throbber
Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 1 of 123 PageID# 9730
`
`IN THE UNITED STATES DISTRICT COURT
`FOR THE EASTERN DISTRICT OF VIRGINIA
`Norfolk Division
`
`
`
`Civil Action No. 2:23-cv-0222 (JKW-DEM)
`
`JURY TRIAL DEMANDED
`
`BioNTech SE, BioNTech Manufacturing
`GmbH, and Pfizer Inc.,
`
`
`
`
`
`CureVac SE (f/k/a CureVac AG),
`
`
`
`
`Plaintiffs and Counter Defendants,
`
`v.
`
`Defendant and Counter Claimant,
`
`and
`
`
`CureVac Manufacturing GmbH,
`
`
`Counter Claimant.
`
`
`
`
`
`
`
`BIONTECH AND PFIZER’S SECOND AMENDED COUNTERCLAIMS AND
`ANSWER TO CUREVAC’S FIRST AMENDED COUNTERCLAIMS
`
`COUNTERCLAIMS
`
`Pursuant to this Court’s Order (ECF No. 240), without admitting any of the allegations of
`
`CureVac SE and CureVac Manufacturing GmbH (collectively, “CureVac” or “Defendants”)
`
`other than those expressly admitted herein, and without prejudice to the right of BioNTech SE
`
`and BioNTech Manufacturing GmbH (collectively, “BioNTech”) and Pfizer Inc. (“Pfizer” and,
`
`together with BioNTech, “Plaintiffs”) to plead additional counterclaims as the facts of the matter
`
`warrant, BioNTech and Pfizer assert the following counterclaims against CureVac.
`
`INTRODUCTION
`
`1.
`
`This action involves Plaintiffs’ and Defendants’ respective independent efforts to
`
`develop vaccines to combat the COVID-19 pandemic. Plaintiffs’ Comirnaty® was the world’s
`
`first mRNA vaccine approved for public use, deployed in record time, and proved to be effective
`
`in preventing severe disease, hospitalization, and death from the COVID-19 pandemic. BioNTech
`
`
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 2 of 123 PageID# 9731
`
`
`
`worked tirelessly to create the mRNA vaccine after years of research and development of mRNA
`
`technology, collaborating with Pfizer to bring the vaccine through regulatory approval and
`
`distribution to combat this global pandemic. All of the investment and work paid off—BioNTech
`
`and Pfizer successfully developed an mRNA vaccine, proved its efficacy, established global
`
`manufacturing and supply chains, and gained regulatory approval. Their efforts played a vital role
`
`in managing the global COVID-19 crisis.
`
`2.
`
`CureVac also tried to develop a vaccine to help the fight against COVID-19. Unlike
`
`BioNTech and Pfizer, CureVac was unsuccessful. Presumably using its alleged patented
`
`technology, CureVac’s vaccine was an unsuccessful treatment and lacked sufficient efficacy for
`
`regulatory approval.
`
`3.
`
`Failing to supply a useful vaccine, CureVac now attempts to profit from BioNTech
`
`and Pfizer’s success through allegations of patent infringement. BioNTech and Pfizer, however,
`
`developed their Comirnaty® vaccine without any contribution from CureVac’s alleged mRNA
`
`technology—which is why BioNTech and Pfizer brought this declaratory judgment action—
`
`instead relying on innovations from their own scientists and coordination with the global scientific
`
`community. CureVac played no part in Comirnaty®’s stunning success.
`
`COMIRNATY® WAS BUILT ON DECADES OF PLAINTIFFS’
`AND THEIR PARTNERS’ FOUNDATIONAL RESEARCH—NOT CUREVAC’S
`
`4.
`
`Comirnaty® was the first-approved vaccine utilizing messenger RNA (“mRNA”)
`
`technology. If efficacious, an mRNA vaccine works by introducing into a person mRNA that
`
`instructs the body to make a certain protein, such as a piece of a virus that the vaccine seeks to
`
`protect against. When that protein is made, or “expressed,” by a person’s cells, that person’s
`
`immune system can recognize the protein as foreign and develop an immune response to it. If that
`
`person is later infected with the actual virus itself, his or her immune system is ready to protect
`
`2
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 3 of 123 PageID# 9732
`
`
`
`against or minimize the severity of the viral infection. This is unlike previously approved
`
`non-mRNA vaccines, developed before the COVID-19 pandemic, such as weakened or inactivated
`
`viruses injected into the patient.
