2,581
Views
16
CrossRef citations to date
0
Altmetric
Perspective

Intellectual property protection

Strategies for antibody inventions

Pages 310-317 | Received 28 Feb 2011, Accepted 18 Mar 2011, Published online: 01 May 2011

Abstract

In the last decade, therapeutic antibodies have become one of the commercially most successful classes of biopharmaceutical drugs. Major drug manufacturers who have successfully managed to occupy this new market, as well as biotechnology firms, some of which have experienced a quick growth and are now on par with the former, owe part of their success to suitable intellectual property strategies. This article provides an overview of the current thinking on antibody-related patents, and discusses strategies for protecting the antibody products of the future.

Introduction

Patent protection for most of the anti- bodies with large global markets will expire within the next three to seven years, thus exposing the respective drugs to competition by biosimilar manufacturers (). To protect their market leadership, numerous companies are therefore in a process of developing or already have on the market, second generation antibodies. An invention for which patent protection is sought has to pass particular tests, which are, among others, the novelty requirement, the inventive step/non-obviousness requirement and the written description and enablement requirement.Citation1 These requirements are, however, not absolute, but subject to constant changes.

The Moving Target

Antibody engineering and design underwent substantial advancements in the past 20 years, including development of recombinant chimerization and humanization techniques and the creation of libraries, display methods and affinity maturation approaches. However, in a global knowledge society, a method that was cutting-edge technology yesterday may be an industry standard today, particularly with respect to technical disciplines that are strongly influenced by academic research, as is the case with therapeutic antibodies.

This situation is reflected in the increasing scrutiny patent authorities exhibit with respect to antibody-related patent applications. The hurdles are steadily set higher or, as the European Patent Office (EPO) puts it, “the bars are raised.”

Patent protection plays a crucial role in the pharmaceutical industry because of its reliance on rapidly changing technology. Because of high upfront disbursements for research and lengthy clinical development and approval procedures (with the respective outcomes by no means predictable), the industry depends on efficient patent protection to assure a sufficient return of investment. This phenomenon can be summed up in the phrase “no patent equals no product”. To ensure that the scientific development of new antibody drugs will continue in the future, it is thus important to co-develop suitable patent strategies.

Inventive Step/Non-Obviousness

Probably due to the rapid technological progress in the antibody industry, arguments that were accepted in support of sufficient inventiveness in the past now may be rejected by the patent authorities as falling under the routine of a skilled artisan. In view of the fact that technologies for the production of a human antibody against every conceivable target are now state of the art (consider, e.g., native antibody libraries and phage display), the mere provision of a human antibody against a target the clinical implications of which are known would have difficulties to meet the inventive step/non-obviousness requirement. In other words, the antibody industry is, in some way, a victim of its own success.

In order to anticipate obvious objections during patent prosecution, applicants should add to their applications fall-back positions, like sequences describing the specific antibody in great detail and/or experimental data with respect to particular binding properties, aggregation behavior, blood clearance, cross-reactivity and the like. Such data can often be used as a last resort to obtain patent protection for the actual antibody. Further, most of these data may also be used to meet the written description and enablement requirement (see below). A look into pertinent databases, like the board of appeal decisions database of the EPOCitation2, shows that patent applications related to therapeutic antibodies have rarely been rejected for lack of inventive step/obviousness alone. One reason for this might be that applicants seem zo provide sufficient data that can be used as fall-back positions.

Written Description and Enablement Requirement

The enablement requirement, which is common to both European and US patent law, strives to ensure that a skilled person can reproduce the subject matter of the invention without undue burden. One example is usually sufficient to provide enablement, as long as no evidence exists that embodiments falling under the scope of the patent are not enabled. In case such evidence exists, the patent examiner may decide to narrow the scope of the claims to the very embodiment for which enabling data have been presented.

