 2011 Leahy-Smith America Invents Act (AIA), "A person shall be entitled to a patent unless . . . the claimed invention was patented, described in a printed publication, or in public use, on sale, or otherwise available to the public before the effective filing date of the claimed invention." 35 U.S.C. §102(a)(1).
The AIA gives inventors a one-year grace period for applications filed for U.S. patents. Under U.S.C. §102(b)(1), inventors must file a patent application within one year of the initial public disclosure. For a period of one year before the effective priority date, any public disclosure made by the inventors—or by someone who obtained the material directly or indirectly from the inventor(s)—will not be deemed as prior art by the USPTO. Independent disclosures by third parties do not meet the requirements for the one-year grace period. Note: the one-year grace period is not available in most jurisdictions outside the United States.
Despite efforts to prevent disclosures, prior art disclosures might occur in a variety of circumstances. For example, while Helsinn Healthcare S. A. was developing a palonosetron product to treat chemotherapy-induced nausea, it granted another company the right to distribute, market, and sell 0.25 mg palonosetron doses in the U.S. following approval. The agreements required that this company keep all proprietary information secret.
Two years later, in January 2003, Helsinn filed a patent application for the 0.25 mg dose of palonosetron. Across the next ten years, Helsinn filed four more patent applications claiming the January 2003 application date as the priority date.
In 2011, Teva Pharmaceuticals sought approval to market a generic version of the 0.25mg palonosetron product. Helsinn sued Teva for infringement of all five palonosetron patents. Teva argued that one of the patents was invalid under the AIA's on-sale bar because the 0.25 mg dose was sold more than one year before Helsinn filed its 2003 patent application.
Teva only challenged one of the five patents (the patent relating to the 0.25 mg palonosetron dose) for invalidity under the on-sale bar, because the other patents were not directly tied to the pre-2003 sale or public disclosure. The on-sale bar only applies to the patent claims that are directly related to an "offer for sale" or sale of the patented invention before the critical filing date (one year prior to filing).
Helsinn won in District Court, arguing that the product details were not disclosed in the terms of sale agreements. But Teva won on appeal, the Federal Circuit Court holding that the sale was a public disclosure regardless of whether the product details were in the sale agreements.
Helsinn appealed to the Supreme Court to determine whether the product's sale to a third party under an agreement to keep the invention confidential meets the requirements for the invention to be "on-sale." The Supreme Court, referring to a pre-AIA decision in Pfaff v. Wells Electronics, Inc., held that the on-sale bar applies as long as the product is (1) the subject of an offer of commercial sale and (2) ready for patent. Helsinn v. Teva 586 U.S. 139 S.Ct. 628 (2019).
To prevent a prior art bar, investigators should consider filing a patent application before any form of public disclosure, including presentations at conferences, journal publications, FDA approvals, product sales, or any form of public use. Provisional patent applications are available for sponsors seeking a less formal, faster, and less expensive route to obtaining a patent filing date. Once a patent is pending under a provisional application, the investigator can continue development while disclosing information where needed for a period of one year. Note that, to maintain protection, the drug maker must file a non-provisional patent application within the one-year period.
Maximizing Drug Patent Opportunities
Pharmaceutical innovators employ multiple approaches to extend the commercial life of their key drug products. In addition to patents obtained during initial product development, later-stage patents can be obtained for novel uses or improvements not proposed or described in the original disclosures. Opportunities to extend a product’s life cycle through the patent system depend on many factors, including the scope of the initial patent disclosures, the willingness and ability to conduct additional product development on new uses and formulations, and the potential benefit of combining the product with other components that enhance its effectiveness or patient convenience.
During the early phases of drug discovery, companies should obtain a base patent that covers the core molecule, the anticipated formulation, and/or the anticipated method of use for the product. From here, drug makers may use this core invention as the basis for pursuing secondary and tertiary patents, thereby extending the life of the core product.
Secondary patents generally represent new uses or new formulations of the original patented product. Tertiary patents might be available if the original product can be paired with other therapeutic products, diagnostic tools, or medical devices.
Various strategies are available for drug sponsors to create new patent opportunities from an existing product. Each of these strategies requires consideration of the cost of developing the change that creates the new patent opportunities, the cost of obtaining regulatory approval for the change, the perceived benefit of the change, and the likely effectiveness of any patent for extending the life cycle of the product.
In addition, to limit the ability of competitors to design around a patent, drug companies should consider pursuing patents on alternative products or methods that can achieve a same or similar result to the innovator product.