`
`5.
`
`Scientists have known since the 1970s that mRNA has the potential to be
`
`administered as a therapeutic to translate a protein that may treat or prevent disease in humans. By
`
`the 1990s, researchers demonstrated that mRNA administered as a therapeutic could be used to
`
`elicit antiviral immune responses in animal models, e.g., encoding proteins expressed by cancer
`
`cells to induce an immune response.
`
`6.
`
`One vexing problem encountered by researchers, however, was that synthetic
`
`mRNA can trigger proteins that result in a non-antigen-specific immune response, such as
`
`activation of toll-like receptors. This can lead to an undesirable reaction in the body, such as
`
`inflammation. Despite this, Dr. Katalin Karikó (a BioNTech scientist and professor at the
`
`University of Pennsylvania), was convinced that mRNA structures could be used to instruct cells
`
`to make their own therapeutic proteins.1
`
`7.
`
`In the mid-2000s, after years of painstaking research, Dr. Karikó and Dr. Drew
`
`Weissman made a key breakthrough while they were both at the University of Pennsylvania: they
`
`discovered that certain chemical modifications to RNA nucleosides could reduce or eliminate the
`
`inflammatory reaction. They showed that the unmodified mRNA that they expressed induced an
`
`
`1 As Dr. Anthony Fauci acknowledged, Dr. Karikó “was, in a positive sense, kind of obsessed with
`the concept of messenger RNA.” (D.I. 104, Ex. 1 at 1.) Despite her tenacity, Dr. Karikó struggled
`to stay afloat in academia, as she sought—and was denied—grant after grant to pursue ideas that
`seemed wild and fanciful to many in the academic community. (Id. at 1-2.) As one of her
`colleagues explained, “[w]hen your idea is against the conventional wisdom that makes sense to
`the star chamber, it is very hard to break out.” (Id. at 1.) Yet, Dr. Karikó’s focus and drive never
`wavered. Her genius was a “willingness to accept failure and keep trying, and her ability to answer
`questions people were not smart enough to ask.” (Id. at 3.)
`
`3
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 4 of 123 PageID# 9733
`
`
`
`immune response, while the control—called transfer RNA (“tRNA”), an intermediary molecule
`
`used during protein translation that links the mRNA and the amino acid sequence of proteins—did
`
`not. In particular, they discovered that a class of nucleotides called pseudouridines found in tRNA
`
`allowed it to evade the cell’s internal immune response.
`
`8.
`
`This led Drs. Karikó and Weissman to investigate the idea of modifying uridines in
`
`mRNA with naturally occurring pseudouridines found in tRNA, including 1-methylpseudouridine.
`
`They discovered that the uridine modification helped synthetic mRNA evade the body’s innate
`
`immune system. Drs. Karikó and Weissman published their insights in a series of research papers,
`
`including a seminal 2005 paper titled “Suppression of RNA Recognition by Toll-like Receptors:
`
`The Impact of Nucleoside Modification and the Evolutionary Origin of RNA.” (D.I. 104, Ex. 2.)2
`
`9.
`
`Drs. Karikó and Weissman presented their ideas to pharmaceutical companies and
`
`venture capitalists. At first, no one was interested. As Dr. Weissman later recounted, “[w]e were
`
`screaming a lot, but no one would listen.” (D.I. 104, Ex. 1 at 4.) BioNTech, however, took notice
`
`of Drs. Karikó and Weissman’s work and began funding Dr. Karikó’s laboratory. (Id.) In 2013,
`
`Dr. Karikó joined BioNTech full-time as a senior vice president.
`
`10.
`
`Drs. Karikó and Weissman’s discovery that modified mRNA nucleosides could
`
`evade the cell’s internal immune response was the critical innovation behind the only fully
`
`
`2 Drs. Karikó and Weissman patented their groundbreaking discovery by submitting, in 2005,
`Provisional Patent Application No. 60/710,164 titled “RNA Containing Modified Nucleosides and
`Methods of Use Thereof.” (D.I. 104, Ex. 3.) The ’164 application describes how “[t]his invention
`provides RNA . . . comprising pseudouridine or a modified nucleoside” and expressly identifies
`N1-methyl-pseudouridine. (D.I. 104, Ex. 3 at 1, 14.) The ’164 application further “provides
`methods of reducing the immunogenicity of RNA” by using mRNA with pseudouridine
`nucleotides. (Id. at 1.) The U.S. Patent Office eventually granted U.S. Patent No. 8,691,966 (“the
`’966 patent”) to Drs. Karikó and Weissman, which claims priority to the ’164 application. (D.I.