One example for the increasing scrutiny with respect to sufficient enablement is given in EPO decision T0601/05,Citation3 which is related to a first generation patent claiming human monoclonal antibodies (mAbs) that bind to human tumor necrosis factor (TNF)α. The only method for the production of the claimed antibodies disclosed in the patent was the hybridoma technique developed by Köhler and Milstein in the 1970s.Citation4 However, the board held that the hybridoma technique would not be suited to prepare high affinity antibodies against TNFα because human peripheral blood cells cannot provide a route to high affinity, neutralizing antibodies against self-antigens, but only to low affinity antibodies. Because the claim language encompassed both, the claim was found to be not sufficiently enabled by the specification.

An example for the ambiguous positions patent authorities have taken with respect to the enablement requirement is EP939804 assigned to Human Genome Sciences (HGS). Said patent comprises claims related to an antibody that binds specifically to Neutrokine-α, which is a member of the TNFα superfamily and was discovered by HGS on the basis of bioinformatic investigations alone. Regarding therapeutic implications, the patent comprised no experimental data, only tissue distribution data of Neutrokine-α mRNA. It was thus alleged that HGS had postulated a potential therapeutic use only on the basis of the known relationship to TNF. In decision T0018/09,Citation5 the EPO judged, however, that the tissue distribution data suffice to provide a valid basis for an industrial application and may be used to develop appropriate means and methods for diagnosis and treatment and, therefore, maintained the patent in slightly amended form. A year before the UK High Court found the patent invalid for lack of industrial applicability, insufficiency and obviousness, said decision having effect for the UK only. This decision was confirmed by the UK Court of Appeal recently in decision Eli Lilly and Company vs. Human Genome Sciences.Citation6

In addition to the enablement requirement, US law also provides a written description requirement in order to ensure that the inventor had, at the filing date, full possession of the entire claimed subject matter. In Centocor Ortho Biotech, Inc. vs. Abbott Laboratories,Citation7 Centocor sued Abbott for patent infringement by selling adalimumab (Humira®). Basis for the legal action was Centocor's US7070775, which relates to human antibodies to human TNFα. The ‘775 patent is a continuation in part (CIP) of an earlier application by Centocor, which was related to chimeric antibodies. However, said earlier patent predated a patent by Abbott related to similar subject matter. The case has generated broad public interest due to a record verdict in the first instance under which Abbott was sentenced to pay $1.67 billion in damages. On appeal, the decision was fully reversed by the US Court of Appeals for the Federal Circuit (CAFC) only for lack of written description. The CAFC considered that most claims of the ‘775 patent lacked written description, because the specification did not describe the claimed human antibody, nor an antibody with a human variable region, and concluded that “the scope of Centocor's right to exclude cannot over-reach the scope of its contribution to the field of art as described in the patent specification.” The claims on which Abbott had been sued were thus declared invalid.

The written description requirement was recently confirmed in the CAFC decision Ariad vs. Eli Lilly,Citation8 related to Ariad's US6410516. The patent, which dealt with transcription factor NFκB and methods of reducing or altering its activity without indicating how this could actually be done, was found invalid for failure to meet the written description requirement. The decision fuels fears that the written description requirement discriminates against universities and start up ventures that have their emphasis in basic research. These entities are under constant pressure to secure their results at the earliest possible date, and to the broadest possible extent, in order to publish them or present them to potential licensees. A requirement for additional data in the future will increase the financial burden for these small or non-commercial entities.

Novelty

Contrary to increasing requirements as to inventive step/non-obviousness and to written description and enablement, the relevant authorities have lowered hurdles with respect to the novelty requirement. Recent case law with respect to small molecules has strengthened the concept of selection inventions, which is established granting practice at the EPO already and which stipulates that the disclosure of a chemical class does not necessarily anticipate the novelty of an individual compound falling within this class (so called “genus-species anticipation,” according to which a species anticipates the genus, whereas the genus does not anticipate a species).