-
Drug Product Modifications
Modifications to the drug product may result in a patentable invention when the new product produces a measurable improvement to the existing prior art. Modifications to the drug product that impact critical quality attributes (identity, quantity, safety, purity, potency) may provide patentable clinical and commercial advantages over the original drug product.
Compounds made from a mixture of enantiomers may be reformulated as a single enantiomer. Single enantiomer products frequently show various improvements over mixtures, including an improved therapeutic index, reduced risk for complex drug interactions, and improved bioavailability. For example, although AstraZeneca's patent for Prilosec (omeprazole) was set to expire in 2002, the effect of this patent expiration was limited. Many years earlier, the company created a single enantiomer of omeprazole—esomeprazole—that improved the bioavailability and clinical efficacy of the original. Patented in 1993 and approved for clinical use in 2000, Nexium brought in around $3 billion for AstraZeneca in 2003.
Similarly, novel polymorphs of a drug (various crystalline structures) can exhibit improvements in bioavailability, solubility, and shelf life from the patented molecule or an alternate crystalline state. Polymorphs of patented molecules may qualify as patentable when sponsors can demonstrate measurable improvements in safety, clinical efficacy, ease of manufacture, or other relevant benefits.
-
New Uses and Treatment Indications
Patents can be obtained for new uses and treatment indications when the product's new use or indication is novel and non-obvious. Patentable new uses and treatment indications may include the use of a drug in new patient populations, the use to treat new disease characteristics, or discoveries regarding use limitations, contraindications, and preexisting conditions.
-
New Strength, Dose, or Delivery Methods
Companies can extend patent protection for a product using new formulations of the prior art that produce clinically superior results, including lessened side effects or improved clinical outcomes. Since improvement in patient compliance benefits patients and the public health, new formulations that improve ease of use, new administration routes, or reduced dosages may be patentable.
-
Manufacturing Process Improvements
A manufacturing process improvement may qualify as a patentable method of manufacture. Likewise, improvements to essential intermediate drugs or biologics that result in new structural entities or improved commercial efficiency may qualify as patentable. If the improved process results in a slight modification to the structure or purity of the product, there may be an additional product patent opportunity.
-
Analytical Procedure Improvements
Inventions that improve analytical procedures, including in-house analytical chemistry procedures, may be patentable when the modification results in improved commercial efficiency.
-
Pharmacodynamics and Pharmacokinetics Improvements
Novel compositions and methods that improve the pharmacodynamics or pharmacokinetics of a given drug product can also meet eligibility requirements for patent protection. Such improvements may include improvements to drug absorption, distribution, elimination, physiological response, or drug-receptor binding dynamics.
-
Combination Products
Combination products encompass therapeutic configurations that include a drug and a device, biological product and device, drug and biological product, or more complex configurations such as a drug, device, and biological product. These may be packaged as a single entity, co-packaged together, or distributed separately with coordinated labeling stating that both (or all) components are needed to achieve the intended therapeutic use. Combination products may be patentable when the integration or coordinated use of these components produces a new or enhanced clinical benefit or functional advantage.
-
Fixed-Dose Drug Combinations
Combinations of two or more active pharmaceutical ingredients formulated into a single product, known as fixed-dose combinations, may be eligible for new patent protection when the combination demonstrates clinical superiority over each component alone. These synergistic drug combinations are commonly used to treat complex conditions such as cancer, cardiovascular disease, and HIV. Fixed-dose drug combinations that improve patient compliance, increase therapeutic efficacy, or reduce adverse effects relative to the original products may qualify for patent protection.
Evaluating Patent Protection Strategies in the Context of Likely Approval Pathways for Competitive Products
In evaluating the strength of patent protection, drug sponsors should consider the demonstration required for follow-on products. Patents focused on attributes that must be demonstrated for the follow-on product provide the greatest enhancement of patent protection for an approved drug product. The evaluation of patent protection in the context of likely approval pathways will help to identify aspects of the product that create competitive vulnerabilities and to identify needs and opportunities for enhanced patent protection.
Abbreviated New Drug Applications (ANDAs)
If the follow-on product is approved through an Abbreviated New Drug Application (ANDA), it must demonstrate that it is the "same" drug and that the product is bioequivalent.