`104, Ex. 4.) The ’966 patent expressly claims mRNA comprising “a modified nucleoside selected
`from the group consisting of (i) 1-methypseudouridine (m1Ψ) and (ii) pseudouridine (Ψ).” (D.I.
`104, Ex. 4 at claim 1.)
`
`4
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 5 of 123 PageID# 9734
`
`
`
`approved mRNA COVID-19 vaccines, BioNTech and Pfizer’s Comirnaty® and Moderna’s
`
`Spikevax®. Moderna’s co-founder, Derrick Rossi, recognized this discovery as “fundamental to
`
`this entire field” of mRNA vaccines and therapeutics. (D.I. 104, Ex. 5 at 2.) In Dr. Rossi’s
`
`estimation, Drs. Karikó and Weissman’s work will “earn them a Nobel Prize because it really is
`
`what allows these mRNA vaccines and any mRNA therapeutic down the road” (id.), and
`
`Moderna’s co-founder reiterated that, “[i]f anyone asks [him] whom to vote for some day down
`
`the line, [he] would put them front and center” (D.I. 104, Ex. 6 at 7). According to Dr. Rossi, Drs.
`
`Karikó and Weissman’s “fundamental discovery is going to go into medicines that help the world.”
`
`(Id.) In fact, Moderna backed up Dr. Rossi’s belief with its pocketbook by taking a license from
`
`the University of Pennsylvania’s successor-in-interest, Cellscript, LLC so it could practice patents
`
`embodying Drs. Karikó and Weissman’s “fundamental discovery,” including patents disclosing
`
`the modified uridine that Moderna’s mRNA vaccine uses. (Id.; D.I. 104, Ex. 7.)
`
`11.
`
`For their discovery, Drs. Karikó and Weissman have been honored on several
`
`occasions by institutions such as the Columbia University Irving Medical Center and the European
`
`Patent Office for their “trailblazing” work, which “laid the foundation for the creation of [an]
`
`incredibly effective COVID-19 vaccine[.]” (Exs. 8, 9; see also Exs. 10 and 11.) Drs. Karikó and
`
`Weissman have also been presented with many other awards, such as the Princess of Asturias
`
`Award, the Albany Medical Center Prize in Medicine and Biomedical Research, the 2022
`
`Breakthrough Prize in Life Sciences, and the 2021 Lasker Award—America’s top biomedical
`
`research prize. (D.I. 104, Exs. 10, 12, 13, 14, 15, and 16.)
`
`12.
`
`Dr. Karikó’s continued research on modified mRNA at BioNTech included
`
`determining that an mRNA vaccine could elicit antibodies against the Zika virus. In 2017, Dr.
`
`Karikó co-authored a paper in Nature (the “2017 Nature Paper”) demonstrating that “a single low-
`
`5
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 6 of 123 PageID# 9735
`
`
`
`dose intradermal immunization with lipid-nanoparticle-encapsulated nucleoside modified mRNA
`
`(mRNA-LNP) encoding the pre-membrane and envelope glycoproteins of a strain from the Zika
`
`Outbreak in 2013 elicited potent and durable neutralizing antibody responses” in animal models.
`
`(D.I. 104, Ex. 17 at 1.) The mRNA vaccine developed by BioNTech and the University of
`
`Pennsylvania against the Zika virus used mRNA that contained the modified nucleoside
`
`1-methylpseudouridine, which is the same modified nucleoside that would later be used in
`
`Comirnaty®. (Id. at 2–3.)
`
`13.