This means, for example, that, despite the fact that the racemate of a given structure is prior art, a patent related to only one enantiomer of said racemate may be considered novel, and thus patentable in case the inventive step requirement is met as well (e.g., due to difficult resolution of the racemate). This view has been consented by courts in the UK, Germany and the US with respect to the (+)-enantiomer of Citalopram (decisions Generics UK vs. Daichi,Citation9 BGH EscitalopramCitation10 and Forest Labs., Inc. vs. Ivax Pharm., Inc.).Citation11

In another example, courts in all three countries agreed that a given compound, which falls within the scope of a general formula disclosed in the prior art, can be considered novel if it is not mentioned explicitly in the latter, but only by means of a Markush group in which some substituents are designated as R1-RX. Courts in UK, Germany and the US came to similar results in cases related to the antipsychotic olanzapine (decisions Dr. Reddy's vs. Eli Lilly,Citation12 BGH OlanzapinCitation13 and Eli Lilly & Co. vs. Zenith Goldline Pharm., Inc.).Citation14

Translated to biomolecules, this means that, e.g., a sequence claim related to a second generation antibody will be considered novel even if said claimed sequence is comprised in the similarity interval of a prior sequence disclosure (e.g., “SEQ ID No 1 or sequences having a similarity of >95% with the former”).

Approaches to Protect Therapeutic Antibody Products

The approaches to protect therapeutic antibody products have been previously discussed.Citation15 In short, the respective antibody can be specified by (1) binding a specific target, (2) having specific binding characteristics against a given target, (3) reference to a specific deposited cell line or a specific production process or (4) by having, or being encoded by, a specific amino acid/DNA sequence with respect to the whole antibody or to sections therof. Crucial issues for selecting the right strategy are the scope of protection that can be obtained with a given claim language, and the prospects of patent allowance by the patent authorities. Generally, for first generation antibody patents, a broad claim language could be used (e.g., by specifying the target only), thus providing a broad scope of protection, while in second or higher generation patents the claim language must be more restrictive (e.g., by specifying the amino acid/DNA sequence), thus providing only a narrow scope of protection. Said trend is illustrated, for example, by the history of patents protecting anti-TNFα antibodies ().

Higher generation antibody patents claim, in most cases, a sequence, at least with respect to one or more complementarity determining regions. In such case, the inventive step/non-obviousness requirement is met more easily, because according to well-established lines of argumentation, the skilled person would very unlikely find any incentive in the prior art to arrive at a specific sequence, at least if such sequence has not merely been isolated from nature, but is the subject of, e.g, an affinity maturation process. Further, the enablement/ disclosure hurdle is passed more easily because a given sequence is fully disclosed and enabled by merely mentioning it in the claims or the specification.

Admittedly, the scope of protection of such claim language is quite narrow, although to date it is still unclear whether or not such claim types enjoy a scope of equivalence and, if so, to what extent. Further, if such patent protects an approved antibody therapeutic, third parties wanting to put a follow-on biological (also called “biosimilar”) on the market cannot simply substitute one or more amino acid residues to avoid a potential patent infringement. Under the respective draft guidelines recently published by the European Medicines Agency (EMA),Citation16 and subject to public consultation until May 31, 2011, the resulting antibody would most probably no longer qualify as a biosimilar, as amino acid sequence identity is considered to be a conditio sine qua non to obtain antibody biosimilar status.Citation17

Antibody Evolution

In view of expiring patent protection for first generation antibody therapeutics, major drug makers and biotechnology firms are currently in a process of developing second generation variants of their or their competitors' antibodies. Generally, the improvements that eventually give rise to a patent allowance have been achieved by use of methods that are now considered to belong to the standard toolbox of antibody engineering, i.e., humanization, affinity maturation and glycoengineering techniques ().

New Targets

While cellular signalling processes are well-understood, new potential targets for antibody therapy are still being discovered. Today, about 100 such targets are addressed by approved biopharmaceuticals,Citation18 but the spectrum of soluble proteins or membrane receptors that represent potential therapeutic targets should be much higher. Although the evaluation of a new target and the subsequent development of a respective antibody are costly endeavors, recent advancements in antibody technology may accelerate the validation of new targets, in particular those relevent to cancer, autoimmune diseases, infectious diseases and neurodegenerative diseases.