Applicants may submit ANDAs for approval of generic versions of listed drugs previously approved under the FDCA. The ANDA pathway allows the applicant to rely on previous FDA findings of safety and effectiveness for listed drugs rather than requiring the ANDA applicant to independently demonstrate the safety and efficacy of its proposed drug.
Section 505(j) of the FDCA generally requires that an ANDA applicant refer to the Reference Listed Drug (RLD) in the application. An RLD is an approved drug product that is compared with proposed generic versions to show bioequivalence. Here, the term "bioequivalence" describes pharmaceutical equivalent or pharmaceutical alternative products that display comparable bioavailability when studied under similar experimental conditions.
Note that the RLD is not always the same as a Reference Standard. The RLD is the marketed, approved drug that serves as the comparator for generic drugs, while the Reference Standard is used for laboratory purposes to ensure the quality and consistency of the drug during manufacturing and testing.
The ANDA applicant must also demonstrate that its product and the RLD are the same with respect to active ingredient(s), dosage form, route of administration, strength, previously approved conditions of use, and, with certain exceptions, labeling.
An assessment of patents that protect the drug itself (and everything considered the same drug, such as polymorphs) would be helpful. Patents covering the pharmacokinetic (PK) profile of the drug include specific features of its absorption, distribution, metabolism, and excretion (ADME). Similarly, patents covering the pharmacodynamic (PD) properties of the drug include specific characteristics, such as receptor binding or downstream biological effects, relate to the drug’s clinical activity. PK and PD are typically considered separately in both patent strategy and regulatory review. Both PK and PD patents can provide meaningful protection because the follow-on product would likely need to infringe the patents to satisfy the ANDA approval requirements.
505(b)(2) Applications
In contrast to an ANDA under Section 505(j), a 505(b)(2) application filed under Section 505(b)(2) of the FDCA permits the applicant to rely in part on data not developed by the applicant, such as published literature or the FDA’s prior findings for an approved drug, while also including new clinical or nonclinical studies to support changes from the original product. This pathway is often used for modified versions of approved drugs, such as new dosage forms, new routes of administration, or combination products.
A key distinction lies in the description of the reference drug used. “Listed drug” is the term used for 505(b)(2) NDAs to refer to the drug or drugs on which the application relies for a demonstration of safety and effectiveness for the proposed product. A similar term, “Reference Listed Drug” (“RLD”) is used in the context of ANDA applications to refer to the approved drug that is the basis for the safety and effectiveness determination for the proposed product. While all RLDs are Listed Drugs, not all Listed Drugs serve as RLDs.
The 505(b)(2) pathway enables brand manufacturers to develop improved or differentiated products (such as extended-release formulations, new indications, or alternative delivery systems) supported by incremental innovation and new intellectual property. From a lifecycle management perspective, 505(b)(2) products can provide a new period of regulatory exclusivity and serve as a bridge between the original brand product and potential generic competition, effectively prolonging the commercial life of the franchise.
Biosimilars
For biologics, patents that cover the molecule and any characteristics that must be demonstrated for a showing of biosimilarity would provide the greatest commercial protection.
In contrast to small molecule drugs, biological products are licensed by FDA through the approval of a Biologics License Application (BLA) under section 351(a) of the Public Health Service (PHS) Act. To show biosimilarity or interchangeability with a licensed biological reference product, applicants can use the 351(k) abbreviated licensure pathway.
Under section 351(i)(2) of the PHS Act, "biosimilarity" means that the biological product is significantly similar to the reference product, aside from slight differences in clinically inactive components, and that no clinically meaningful differences exist regarding product safety, purity, and potency.
Under section 351(k)(2) of the PHS Act, a 351(k) application must contain (among other things) information showing that the biological product is biosimilar to a reference product using data derived from analytical studies, animal studies, and a clinical study or studies, including an assessment of immunogenicity, pharmacokinetics, and pharmacodynamics (mechanism of action, dose-response relationships), unless FDA determines, in its discretion, that certain studies are unnecessary in a 351(k) application.
To demonstrate biosimilarity, FDA’s regulations and guidance require sponsors to perform extensive comparative functional and structural studies to determine the level of similarity between a biosimilar product and the reference product. Such a comparative analytical assessment may require evaluation of several factors, including:
- Target binding properties
- Present impurities
- Physiochemical aspects
- Manufacturing process
- Functional activities
- Expression mechanics
FDA deems a biological product "interchangeable" with a reference product when it concludes that the information included in an application or supplement is sufficient to show that the product is (1) biosimilar to the reference product and (2) is expected to produce comparable clinical results in any given patient.
Achieving an interchangeability designation is desirable because it permits the biosimilar to be substituted for the reference product at the pharmacy level without intervention from the prescribing healthcare provider, similar to generic substitution for small molecule drugs. The interchangeability standard is also more rigorous than biosimilarity alone. It requires additional data, often including switching studies, to demonstrate that alternating between the biosimilar and the reference product presents no greater risk in terms of safety or diminished efficacy.
For products requiring multiple administrations to a single patient, switching between the reference product and biological product during the treatment regimen must not decrease safety, purity, or potency.
Limits of Patent Protection for Pharmaceutical Products
Any of the following factors may limit the commercial effectiveness of a patent:
- Indications and conditions of use may be subject to potential label carve-outs by ANDA or 505(b)(2) applicants. Carve-outs permit ANDA and 505(b)(2) applicants to avoid patent certification and infringement of patents listed in the Orange Book
- Competitors may be able to "design around" a patent
- USPTO office procedures may render the patent invalid or unenforceable
- Litigation may determine a patent invalid, unenforceable, or not infringed
Drug sponsors should pursue opportunities to reduce the impact of these limitations. In addition to thorough patent prosecution and robust litigation strategies, companies should consider strategies to reduce the ability of follow-on competitors to avoid the patents.
For example, the Hatch-Waxman Act requires ANDA applicants to make a certification for each patent listed in the Orange Book for the reference listed drug (RLD). However, when the RLD has multiple indications and the generic applicant is not seeking approval for an indication or condition of use that is covered by an Orange Book-listed method-of-use patent, the applicant may bypass a Paragraph IV certification by submitting a Section VIII Statement instead. This allows the ANDA applicant to obtain approval to market the generic product for the non-patented use only, without addressing the patents covering other indications.
In the Section VIII Statement, the ANDA applicant must “carve out” the patented methods of use in its proposed generic labeling. The proposed carve-out label must not overlap with the protected Use Code listed in the Orange Book.
The ability of the ANDA applicant to use a label carve-out depends, in part, on the Use Code that the NDA holder submitted for the patent. A Use Code is a specific identifier (e.g., U-1276) that FDA assigns to a patent to describe the scope of the patent's method-of-use claim. The Use Code helps determine which uses of a drug are protected by a patent, and therefore, which uses an ANDA applicant can and cannot claim in their application.
The Use Code ultimately determines whether the ANDA applicant must file a patent certification or Section VIII statement. In this regard, drug makers should work to craft strategically useful Use Codes that comply with FDA regulations and guidance to limit any indications and conditions of use subject to potential label carve-outs. Nevertheless, once a product is approved with a carve-out, an ANDA product can be prescribed by physicians and filed by pharmacists for the “off-label” infringing use.
Limitation of Patent Protection Under the Hatch-Waxman Safe Harbor Provision
The Hatch-Waxman amendments included a "safe harbor" provision that protects the use of patented products from patent infringement liability as long as activities are reasonably required to develop and submit information to FDA. The safe harbor applies to drugs, devices, and the use of patented technology reasonably related to the development and submission of information to FDA.
35 U.S.C. § 271(e)(1) states, "it shall not be an act of infringement to make, use, offer to sell, or sell within the United States or import into the United States a patented invention solely for uses reasonably related to the development and submission of information under a Federal law which regulates the manufacture, use, or sale of drugs or veterinary biological products."
The scope of the safe harbor provision is continually expanding. For example, many courts use a "routine" versus "non-routine" litmus test to determine whether the safe harbor provision applies. But much debate still surrounds what activities are considered "routine" versus "non-routine."
In general, courts consider "routine" FDA submissions as not protected by the safe harbor. In 2011, a Court held that safe harbor does not protect acts performed to gather "information that may be routinely reported to FDA long after marketing approval has been obtained." Classen Immunotherapies, Inc. v. Biogen Idec, 659 F.3d 1057 (Fed. Cir. 2011).
Routine submissions include using a patented invention for post-approval, FDA-mandated, commercial-release, and quality control testing. However, in a 2012 ruling, the Federal Circuit held that safe harbor does apply to post-approval submissions required to maintain FDA approval, deeming these submissions "non-routine." Momenta Pharmaceuticals, Inc. v. Amphastar Pharmaceuticals, Inc., 686 F.3d 1348 (Fed. Cir. 2012).
Patent Term Restoration
Although utility patents have a term of 20 years from the application filing date, commercially useful patent life following product approval could be lost due to the time it takes to conduct pre-market safety and efficacy clinical trials. To remedy the potential loss of post-approval patent life, the Hatch-Waxman Act includes Patent Term Restoration (PTR) provisions to restore part of the patent life lost during pharmaceutical product development and approval.
Patent Term Restoration is available for small molecule, biologic, manufacturing, and method-of-use patents. A product is eligible for PTR when (1) it is the first commercial marketing or use of the active ingredient subject to the regulatory review period and (2) the patent has not been previously extended under this provision.
The duration of PTR is based on the regulatory review period and is calculated as half of the clinical trials period plus all the FDA review period. The extension is capped at a maximum of 5 years, and under no circumstance may the total patent term exceed 14 years from the date of FDA approval.
Coordinating Patent Protection with FDA Regulatory Exclusivities
In the context of these limitations, other strategies for strengthening product life cycle become important. In addition to new patent protection, drug companies should consider all available regulatory incentives that could complement and enhance existing patents and extend the commercial lifecycle.
New Chemical Entity (NCE) Exclusivity is a 5-year market exclusivity available for FDA-approved drug products for which the active moiety has not been approved previously. For five years, FDA may not accept an application for review and may not approve a marketing application for a drug that relies on the safety and effectiveness data for the product awarded NCE exclusivity. Although NCE exclusivity lasts 5 years, a 505(b)(2) application or ANDA may be submitted after 4 years if the 505(b)(2) application or ANDA contains a paragraph IV certification to a patent listed in the Orange Book for the approved drug product.
New Product Exclusivity is a three-year market exclusivity available for FDA-approved new indications or conditions of use for which a new clinical investigation was essential to the approval of the change. For a period of three years after the approval date, FDA will not approve any 505(b)(2) application or ANDA for the indications or conditions of use of the NDA approval, or an ANDA submitted pursuant to an approved petition under section 505(j)(2)(C) of the Federal Food, Drug, and Cosmetic Act that relies on the information supporting the approval of an original NDA.
Orphan Drug Exclusivity is a seven-year market exclusivity available for products intended for use in fewer than 200,000 U.S. patients per year or products that are unlikely to recover research and development costs. Drug companies can sometimes achieve more than one non-consecutive periods of exclusivity for multiple orphan indications approved under the same orphan designation.
Pediatric Exclusivity provides as additional six months of protection for products that have completed at least one clinical investigation in pediatric age groups where a drug is anticipated to be used. Under the provisions of section 505A of the Food, Drug, and Cosmetics Act (FDCA), the six-month period attaches to New Chemical Entity (NCE) Exclusivity, New Product Exclusivity, Orphan Drug Exclusivity, and the patent period and is included in the Orange Book's patent listing.
Qualified Infectious Disease Product (QIDP) Exclusivity is awarded to drug products intended to treat serious or life-threatening infections. QIDP exclusivity adds a five-year extension to any existing regulatory exclusivities for the product, including the timing of patent challenges
Under the Biologics Price Competition and Innovation Act (BPCIA), biological products filed under a 351(a) application may be eligible for 12-year Reference Product Exclusivity. This includes four years of protection from FDA- review of biosimilar applications and eight additional years of protection before the FDA can approve an application.
Biosimilar products that can be licensed as interchangeable with the reference product under a 351(k) application may be eligible for Interchangeability Exclusivity. The duration of interchangeability exclusivity varies between 12 to 42 months [42 USC §262(k)(6)], depending on the timing of commercial marketing and the status of any ongoing litigation.
The numerous regulatory requirements and limitations involved in patent and exclusivity regulations can be complex. Understanding the interaction between patent protection and Food and Drug Administration (FDA) regulations will help companies maximize the commercial life cycle for pharmaceutical and biotechnology products. Sponsors should act early to implement these approaches into drug development programs and understand how the various protective mechanisms will impact developmental, regulatory, and competitive strategies.
Ultimately, a proactive and integrated approach to patent protection enables companies not only to preserve return on investment but also to sustain innovation that benefits patients and strengthens long-term market leadership. The most effective life cycle management plans are those that anticipate competitive pathways, mitigate risks under provisions such as Hatch-Waxman, and maximize opportunities for patent protection. By aligning patent strategies with regulatory exclusivities and leveraging opportunities across product modifications, new indications, and process improvements, drug developers can meaningfully extend market presence and protect revenue streams well beyond initial approval.