`
`BioNTech’s development work included additional discoveries as part of its mRNA
`
`platform. For example, by 2014, scientists at BioNTech created the disrupted poly(A) tail that
`
`would later be used in Comirnaty®. (See, e.g., D.I. 104, Ex. 18.) BioNTech’s development work
`
`also involved collaborations with various partners. For example, by 2017, Acuitas Therapeutics,
`
`Inc. (“Acuitas”) and BioNTech were collaborating on the development of mRNA therapeutic
`
`products using Acuitas’ technology as a delivery system. During this time, Acuitas painstakingly
`
`engineered a microscopic sphere of fats called a lipid nanoparticle (“LNP”) that can envelop and
`
`protect the mRNA. These LNPs allow the mRNA to cross the membrane of a human cell and then
`
`release the mRNA payload so it can be used to create the proteins that can potentially generate a
`
`protective immune response. BioNTech licensed LNPs from Acuitas for use with mRNA
`
`therapeutic products.
`
`14.
`
`BioNTech’s scientists, including Dr. Karikó, demonstrated that modified mRNA
`
`vaccines successfully conferred immunity against HIV, Zika, and influenza viruses in animal
`
`models; and published these results in the Journal of Experimental Medicine (the “2018 JEM
`
`Paper”). (See D.I. 105, Ex. 19.) The 2018 JEM Paper recognized that BioNTech’s mRNA vaccine
`
`6
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 7 of 123 PageID# 9736
`
`
`
`platform has the “advantages of a favorable safety profile, potentially inexpensive manufacturing,
`
`and the capacity for rapid development in emerging epidemics.” (Id. at 1580 (emphasis added).)
`
`15.
`
`That same year, Pfizer and BioNTech partnered to develop an mRNA-based
`
`vaccine for influenza. As part of the agreement, BioNTech and Pfizer would jointly conduct
`
`research and development to advance mRNA-based flu vaccines. In announcing the collaboration,
`
`the head of Pfizer’s vaccine research and development unit, Dr. Kathrin Jansen, noted that
`
`“[i]nnovative vaccine approaches are urgently needed to provide improved protection against
`
`seasonal flu, and to respond rapidly and in quantity to pandemic influenza threats.” (D.I. 105,
`
`Ex. 20 at 1 (emphasis added).) Dr. Jansen further emphasized that “mRNA vaccines offer a novel
`
`approach to code for any protein or multiple proteins, and the potential to manufacture higher
`
`potency flu vaccines more rapidly and at a lower cost than contemporary flu vaccines.” (Id.)
`
`16.
`
`In December 2019, SARS-CoV-2 was reported in Wuhan, China. When this novel
`
`coronavirus emerged, BioNTech was well-positioned to respond rapidly by constructing a vaccine
`
`around its existing modified mRNA platform, which had already been tested against viruses such
`
`as HIV, Zika, and influenza. Leveraging decades of foundational research, BioNTech rapidly
`
`identified several candidates for clinical testing as mRNA-based vaccines to protect against
`
`COVID-19.
`
`17.
`
`By early 2020, soon after the genetic sequence for SARS-CoV-2 (the virus that
`
`causes COVID-19) was published, BioNTech and its development partner Pfizer initiated “Project
`
`Lightspeed,” an accelerated vaccine development program to fight COVID-19. BioNTech and
`
`Pfizer’s COVID-19 vaccine development program leveraged BioNTech’s experience and
`
`expertise with mRNA technologies, as well as the work of other partners, including Acuitas and
`
`the National Institutes of Health (“NIH”).
`
`7
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 8 of 123 PageID# 9737
`
`
`
`18.
`
`For example, BioNTech developed
`
`innovative, proprietary mRNA-based
`
`technologies to achieve effective translational performance and direction of the immune response.
`
`BioNTech also leveraged its prior work with Acuitas on LNP technology and used Acuitas lipids
`
`ALC-315 and ALC-159. Further, BioNTech licensed and incorporated the work of NIH scientists
`
`relating to a particular modification to the sequence of the coronavirus spike protein (i.e., the
`
`protein structures covering the exterior of the SARS-CoV-2 virus), which causes the modified
`
`spike protein to be locked in a certain configuration. (See, e.g., D.I. 105, Ex. 21.) This
`
`configuration allows the modified spike protein to be recognized more easily by human cells and
`
`elicit a more robust bodily response that results in immunity.
`
`19. With the benefit of its prior development work for its mRNA platform, BioNTech
`
`rapidly developed and performed numerous toxicological and pharmacological studies to
`
`determine the safety and efficacy of the Comirnaty® vaccine. For example, BioNTech’s studies
`
`showed, inter alia, that the vaccine is highly immunogenic in animal models and provided the
`
`confirmation needed to move quickly into Phase 1 clinical studies.
`
`20.