Filing a patent application for an antibody against a new target is, usually, a safe bet. In case a patent application describes, and claims, a new protein that may play a physiological role in the human body, it is common to also draft a claim related to a theoretical antibody against said protein, i.e., an antibody which has not actually been manufactured. Such type of claim is routinely granted in the case when the target protein is novel and substantially defined, even if the applicant has not produced such an antibody or provides no data or enablement related to such antibody (see, for example, US decision Noelle vs. LedermanCitation19 or EPO technical board decision T0542/95).Citation20

In both cases, the rationale behind this position was that the provision and correct specification, of a novel protein X enables a skilled person to produce an antibody against said protein. Therefore, it is considered a fair reward for the applicant of protein X to be granted a claim related to a theoretical antibody against said protein.

However, publication of limited data for a protein that is part of a cellular signalling process does not automatically compromise the inventive step/non-obviousness of a patent claim related to an antibody against such protein. This is because it may remain unclear whether or not said moiety is involved in a pathogenic process and, if so, whether underexpression or overexpression, or expression of a dysfunctional or misfunctional product, is responsible for the pathologic condition, or if the moiety is causative for, or a consequence of, said pathologic condition.

Ganymed (Mainz, Germany) developed a series of antibodies against newly found antigens that are specific for different types of cancer. The physiological role of these antigens is not fully clear. However, it seems that the respective antibodies are not meant to interfere in cellular signalling processes, as is the case, for example, in anti VEGF therapy. The idea is rather to evoke antibody-dependent cell-mediated cytotoxicity responses against the respective cells.

LPath (San Diego, CA) has pursued a different concept. The company developed a mAb against a non-protein target, i.e., the sphingolipd sphingosine-1-phosphate, which is a tumor growth factor. LPath claims that this drug has a direct effect on angiogenesis in addition to a direct effect on tumor cells themselves, i.e., inhibition of metastasis, tumor cell growth and apoptosis, and thus combines effects of some marketed antibodies.

Merrimack (Cambridge, MA) is developing MM-111, which is an IgG-like bispecific antibody whereby, unlike natural antibodies, one arm binds the human epidermal growth factor receptor 2 (HER2) and a second arm binds the HER3 receptor. It is claimed that the antibody uses the HER2 target to block the HER3 pathway, as it appears that HER3 signalling is an important therapeutic target in HER2-positive cancers.Citation21 summarizes the above examples.

New Antibody Formats

Strictly speaking, when first introduced, formats such as chimerized antibodies, humanized antibodies, antigen binding fragments (Fab) and single chain variable fragments were considered new antibody formats, and were (or still are) subject to patent protection. The basic concept of rearranging and recombining different components of IgGs was further pursued in the last decade. Potential advantages of new antibody formats compared to full-size molecules depend on the respective nature of the format and encompass, for example, lack of glycosylation, lack of disulfide bridges, reduced molecular weight, better stability and serum half life, better tissue penetration, lower immunogenicity, straightforward transfer from animal trials to humans, suitability for oral administration, expression advantages (e.g., expression in E. coli or yeast instead of Chinese hamster ovary cells), higher efficiency and ease of selection/screening. These advantages can be referred to to meet the inventive step/non-obviousness requirement in first generation patents claiming the respective antibody formats or their technology. Relevant patents on major advancements in this field are listed in .

In most cases, companies have first established, and protected, the basic enabling technologies related to the new format as such. In a subsequent step, specific drug candidates are developed, thereby forming the subject of respective patent applications.

One example resulting from a new antibody format is Symphogen's (Lyngby, Denmark) Sym004, which is a recombinant IgG1 antibody product consisting of two antibodies targeting distinct non-overlapping epitopes in epidermal growth factor receptor (EGFR) extracellular domain III and which may, one day, compete with JmClone's Cetuximab. In comparison to the latter, Sym004 is said to induce removal of the receptor from the cancer cell surface, leading to more pronounced cancer growth inhibition. The product is undergoing evaluation in a clinical Phase 1 study [NCT01117428] of patients with advanced solid tumors, and is pursued, among others, under the European Patent application EP2132229A1 and related patent family members. The basis for upcoming inventive step/non-obviousness considerations will probably be advantageous binding properties compared to prior art products (e.g., Cetuximab) due to the polyclonality of the product.