`
`BioNTech partnered with Pfizer on
`
`the development, clinical
`
`testing,
`
`manufacturing, distribution, and regulatory approval of the Comirnaty® vaccine. (See D.I. 1,
`
`Exhibit 6.) By March 2020, when the World Health Organization (“WHO”) declared the COVID-
`
`19 outbreak a global pandemic, Pfizer and BioNTech had already begun their collaborative effort.
`
`21.
`
`Clinical trials of the Comirnaty® vaccine began in late April 2020, with preliminary
`
`results demonstrating its safety and efficacy published within six months. This rapid development
`
`and start of clinical trials of product candidates was not a chance event, the result of sudden
`
`inspiration, or copying someone else’s work. It was the result of the relentless work by dedicated
`
`scientists and the vision of BioNTech and Pfizer working together.
`
`8
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 9 of 123 PageID# 9738
`
`
`
`22.
`
`On May 5, 2020, BioNTech and Pfizer announced that the first participants had
`
`been dosed in the United States in the Phase 1/2 clinical trial for their vaccine, codenamed
`
`BNT162, designed to determine safety and efficacy against COVID-19. (See D.I. 1, Exhibit 9.)
`
`After attaining promising Phase 1/2 clinical study results, on July 27, 2020, BioNTech and Pfizer
`
`began a Phase 2/3 study on their Comirnaty® vaccine. (See D.I. 1, Exhibit 12.) The pivotal Phase
`
`3 study was conducted on a global scale—encompassing more than 44,000 patients—to continue
`
`determining its safety and efficacy in humans. (See D.I. 1, Exhibit 8.)
`
`23. Meanwhile, Pfizer was also working on the logistics and infrastructure needed to
`
`successfully manufacture and distribute the Comirnaty® vaccine. Pfizer leveraged its extensive
`
`manufacturing network to produce an approved COVID-19 vaccine as quickly as possible for those
`
`most in need in the United States.
`
`24.
`
`In November 2020, the Comirnaty® vaccine was shown to have met all the primary
`
`efficacy endpoints in a Phase 3 clinical trial, demonstrating an “efficacy rate of 95% (p < 0.0001)
`
`in participants without prior SARS-CoV-2 infection (first primary objective) and in participants
`
`with and without prior SARS-CoV-2 infection (second primary objective),” as measured from
`
`seven days after the second dose of the vaccine. (See D.I. 1, Exhibit 13.)
`
`25.
`
`On November 20, 2020, Pfizer, on behalf of itself and BioNTech, submitted the
`
`clinical trial data as part of an Emergency Use Authorization (“EUA”) request to the Food and
`
`Drug Administration (“FDA”) for administering the Comirnaty® vaccine to people 16 years of age
`
`and older.
`
`9
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 10 of 123 PageID# 9739
`
`
`
`26.
`
`On December 11, 2020, the FDA granted the first EUA for a COVID-19 vaccine
`
`to Pfizer and BioNTech’s Comirnaty® vaccine with vaccinations rolling out immediately
`
`thereafter,3 completing the fastest development of a vaccine in history.
`
`27.
`
`Based on a comprehensive data package and real-world results demonstrating
`
`overwhelming safety and efficacy, on August 23, 2021, the FDA granted full approval of the
`
`Comirnaty® vaccine for individuals 16 years of age and older. In late 2021 and early 2022, the
`
`FDA amended its emergency use authorization for Comirnaty® to permit administration of a
`
`booster dose in certain individuals after completing their primary two-dose series with an FDA-
`
`authorized or approved COVID-19 vaccine, including Comirnaty®. The Comirnaty® vaccine was
`
`both the first mRNA drug product and the first COVID-19 vaccine to receive full FDA approval.
`
`BioNTech Manufacturing GmbH is the Biologics License Application holder for Comirnaty® in
`
`the United States.
`
`28.
`
`Since receiving the first EUA from the FDA, Comirnaty® vaccine has contributed
`
`to saving at least 14 million lives that otherwise may have been lost due to the pandemic. (See
`
`D.I. 1, Exhibit 16.) Furthermore, widespread vaccination of populations with Comirnaty® vaccine
`
`allowed jurisdictions worldwide, including Virginia, to remove restrictions on movement, easing
`
`the economic and social burdens countless individuals suffered during forced isolation.
`
`
`3 FDA Memorandum, Emergency Use Authorization (“EUA”) for an Unapproved Product
`Review (Dec. 11, 2020), https://www.fda.gov/media/144416/download; Press Release, FDA,
`FDA Takes Key Action in Fight Against COVID-19 By Issuing Emergency Use Authorization
`for First COVID-19 Vaccine (Dec. 11, 2020),
`https://www.fda.gov/newsevents/pressannouncements/fda-takes-key-action-fight-against-covid-
`19-issuing-emergency-useauthorization-first-covid-19.
`
`10
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 11 of 123 PageID# 9740
`
`
`
`CUREVAC NOW SEEKS TO PROFIT FROM
`PFIZER AND BIONTECH’S VACCINE AFTER ITS OWN VACCINE FAILED
`
`29.
`
`Like BioNTech and Pfizer, CureVac also tried to develop a vaccine containing
`
`mRNA packaged in an LNP to fight the COVID-19 pandemic. Unlike BioNTech and Pfizer,
`
`however, CureVac failed to create an mRNA vaccine against COVID-19 that induced a sufficient
`
`protective immune response.
`
`30.
`
`Upon information and belief, on or about June 16, 2021, a pivotal Phase 2b/3
`
`clinical trial showed that CureVac’s COVID-19 vaccine candidate, “CVnCoV,” had an interim
`
`vaccine efficacy of about 47% against COVID-19 of any severity, which did not meet the pre-
`
`specified statistical success criteria. (See D.I. 1, Exhibit 4.)
`
`31.
`
`Upon information and belief, an earlier Phase 1 study of CVnCoV showed that
`
`serum levels of so-called neutralizing antibodies, which prevent the virus from binding to cells,
`
`were relatively low in vaccine recipients compared with people who were naturally infected with
`
`SARS-CoV-2.
`
`32.
`
`Upon information and belief, after failing to develop its own effective COVID-19
`
`vaccine, CureVac set its sights on profiting off BioNTech and Pfizer’s Comirnaty® vaccine through
`
`baseless allegations of patent infringement. In its Counterclaims, CureVac improperly attempts to
`
`inflate its scientific contributions to the field of mRNA therapeutics. For example, CureVac
`
`alleges that its “discoveries span all aspects of mRNA medicines, including methods to stabilize
`
`mRNA, to modify it, to manufacture it on a commercial scale, to increase the yield of the protein
`
`it encodes, and to formulate it for safe and effective administration to patients.” (D.I. 106 at ¶ 4.)
`
`Despite all of these “discoveries,” and CureVac’s allegation that it “was the first company in the
`
`world to harness mRNA for medical purposes” (D.I. 106 at ¶ 3), when the global community called
`
`for a COVID-19 vaccine, CureVac was unable to provide one.
`
`11
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 12 of 123 PageID# 9741
`
`
`
`33.
`
`CureVac also alleges that, “[w]hen the COVID-19 pandemic struck, CureVac
`
`scientists leveraged their resources and expertise to find the optimal mRNA sequence encoding
`
`the full-length COVID-19 spike protein and packaged that mRNA in the LNP that resulted from
`
`CureVac’s selection and validation process in the human rabies vaccine trial.” (D.I. 106 at ¶ 26.)
`
`CureVac did not discover the sequence of the SARS-CoV-2 spike protein, despite citing it in U.S.
`
`Patent Nos. 11,241,493 (“the ’493 patent”), 11,471,525 (“the ’525 patent”), 11,576,966 (“the ’966
`
`patent”), and 11,596,686 (“the ’686 patent”); that information was publicly available before
`
`February 2020. (See, e.g., D.I. 105, Exs. 22 and 23.) Likewise, CureVac did not discover the basic
`
`idea of an mRNA vaccine to elicit an immune response against a specific viral antigen or the
`
`particular modification to the sequence of a coronavirus spike protein that stabilizes the spike
`
`protein, which allows it to be used effectively as an mRNA vaccine antigen. Instead, NIH
`
`scientists described the specific spike protein sequence modification. (See, e.g., D.I. 105, Ex. 21.)
`
`34.