Another example is Philogen's (Sovicille, Italy) L19-TNFα, which consists of the human antibody L19, which targets the extradomain B of fibronectin, fused to human TNF. In this construct, the L19 domain provides vascular targeting of the TNF domain to the site of disease, where the latter exerts its antitumor activity. The product is said to have superior anti-carcinogenic effect. Respective experimental data put the corresponding patent application ready for grant by the EPO, and the product candidate is now protected under EP1257297B1.

Yet another example is ATN-103, which is an anti-TNF Nanobody® developed by Ablynx (Ghent, Belgium) that is currently undergoing evaluation in clinical studies as a treatment for rheumatoid arthritis. ATN-103 targets the same antigen as the marketed antibody drugs Adalimumab (Humira®), Infliximab (Remicade®), Golimumab (Simponi®) and Certolizumab pegol (Cimzia®), as well as the fusion protein Etanercept (Enbrel®) and is said to have a variety of advantages related to administration and pharmacokinetics, which are currently used as a basis for inventive step/non-obviousness argumentation in the patent prosecution of European Patent Application EP1558647A1.

Antibody Mimetics

Proteins not belonging to the immunglobulin family and even non-proteins such as aptamers or synthetic polymers, have also been suggested as alternatives to antibodies.Citation22 One reason for the increasing interest in these so-called “alternative scaffolds,” or “antibody mimetics,” is the barrier to entry into the field created by existing antibody IP. As with new antibody formats, potential advantages of new antibody mimetics depend on their respective structural characteristics. These specific advantages may be used as a basis for patentability, i.e., in order to meet the requirements towards novelty and inventive step/non-obviousness. An overview of some selected approaches is shown in . Some product candidates derived from these approaches have already entered the clinical phase, while others are still in the preclinical phase.

Companies have in most cases first established and protected the basic scaffold technologies, and have then started to develop specific drug candidates, i.e., scaffold-based products that bind a given target. The approach has the risk that the respective patent applications meant to protect these products may not be considered as inventive/non-obvious by the respective authorities. The rationale behind such considerations is that both (1) the respective scaffold and its implicit advantages and (2) the respective target and its clinical implications were already known to the skilled person at the priority date of said second-generation patent application. The mere combination of a known scaffold and a known target, although novel, may thus be considered obvious to the skilled person.

Therefore, to obtain patent protection for such products, advantageous properties of the product, or, ideally, an unexpected synergism between the scaffold and the target, should be disclosed in the patent, in order to be at hand as fallback position or as basis for a respective argumentation with respect to meet the inventive step/non-obviousness requirement.

Molecular Partners (Schlieren, Switzerland) has developed an ankyrin-based drug for the treatment for age-related macular degeneration (MP0112), which targets vascular endothelial growth factor (VEGF) A and, thus, would compete, if approved, with the marketed Fab fragment ranibizumab (Lucentis®). MP0112, which is pursued under the international patent application WO2010060748A1, is said to have a higher target affinity than the former. In the respective claims, a minimum Kd of less than 10−7 M is claimed, the latter being the basis for upcoming inventive step/non-obviousness considerations in the respective prosecution proceedings.

Another example is PRS-050 by Pieris AG (Freising, Germany). Clinical trials for this drug, which is a PEGylated Anticalin®, were initiated in 2010. PRS-050 binds and neutralizes human and murine VEGF with picomolar affinity, and is said to be used as a drug for treating solid tumors. Pieris claims that, compared to other anti-VEGF antibodies, PRS-050 exhibits better tissue penetration and, consequently, can infiltrate tumors much easier, and has a shorter half-life which supports VEGF removal from the body. Further, Pieris claims that the drug does not aggregate. The drug is currently protected under EP2046820 B1 and related patents.

Scil proteins (Halle, Germany) has in its pipeline proprietary ubiquitin-based drug candidates called Affilin® therapeutics. The lead candidate SPVF2801-30 is in preclinical development. SPVF2801-30 is a fusion protein comprising a heterodimeric Affilin®, plus a specific cytokine. Despite systemic advantages of their proprietary ubiquitin-based Affilin® therapeutics, Scil provides data supporting superior target affinity and specificity of the claimed drug. Patent protection is currently pursued under PCT application No PCT/EP2010/069665.

Conclusion

The quick advancements of antibody technologies require a steady adaptation of patent strategies, to ensure that future products will still be protected by IP rights. While requirements as to inventive step/non-obviousness and written description and enablement seem to be on the rise, the hurdles with respect to the novelty requirement have been lowered. Companies and research institutions which are involved in the development of new therapeutic antibody products should develop an adequate IP expertise or seek expert advice, to account for these developments, in order to be able to protect their investments for research and development.

Disclaimer

The information provided herein reflect the personal views and considerations of the author. They do not represent legal counsel and should not be attributed to Michalski • Hüttermann & Partner Patent Attorneys or to any of its clients. Patent numbers and patent lifetimes have been verified with utmost care, but no liability is taken for their correctness.

Figures and Tables

Table 1 Commercially most successful therapeutic antibodies and their key patents

Table 2 Selected key patents protecting anti-TNFα antibodies

Table 3 2nd generation antibodies and their key patents

Table 4 Selected therapeutic antibodies against new targets

Table 5 Selected new antibody formats and their key patents

Table 6 Selected antibody mimetics and their key patents

References

  • McCabe K. Guardians at the gate: Patent protection for therapeutic monoclonal antibodies-part 1. mAbs 2009; 1:382 - 384
  • www.epo.org/law-practice/case-law-appeals/search.html
  • EPO Technical Board decision T0601/05 2009; EPO Board of appeal decisions database T0601/05
  • Köhler GF, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975; 256:495 - 497
  • EPO Technical Board decision T0018/09 2009; EPO Board of appeal decisions database T0018/09
  • Eli Lilly and Company vs. Human Genome Sciences, [2008] EWHC 1903 (Pat), 2008 Bailii EWHC 1903
  • Centocor Ortho Biotech, Inc., v. Abbott Labs, 07-CV-0139, 2011 US App. LEXIS 3514
  • Ariad Pharmaceuticals, Inc., v. Eli Lilly and Company, 598 F.3d 1336, 2010 US App. LEXIS 5966
  • Generics UK vs. Daiichi, [2008] EWHC 2413 (Pat), 2008 Bailii EWHC 2413
  • Escitalopram, Xa ZR 130/07 (BPatG), 2009. GRUR 2010; 123
  • Forest Labs., Inc., v. Ivax Pharm., Inc., 501 F.3d 1263, 2007 US App. LEXIS 21165
  • Dr. Reddy's vs. Eli Lilly and Company, [2008] EWHC 2345 (Pat), 2008 Bailii EWHC 2345
  • Olanzapin X ZR 89/07 (BPatG) 2008. GRUR 2009; 382
  • Eli Lilly & Co., v. Zenith Goldline Pharm., Inc., 05-1396, 05-1429, 05-1430, 2007 US App. LEXIS 8750
  • Storz U. Kontermann R, Duebel S. IP Issues in the Therapeutic Antibody Industry. Antibody Engineering 2010; 2nd edition Springer 517 - 581
  • Guideline on similar biological medicinal products containing monoclonal antibodies (EMA/CHMP/BMWP/403543/2010), and Guideline on immunogenicity assessment of monoclonal antibodies intended for in vivo clinical use (EMA/CHMP/BMWP/86289/2010) www.ema.europa.eu
  • Schneider CK, Kalinke U. Toward biosimilar monoclonal antibodies. Nat Biotechnol 2008; 26:985 - 990
  • Overington JP, Al-Lazikani B, Hopkins AL. How many drug targets are there?. Nat Rev Drug Discov 2006; 5:993 - 996
  • Noelle v. Lederman, 355 F.3d 1343, 2004 US App. LEXIS 774
  • EPO Technical Board decision T0542/95. 1999 EPO Board of appeal decisions database T0542/95
  • Denlinger CS, et al. A phase I/II and pharmacologic study of MM-111 in patients with advanced, refractory HER2-positive (HER2+) cancers. J Clin Oncol (Meeting Abstracts) 2010; 5 28:15
  • Gebauer M, Skerra A. Engineered protein scaffolds as next-generation antibody therapeutics. Curr Opin Chem Biol 2009; 13:245 - 255

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.