`
`CureVac also did not create the LNP in which the mRNA is packaged because that
`
`was done by Acuitas and disclosed publicly prior to the filing of CureVac’s patents in suit. (See,
`
`e.g., D.I. 105, Exs. 24-26.) Moreover, Acuitas has an ownership interest in the ’493, ’525, ’966,
`
`and ’686 patents (collectively, the “’493 Patent Family”). CureVac lacks standing to assert the
`
`’493 Patent Family against BioNTech and Pfizer because Acuitas has not and would not consent
`
`to join in an infringement lawsuit by one business partner, CureVac, against its other business
`
`partner, BioNTech. (See, e.g., D.I. 136 at 7.)
`
`35.
`
`CureVac also alleges that it “discovered that increasing the proportions of
`
`guanosine and cytidine nucleosides in an mRNA stabilizes the molecule and results in an increased
`
`expression of the protein it encodes.” (D.I. 106 at ¶ 19.) This is incorrect. U.S. Patent
`
`No. 11,135,312 (“the ’312 patent”) does not disclose the stabilization of any mRNA or increased
`
`12
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 13 of 123 PageID# 9742
`
`
`
`expression of any protein. Regardless, codon optimization of viral nucleic acid coding sequences
`
`for increasing expression in mammalian cells over the wild type virus was known years before
`
`CureVac’s patent applications were filed. (See, e.g., D.I. 105, Exs. 27-29.)
`
`36.
`
`CureVac’s broad allegations demonstrate that CureVac is not attempting to protect
`
`a specific composition, but rather seeks to preempt others from using prior art methods that were
`
`known and practiced by scientists before CureVac’s applications were filed. (D.I. 106 at ¶ 19.)
`
`CureVac should not be permitted to monopolize an entire class of alleged inventions by claiming
`
`them according to their function (e.g., the stabilization of any mRNA or enhanced expression of
`
`any protein), especially where its patent specification does not even show that claimed function.
`
`37.
`
`CureVac further asserts that it “discovered that modifying one of [mRNA’s]
`
`untranslated regions, composed of sequential adenosine nucleotides at one end of the mRNA (often
`
`called ‘polyA’), results in a substantial increase in the amount of protein expressed by the cell or
`
`organism.” (D.I. 106 at ¶ 21.) But again, this concept of modifying mRNA using “polyA,” as
`
`CureVac attempts to claim in U.S. Patent Nos. 11,149,278 (“the ’278 patent”), 11,286,492 (“the
`
`’492 patent”), and 11,345,920 (“the ’920 patent”) (see D.I. 106 at ¶ 21), is broad and was
`
`previously known in the art (see, e.g. D.I. 104, Ex. 18; D.I. 105, Exs. 30 and 31). Also, upon
`
`information and belief, the U.S. Government has certain rights, including “have made” rights, in
`
`the asserted patents, including at least the ’278, ’492 and ’920, patents.
`
`38.
`
`CureVac also alleges that it “developed new methods to purify the mRNA, as well
`
`as the DNA template which encodes the mRNA, using a technique called Tangential Flow
`
`Filtration (‘TFF’).” (D.I. 106 at ¶ 23.) Once again, this is not true. The prior art reveals that the
`
`use of TFF was known to purify nucleic acids, including plasmid DNA and RNA, before the
`
`applications leading to U.S. Patent Nos. 10,760,070 (“the ’070 patent”) and 11,667,910 (“the ’910
`
`13
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 14 of 123 PageID# 9743
`
`
`
`patent”) were filed. (See, e.g., D.I. 105, Exs. 32-36.) Also, upon information and belief, the U.S.
`
`Government has certain rights, including “have made” rights, in the asserted patents, including at
`
`least the ’070 and ’910 patents.
`
`39.
`
`Upon information and belief, CureVac sought to broaden its patent portfolio
`
`beyond what it actually invented, in an effort to collect and leverage well known and fundamental
`
`research discoveries made by others for its own financial gain.
`
`40.
`
`For these reasons, and as set forth more fully in these Counterclaims and Answer
`
`to Counterclaims, BioNTech and Pfizer deny CureVac’s allegations, deny that CureVac is entitled
`
`to any relief, and seek the relief described below.
`
`PARTIES
`
`41.
`
`BioNTech SE is a company organized and existing under the laws of Germany,
`
`having a principal place of business at An der Goldgrube 12, D-55131 Mainz, Germany.
`
`42.
`
`BioNTech Manufacturing is a company organized and existing under the laws of
`
`Germany, having a principal place of business at An der Goldgrube 12, D-55131 Mainz, Germany.
`
`43.
`
`44.
`
`BioNTech Manufacturing is a wholly owned subsidiary of BioNTech SE.
`
`Pfizer is a corporation organized and existing under the laws of the State of
`
`Delaware, with its principal place of business at 66 Hudson Blvd E, New York, NY 10001-2192,
`
`USA.
`
`45.
`
`Upon information and belief, CureVac SE is a corporation organized and existing
`
`under the laws of Germany, having a place of business at Friedrich-Miescher-Straße 15, 72076
`
`Tübingen, Germany.
`
`46.
`
`Upon information and belief, CureVac Manufacturing GmbH is a corporation
`
`organized and existing under the laws of Germany, having a place of business at Friedrich-
`
`Miescher-Straße 15, 72076 Tübingen, Germany. CureVac Manufacturing GmbH is a
`
`14
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 15 of 123 PageID# 9744
`
`
`
`wholly-owned subsidiary of CureVac N.V.
`
`
`
`
`
`JURISDICTION AND VENUE
`
`47.
`
`BioNTech and Pfizer seek a declaratory judgment pursuant to 28 U.S.C. §§ 2201
`
`and 2202. The Court has jurisdiction over these Counterclaims pursuant to 28 U.S.C. §§ 1331 and
`
`1338(a).
`
`48.
`
`Venue is proper under 28 U.S.C. §§ 1391 and 1400(b), and by CureVac’s choice
`
`of forum.
`
`49.
`
`This action is based upon an actual controversy between the parties arising from
`
`allegations of infringement of the ’312, ’278, ’492, ’920, ’070, ’910, ’493, ’525, ’966, and ’686
`
`patents.
`
`COUNT I - DECLARATION OF INVALIDITY OF THE ’312 PATENT
`
`50.
`
`Pfizer and BioNTech reallege and incorporate by reference paragraphs 1 through
`
`49 of its Counterclaims as if fully set forth herein.
`
`51.
`
`As set forth above, and in particular, in paragraphs 32, 35-36, and 39, the claims of
`
`the ’312 patent are invalid for failure to satisfy one or more of the provisions set forth in 35 U.S.C.
`
`§§ 100 et seq., including, without limitation, the requirements of 35 U.S.C. §§ 102, 103, 112,
`
`and/or any other judicially created requirements for patentability and enforceability of patents,
`
`such as obviousness-type double patenting, and/or in view of the defenses recognized in 35 U.S.C.
`
`§ 282.
`
`52. Without limitation, the claims of the ’312 patent are anticipated and/or obvious
`
`based on prior art, including but not limited to, prior art described above in paragraph 35.
`
`15
`
`

`

`Case 2:23-cv-00222-JKW-DEM Document 252 Filed 03/29/24 Page 16 of 123 PageID# 9745
`
`
`
`53.
`
`In addition, the claims of the ’312 patent are not adequately described, defined,
`
`and/or enabled across the full scope of the claims.
`
`COUNT II - DECLARATION OF INVALIDITY OF THE ’278 PATENT
`
`54.
`
`Pfizer and BioNTech reallege and incorporate by reference paragraphs 1 through
`
`49 of its Counterclaims as if fully set forth herein.
`
`55.
`
`As set forth above, and in particular, in paragraphs 32, 37 and 39, the claims of the
`
`’278 patent are invalid for failure to satisfy one or more of the provisions set forth in 35 U.S.C. §§
`
`100 et seq., including, without limitation, the requirements of 35 U.S.C. §§ 102, 103, 112, and/or
`
`any other judicially created requirements for patentability and enforceability of patents, such as
`
`obviousness-type double patenting, and/or in view of the defenses recognized in 35 U.S.C. § 282.
`
`56. Without limitation, the claims of the ’278 patent are anticipated and/or obvious
`
`based on prior art, including but not limited to, prior art described above in paragraph 37.
`
`57.
`
`In addition, the claims of the ’278 patent are not adequately described, defined,
`
`and/or enabled across the full scope of the claims.
`
`COUNT III - DECLARATION OF INVALIDITY OF THE ’492 PATENT
`
`58.
`
`Pfizer and BioNTech reallege and incorporate by reference paragraphs 1 through
`
`49 of its Counterclaims as if fully set forth herein.
`
`59.
`
`As set forth above, and in particular, in paragrap

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket