Tuesday, August 25, 2020

China Grants Patent Covering the Spike Protein (S Protein) of COVID-19

On August 17, 2020, LAW360 reported that "China has granted CanSino Biologics Inc. and a military scientist the first patent for a COVID-19 vaccine candidate, state media reported Sunday."

Apparently, claim 1 of the patent that was granted is directed to the spike protein (S protein) of COVID-19:

Issued claim: A polynucleotide encoding S protein of 2019 novel coronavirus, characterized in that the sequence of the polynucleotide is shown in SEQ ID NO:1.

The spike protein (S protein) mediates the attachment between the virus and the host cell surface receptors.

If you need additional information on the granted Chinese patent, please contact this law firm in China: czipa@czipa.com


Thursday, April 16, 2020

FEDERAL CIRCUIT PHARMA PATENT SUMMARY


We are open and available for pharma patent work.  Stay safe.

REMBRANDT DIAGNOSTICS, LP v. ALERE, INC. (April 10, 2020)

The plain claim language suggests a broader meaning of “equivalent to” than “equal to.” Claim 1 recites, in relevant part, that the “entry of fluid into the flow control channel creates an ambient pressure within the flow control channel equivalent to the ambient pressure outside of the flow control channel, thereby eliminating a pressure gradient along which excess sample fluid could flow into the flow control channel.” Similar to the oft-used claim term “substantially,” we consider “equivalent,” as used in claim 1, to be a term of degree that does not require mathematical precision.  (“[T]he term ‘substantially’ is a descriptive term commonly used in patent claims to ‘avoid a strict numerical boundary to the specified parameter.’”  Indeed, as Rembrandt correctly observes, the patent law doctrine of equivalents refers to the legal comparison of two elements that are, in fact, different, which supports that “equivalent” is not limited to “equal.” If the applicants meant “equal,” they would have used the word “equal” rather than the broader word “equivalent.” Claim 1 neither uses the term “equal” nor refers to a numerical measurement, numerical comparison between pressures, or even the pressure unit of measurement—pascals.

BASF CORPORATION v. SNF HOLDING COMPANY (April 8, 2020)

In Shimadzu, the Supreme Court explained that the public-use bar applies to uses of the invention “not purposely hidden” and held that the use of a process in the ordinary course of business—where the process was “well known to the employees” and no “efforts were made to conceal” it from anyone else—is a public use. In our public-use cases, we have applied the rule of Shimadzu to assess whether a use was successfully concealed or hidden and therefore inaccessible to the public.  If the use identified by the defendant was not successfully concealed or hidden from those who lack any “limitation or restriction, or injunction of secrecy,” then it is a public use within the meaning of § 102(b). (experimental use is not public use). The parties disagree whether Celanese’s use of the Sanwet® Process meets this standard. The district court cited evidence that Celanese had given tours of the Portsmouth plant to local officials, potential customers, and members of the public and that newspaper articles and a commemoration video depicted the interior of the plant. BASF principally contends that, while certain elements of the claimed process were unquestionably made public—such as the shape of the conical taper—the rest were not, precluding a finding of public use. SNF responds that Celanese failed to fully conceal the process, which qualifies as public use as a matter of law. We conclude that the district court erred in finding, on summary judgment, that the claimed process was publicly accessible. As an initial matter, we disagree with BASF that public use under § 102(b) requires a finding that the public was made aware of every limitation of the claimed process. Our precedent is clear that a factfinder should consider whether certain elements of the claimed invention were already in the public domain at the time of the alleged public-use or on-sale activity. Thus, we have held that § 102(b) “does not require a strict identity between the claimed invention and the device involved in the public use or on sale activities;” it only requires that “the differences between the claimed invention and the device used or sold would have been obvious to one skilled in the art.” In re Smith, 714 F.2d 1127, 1137 n.13 (Fed. Cir. 1983) (citing In re Corcoran, 640 F.2d 1331, 1333 (CCPA 1981)); see also In re Blaisdell, 242 F.2d 779, 783 (CCPA 1957) (finding public use even where some elements of the invention are by their nature concealed from view) (citing Hall v. Macneale, 107 U.S. (17 Otto) 90, 96–97 (1883)).

VALEANT PHARMACEUTICALS INTL. v. MYLAN PHARMACEUTICALS INC. (April 8, 2020)

Claim 1: A stable pharmaceutical preparation comprising a solution of methylnaltrexone or a salt thereof, wherein the preparation comprises a pH between about 3.0 and about 4.0.
Claim 8 recites “[t]he pharmaceutical preparation of claim 1, wherein the preparation is stable to storage for 24 months at about room temperature.”. Notably, claim 8 recites the same preparation as claim 1, but with a newly stated result: 24-month stability.

Mylan cites three prior art references involving different compounds, but each discloses formulations with pH ranges that overlap with the range recited in claim 8, pH between about 3 and about 4. Specifically, Bahal teaches a naloxone composition with a pH of 3 to 3.5, Oshlack teaches a naltrexone composition with a pH of about 3 to about 5 and about 4, and Fawcett discloses a naltrexone formulation with a pH of 3.5 that fell to 3.2 over 90 days. In Mylan’s view, these references establish a prima facie case of obviousness because the pH ranges they teach overlap with those in claim 8. While no reference contemplates methylnaltrexone specifically, Mylan submits that methylnaltrexone bears significant structural and functional similarity to both naloxone and naltrexone such that a person of skill in the art would seek to use prior disclosed pHs for naloxone and naltrexone when formulating solutions of methylnaltrexone. 

Valeant responds that overlapping ranges for different chemical compounds that fail to meet claim 8’s stability requirement do not establish obviousness. According to Valeant, the structural and functional similarities of the compounds are not relevant because claim 8 recites a solution of methylnaltrexone with a stability profile unrecognized and unattained in the prior art. Nevertheless, Valeant submits, methylnaltrexone, naloxone, and naltrexone function differently because of their structural differences, and nothing about the shared function of the drugs is relevant to their stability in solution. 

We agree with Mylan that the record supports a prima facie case of obviousness here. In Peterson, this court recognized that “[a] prima facie case of obviousness typically exists when the ranges of a claimed composition overlap the ranges disclosed in the prior art.” In re Peterson, 315 F.3d 1325, 1329 (Fed. Cir. 2003) (citing In re Geisler, 116 F.3d 1465, 1469 (Fed. Cir. 1997)); In re Woodruff, 919 F.2d 1575, 1578 (CCPA 1990); In re Malagari, 499 F.2d 1297, 1303 (CCPA 1974)). At issue in Peterson was a claim to a nickel-base single-crystal superalloy used in the manufacture of turbine engines. The claimed composition included a relatively small amount of rhenium—about 1 to 3 percent. The prior art of record taught compositions with 0 to 7 percent rhenium, an overlapping range within which the narrower, claimed range fell. We explained that “[s]electing a narrow range from within a somewhat broader range disclosed in a prior art reference is no less obvious than identifying a range that simply overlaps a disclosed range.” Peterson, 315 F.3d at 1329–30. We thus held that the overlapping ranges were sufficient to establish a prima facie case of obviousness, shifting the burden to the patentee to show that the invention would not have been obvious. Here, the pH range recited in claim 8 clearly overlaps with the pH range in the record art, but none of the references disclose the same drug as the one claimed. We are thus presented with the question whether prior art ranges for solutions of structurally and functionally similar compounds that overlap with a claimed range can establish a prima facie case of obviousness. We conclude that they can and, in this case, do.

GENENTECH, INC. v. IANCU (March 30, 2020)

Claim 1. A method for the treatment of a human patient with breast cancer that overexpresses ErbB2 receptor, comprising administering a combination of an antibody that binds ErbB2, a taxoid, and a further growth inhibitory agent to the human patient in an amount effective to extend the time to disease progression in the human patient, wherein the antibody binds to epitope 4D5 within the ErbB2 extracellular domain sequence.

Claim 5. A method for the treatment of a human patient with breast cancer characterized by overexpression of ErbB2 receptor, comprising administering an effective amount of a combination of an anti-ErbB2 antibody which binds epitope 4D5 within the ErbB2 extracellular domain sequence, a taxoid, and a further therapeutic agent, to the human patient.

The claims and specifications of the ’441 and ’549 patents do not clearly define what comparison is required by the disputed claim terms. The examiner recognized as much during prosecution of the ’441 patent, rejecting as indefinite the claim term “extend the time to disease progression.” The examiner found the term to be a relative term undefined by the claim, without “a standard for ascertaining the requisite degree,” and that “one of ordinary skill would not be reasonably apprised of the scope of the invention.”

The examiner inquired:

Specifically, it is never set forth what the extension of time to disease progress is relative to, for example, is the extension of time to disease progress relative to untreated patients? Patients who received antibody or taxoid alone? Patients who received antibody and an anthracycline?

Genentech responded:

[T]he expressions “extend the time to disease progression” and “response rate” are clear from the specification and would be readily understood by a skilled oncologist. Clearly, the combination of an anti-ErbB2 antibody and a taxoid is administered in an amount effective to extend the time to disease progression relative to an untreated patient.

The Board determined that Genentech’s response to the examiner was an express choice, which defined the claim term and led to the issuance of the  Genentech argues that the Board erred in its reliance on this exchange by using it “to override the meaning evident from the specification.” The specification does not, however, expressly define the disputed terms. And Genentech did not view this choice as inconsistent with the specification during prosecution. Instead, Genentech interpreted its own claim language, based on its own specification’s disclosure, as referring to a comparison to untreated patients. The examiner even provided Genentech with the very alternative option (“taxoid alone”) for which Genentech now advocates. Genentech expressly rejected this comparator during prosecution and instead clearly stated that it was effectiveness relative to an untreated patient. Genentech provided an unequivocal, direct response to the examiner’s inquiry—that the term “extend the time to disease progression” was compared to an untreated patient. Genentech’s comparator choice during prosecution of the ’441 patent applies equally to the same claim term that appears in the ’549 patent, which shares a specification and is in the same patent family. Given this relationship between the patents, the Board construed the relevant terms consistently in all six inter partes reviews. We see no error in the Board’s constructions.

GALDERMA LABS., L.P. v. AMNEAL PHARMS. LLC (March 29, 2020)

Claim: An oral pharmaceutical composition of doxycycline, which at a once-daily dosage will give steady state blood levels of doxycycline of a minimum of 0.1 μg/ml and a maximum of 1.0 μg/ml, the composition consisting of (i) an immediate release (IR) portion comprising 30 mg doxycycline; (ii) a delayed release (DR) portion comprising 10 mg doxycycline; and optionally, (iii) one or more pharmaceutically acceptable excipients.
“Immediate release” or IR is defined by the ’patent as “a dosage form that is intended to release substantially all of the active ingredient on administration with no enhanced, delayed or extended release effect.” 
“Delayed release” or DR is not expressly defined. 

We have held that “statements made by a patent owner during an IPR proceeding can be considered during claim construction and relied upon to support a finding of prosecution disclaimer” so long as the statements are “both clear and unmistakable.” 

There is no doubt that the Board rejected the Patent Owner’s attempt to limit the meaning of delayed release. See J.A. 17023 (“[W]e agree with Amneal that the broadest reasonable construction of ‘delayed release,’ in light of the specification of the ’740 patent, is not limited to formulations requiring that there be no substantial release in the stomach.”). Because the Board rejected the Patent Owner’s arguments regarding the meaning of delayed release, the record before the Patent Office clearly put the public on notice that the meaning of delayed release with respect to the Chang Patents is not limited to formulations requiring that there be no substantial release in the stomach. While clear and limiting statements made by the patent owner can give rise to disclaimer, they do not in this case where those statements were clearly and expressly rejected by the Patent Office. Because the record makes clear to a skilled artisan that Patent Owner’s arguments were rejected, those arguments do not impact claim scope. Accordingly, we see no error in the district court’s conclusion that Galderma was not precluded by these statements from asserting the doctrine of equivalents.

KAKEN PHARMACEUTICAL CO., LTD. v. IANCU (March 13, 2020)

Kaken responded by submitting an amendment that cancelled claim 1, clarified the wording of claim 18, and asserted the important difference between mycosis and onychomycosis. Noting that the “[t]reatment of onychomycosis significantly differs from the general treatment of mycoses claimed in ’994,” Kaken explained that “[o]nychomycosis is a condition that specifically affects the nail plate.” Kaken further argued that the “present invention shows the unexpected ability of an azolylamine derivate to penetrate nail and be retained by the nail.”

In allowing the claims, the examiner stated, under the heading “Examiner’s Reasons for Allowance,” that “unexpectedly and in contrast to previously evaluated compositions/methods, the instantly claimed method cures the onychomycosis because the medicament upon direct administration to the nail, penetrates through the nail plate and eradicates the infection at the site.”  


This exchange would leave a skilled artisan with no reasonable uncertainty about the scope of the claim language in the respect at issue here. Kaken is bound by its arguments made to convince the examiner that claims 1 and 2 are patentable. Thus, Kaken’s unambiguous statement that onychomycosis affects the nail plate, and the examiner’s concomitant action based on this statement, make clear that “treating onychomycosis” requires penetrating the nail plate to treat an infection inside the nail plate or in the nail bed under it.

Similarly, the Board rejected Kaken’s objective indicia of non-obviousness because it concluded, relying on the claim construction we have concluded is erroneous, that there is no nexus between the objective indicia and the challenged claims. In its Patent Owner’s response, Kaken identified KP-103 as the “active pharmaceutical ingredient in Jublia®, the first FDA-approved monotherapy for the topical treatment of onychomycosis.”  Kaken made several objective-indicia arguments based on Jublia®: that Jublia® produced unexpected results; that Jublia® has had significant commercial success; that Jublia® received industry praise; and that Jublia® fulfilled a long-felt, but unmet need.  But before considering any of those arguments, the Board pointed to evidence that the FDA approved Jublia® as a “topical treatment of onychomycosis of the toenails,” and Jublia®’s label directs the user to “apply Jublia® to affected toenails once daily.” Because “Jublia® is directed to treatment of specific fungal infections in toenails, and not to a ‘nail’ or to treat ‘onychomycosis,’” the Board explained, the “method for Jublia®’s use is not reasonably commensurate with the [scope] of the challenged claims.” Thus, the Board concluded, the objective indicia presented by Kaken “do not weigh in favor of a finding that the subject matter of the claims would not have been obvious.” I
The foregoing determinations are infected by the erroneous claim construction. In this court, the Director has sought to support the Board’s factual findings with little or no reliance on the claim construction we have held to be erroneous. But that effort is more a reconstruction of the Board’s analysis than a description of the Board’s actual reasoning. We conclude that the appropriate course in this case, as in so many others involving a reversal of a Board claim construction, is to vacate the Board’s decision and remand the matter. 

ILLUMINA, INC. v. ARIOSA DIAGNOSTICS, INC. (March 17, 2020)

Claim 1. A method for preparing a deoxyribonucleic acid (DNA) fraction from a pregnant human female useful for analyzing a genetic locus involved in a fetal chromosomal aberration, comprising: (a) extracting DNA from a substantially cell-free sample of blood plasma or blood serum of a pregnant human female to obtain extracellular circulatory fetal and maternal DNA fragments; (b) producing a fraction of the DNA extracted in (a) by: (i) size discrimination of extracellular circulatory DNA fragments, and  (ii) selectively removing the DNA fragments greater than approximately 500 base pairs, wherein the DNA fraction after (b) comprises a plurality of genetic loci of the extracellular circulatory fetal and maternal DNA; and (c) analyzing a genetic locus in the fraction of DNA produced in (b). ’751 patent col. 7 l. 54–col. 8 l. 57. 1. A method, comprising: (a) extracting DNA comprising maternal and fetal DNA fragments from a substantially cell-free sample of blood plasma or blood serum of a pregnant human female; (b) producing a fraction of the DNA extracted in (a) by: (i) size discrimination of extracellular circulatory fetal and maternal DNA fragments, and (ii) selectively removing the DNA fragments greater than approximately 300 base pairs, wherein the DNA fraction after (b) comprises extracellular circulatory fetal and maternal DNA fragments of approximately 300 base pairs and less and a plurality of genetic loci of the extracellular circulatory fetal and maternal DNA fragments; and (c) analyzing DNA fragments in the fraction of DNA produced in (b).

In Ariosa, we recognized that the inventors had made a discovery with implications that would allow what had previously been discarded as medical waste to be used as a tool for determining fetal characteristics. We acknowledged the profound impact that the discovery had on the field of prenatal medicine, including that it “created an alternative for prenatal diagnosis of fetal DNA that avoids the risks of widely-used techniques that took samples from the fetus or placenta.” Nevertheless, under guidance from the Supreme Court, we determined that the discovery of that natural phenomenon, no matter how significant it was to the medical field, was not itself patentable, and neither was a method for detecting it. The invention in this case is the product of further research on cell-free fetal DNA. This time, the inventors discovered that, not only does the fetal DNA exist in the bloodstream of a pregnant mother, but it has characteristics that make it distinguishable, and therefore separable, from the maternal DNA. Again, regardless how groundbreaking this additional discovery may have been, the inventors were not entitled to patent the natural phenomenon that cell-free fetal DNA tends to be shorter than cell-free maternal DNA. “Groundbreaking, innovative, or even brilliant discovery does not by itself satisfy the § 101 inquiry.”  Thus, they could not claim a method directed to the natural phenomenon, e.g., a method for determining whether a fragment of cell-free DNA is fetal or maternal based on its length. And they did not attempt to patent such a method. The inventors here patented methods of preparing a DNA fraction. The claimed methods utilize the natural phenomenon that the inventors discovered by employing physical process steps to selectively remove larger fragments of cell-free DNA and thus enrich a mixture in cellfree fetal DNA. Though we make no comment on whether the claims at issue will pass muster under challenges based on any other portion of the patent statute, under § 101 the claimed methods are patent-eligible subject matter.

GENENTECH, INC. v. HOSPIRA, INC. (Jan. 10, 2020)

Claim 1: A method of purifying a protein which comprises CH2/CH3 region, comprising subjecting a composition comprising said protein to protein A affinity chromatography at a temperature in the range from about 10°C to about 18°C.

A prior art reference that discloses an overlapping but different range than the claimed range can be anticipatory, even where the prior art range only partially or slightly overlaps with the claimed range. See Ineos USA LLC v. Berry Plastics Corp., 783 F.3d 865, 870–71 (Fed. Cir. 2015) (affirming summary judgment of anticipation of patent claims for composition with “0.05 to 0.5% by weight of at least one saturated fatty acid amide” lubricant in view of a prior art reference disclosing the same class of lubricant in an overlapping range of “0.1 to 5 parts by weight,” and the parties agreed that a measurement in “% by weight” was equivalent to one in “parts by weight”). Once the patent challenger has established, through overlapping ranges, its prima facie case of anticipation, “the court must evaluate whether the patentee has established that the claimed range is critical to the operability of the claimed invention.” Id. at 871; see also E.I. DuPont de Nemours & Co. v. Synvina C.V., 904 F.3d 996, 1008 (Fed. Cir. 2018) (“‘where there is a range disclosed in the prior art, and the claimed invention falls within that range, the burden of production falls upon the patentee to come forward with evidence’ of . . . criticality”) (quoting Galderma Labs., L.P. v. Tolmar, Inc., 737 F.3d 731, 738 (Fed. Cir. 2013)). Here, the Board found that Genentech failed to establish that the claimed temperature range of “about 10°C to about 18°C” is critical to performing protein A chromatography. ’837 Decision, 2018 WL 1187484, at *10–11. Genentech does not challenge the Board’s finding as to criticality, and accordingly, whether or not the claimed temperature range achieves different performance results than WO ’389’s disclosed temperature range is not at issue on appeal. 

Hospira responds that WO ’389 uses the term, “room temperature (18–25°C),” to describe the temperature for conducting protein A affinity chromatography, and all the components involved in that process, including the composition to be purified. According to Hospira’s expert, Dr. Przybycien, in the field of antibody purification, absent contrary language, a skilled artisan would understand that experiments are being conducted at ambient temperature with all materials equilibrated in order to obtain robust scientific data. Based on WO ’389’s disclosure that the composition was cold or frozen after the viral inactivation step, Hospira contends that WO ’389 specifically called out the temperature of the composition when requiring it to be at a temperature other than room temperature. Here, substantial evidence supports the Board’s finding that the HCCF subject to protein A affinity chromatography in WO ’389 is within the claimed temperature range of claim 1. The Board reasoned that it would have been redundant to specifically call out the temperature of the HCCF during protein A affinity chromatography in light of WO ’389’s blanket statement to carry out all steps at “ The Board considered, but disagreed with, Dr. Cramer’s interpretation of WO ’389 as disclosing a process with HCCF that was warmer than “18–25°C” because his opinion was predicated on the view that the ’799 patent is directed to large-scale, industrial processes, which it is not. Id. Further, Dr. Cramer testified that even for large-scale industrial processes, he was not aware of any process where HCCF was applied directly into the chromatography column after being removed from the bioreactor and filtered. Id. at *10 (citing J.A. 1075 at 85:6–15). The Board instead credited Dr. Przybycien’s testimony that no skilled artisan would contact 37°C HCCF to the chromatography column, and report having performed the step at “room temperature (18–25°C)” because using HCCF that was warmer than the chromatography column would raise the temperature of the entire system, making it impossible to conduct “[a]ll steps . . . at room temperature.” To the extent that the experts disagreed with each other, the Board reasonably chose to credit the testimony of Dr. Przybycien over the testimony of Dr. Cramer. 

ELI LILLY AND COMPANY v. HOSPIRA, INC. (August 9, 2019)

Claim 12. An improved method for administering pemetrexed disodium to a patient in need of chemotherapeutic treatment, wherein the improvement comprises: a) administration of between about 350 µg and about 1000 µg of folic acid prior to the first administration of pemetrexed disodium; b) administration of about 500 µg to about 1500 µg of vitamin B12, prior to the first administration of pemetrexed disodium; and c) administration of pemetrexed disodium.

We agree with Hospira. It was clearly erroneous for the district court to hold that the “administration of pemetrexed disodium” step was met because Hospira’s pemetrexed ditromethamine product will be dissolved in saline before administration. A solution of pemetrexed and chloride anions and tromethamine and sodium cations cannot be deemed pemetrexed disodium simply because some assortment of the ions in the solution consists of pemetrexed and two sodium cations. As Lilly acknowledges throughout its brief, pemetrexed disodium is a salt. (pemetrexed toxicity is caused “by pemetrexed itself once dissociated in solution,” not pemetrexed disodium); see also Hospira J.A. 1596 (October 2017 Alimta® Label referring to the drug substance as the “disodium salt” of pemetrexed). Once diluted, the salt’s crystalline structure dissolves, and the individual ions dissociate. See Hospira J.A. 2820 (declaration of Lilly’s expert). In other words, pemetrexed disodium no longer exists once dissolved in solution, and, as a corollary, a different salt of pemetrexed dissolved in saline is not pemetrexed disodium.

We conclude that to literally practice the “administration of pemetrexed disodium” step under the district court’s claim construction, the pemetrexed disodium salt must be itself administered.  (“‘[A]dministration of pemetrexed disodium’ . . . refer[s] to a liquid administration of pemetrexed disodium. . . ., accomplished by dissolving the solid compound pemetrexed disodium into solution . . . .”); see also Tex. Instruments Inc. v. Cypress Semiconductor Corp., 90 F.3d 1558, 1563 (Fed. Cir. 1996) (“To literally infringe, the accused . . . process must contain every limitation of the asserted claim.” (citing Laitram Corp. v. Rexnord, Inc., 939 F.2d 1533, 1535 (Fed. Cir. 1991))). There is no dispute that Hospira has only sought approval to market pemetrexed ditromethamine,  and that neither its proposed product nor methods of administering it will constitute administering the pemetrexed disodium salt. Accordingly, Hospira will not practice the step of “administration of pemetrexed disodium,” and the district court’s finding of literal infringement must be reversed.

Doctrine of equivalents
Prosecution History Estoppel

But here, we conclude that this consideration is not dispositive because the rest of the prosecution history, and the ’209 patent itself, show that it is implausible that the reason for Lilly’s amendment was to surrender other pemetrexed salts. Indeed, such a relinquishment would effectively dedicate the entirety of Lilly’s invention to the public and thereby render the ’209 patent worthless, and it would have been irrelevant for distinguishing the prior art. Again, the prosecution history strongly indicates a less sweeping and more sensible reason for Lilly’s amendment: to surrender antifolates other than pemetrexed. Thus, we conclude on this prosecution record that Lilly’s amendment was merely tangential to pemetrexed ditromethamine.

Disclosure-Dedication Rule

We are unpersuaded by DRL’s arguments. As the district court noted, Akimoto’s formula, includes seven functional group variables and encompasses thousands of compounds, and while Akimoto discloses about fifty exemplary compounds, none of them is pemetrexed. Moreover, Akimoto does not even disclose tromethamine expressly but only generically among dozens of other salts. At most, Akimoto discloses ammonium salts generally, which is far from a description of tromethamine. In similar circumstances, we have held that “sufficient description of a genus” requires that a skilled artisan be able to “‘visualize or recognize’ the members of the genus.” See Ariad Pharm., Inc. v. Eli Lilly & Co., 598 F.3d 1336, 1350 (Fed. Cir. 2010) (quoting Regents of the Univ. of Cal. v. Eli Lilly & Co., 119 F.3d 1559, 1568–69 (Fed. Cir. 1997)). Akimoto does not so describe pemetrexed ditromethamine, and we see no reason why a skilled artisan would set out on DRL’s winding path to cobble together pemetrexed ditromethamine. While the ’209 patent teaches that pemetrexed disodium is the “most preferred” antifolate, that knowledge would not change the skilled artisan’s understanding of what Akimoto discloses. Because Akimoto contains only a “generic reference” to pemetrexed ditromethamine, PSC Comput., 355 F.3d at 1360, we conclude that it was not dedicated to the public.

SANOFI-AVENTIS U.S., LLC v. FRESENIUS KABI USA, LLC (August 9, 2019)

Even assuming there was some general motivation to make docetaxel more lipophilic to combat drug resistance, the district court also did not clearly err in finding that Fresenius failed to establish a motivation to do so by specifically making simultaneous methoxy substitutions at C7 and C10. The court found that taxane researchers investigated substitutions at many positions, and the voluminous references in this case support that finding. For example, Commerçon disclosed that C3′, C7, C9, and C10, and to a more limited extent C2′, were modifiable. And as summarized above, the other references investigated a diverse set of substitutions. Fresenius reads this panoply of teachings as rendering obvious simultaneous C7 and C10 methoxy substitutions. But despite the apparent interest in taxane analogs, not a single reference relied on by Fresenius made simultaneous substitutions of any kind at C7 and C10. And of the references that made individual methoxy substitutions at C7 or C10, none tested those taxane analogs against drug resistant cell lines or taught that the analogs would overcome drug resistance. On this record, the court did not clearly err in finding no motivation to make C7 and C10 methoxy substitutions on docetaxel to improve its activity against drug-resistant cancers.

Considering Fresenius’s reference-specific arguments, we agree with the district court that they are emblematic of hindsight reasoning. Fresenius contends that Commerçon would have pointed a skilled artisan towards C7, C10, and (less desirably) C9 substitutions because those positions were “flexible,” and away from C2′ and C3′ substitutions because those positions were “crucial.” CrossAppellants’ Br. 57–58. However, this argument plainly mischaracterizes the reference. Commerçon expressly identified the sidechain position C3′ as “flexible,” and indicated that C2′ could be modified with certain substitutions if the configuration was maintained. 

PHIGENIX, INC. v. GENENTECH, INC. (Sept. 5, 2019)

Claim 1 recites “[a] method for treating a breast condition” by administering a composition that inhibits PAX2 expression or activity, and/or expresses DEFB1.

In its complaint, Phigenix alleged that Genentech induced infringement of the ’534 patent by encouraging health care professionals to prescribe and administer Kadcyla to breast cancer patients who had previously received the chemotherapy drugs “trastuzumab and a taxane, separately or in combination.”

In February 2017, the district court issued a summary judgment order holding that the asserted claims of the ’534 patent are not entitled to the priority date of the 2005 provisional application. Phigenix did not move to amend its infringement contentions in response. Several months later—after fact discovery had closed and expert reports had been exchanged—Phigenix narrowed the relevant population to Kadcyla patients who were pretreated exclusively with trastuzumab and a taxane—i.e., trastuzumab, a taxane, and “and nothing else.” J.A. 317–19. Phigenix did not do so by affirmatively moving for leave to amend its infringement contentions. Instead, Phigenix first revealed its narrowed definition during the deposition of its expert on May 23, 2017, and only in response to questioning by Genentech. Phigenix does not dispute that the narrowed population of relevant patients comprises only about 4% of the total population of Kadcyla patients. Yet at no point did Phigenix move to amend its infringement contentions to reflect this narrower population.

After learning about the narrowed patient population, Genentech moved to strike the infringement opinion of Phigenix’s expert. Agreeing that Phigenix had failed to provide adequate notice of its narrowed infringement theory, the district court struck Phigenix’s expert infringement opinion and granted summary judgment of noninfringement based on the resulting lack of direct infringement evidence. The district court specifically noted that if its determination regarding the 2005 priority date was the impetus for Phigenix’s narrowed infringement theory, then “Phigenix could have moved soon thereafter to amend its infringement contentions and to notify Genentech of this change in position.” J.A. 16. Phigenix’s failure to do so “deprived Genentech [of] a timely disclosure of this new theory, as well as any potential accommodation in the case schedule the Court would entertain.” 

Phigenix first argues that its revised infringement theory is not newly propounded because it falls wholly within the scope of its original infringement contentions. While technically accurate, this argument fails to acknowledge the notice function served by infringement contentions. Patent local rules bolster discovery under the Federal Rules because they “allow the defendant to pin down the plaintiff’s theories of liability . . . thus confining discovery and trial preparation to information that is pertinent to the theories of the case.”  The Northern District’s patent local rules, furthermore, expressly require that the identification of the accused methods in infringement contentions “shall be as specific as possible.” N.D. Cal. Patent L.R. 3-1(b). We agree with the district court that the broader theory disclosed in Phigenix’s infringement contentions is not sufficiently specific to disclose its narrowed theory. If, as Phigenix suggests, plaintiffs could include broadly scoped infringement theories in their contentions only to unilaterally narrow them after the close of fact discovery, infringement contentions would provide little relief to defendants.

Finally, Phigenix argues that striking Phigenix’s entire expert infringement opinion is an abuse of discretion because it is (at least in Phigenix’s view) heavy-handed. In support, Phigenix avers that Genentech has not adequately explained how it was prejudiced. Phigenix further argues that Genentech fails to explain why a less severe sanction would not address any concern the district court had. Phigenix even boldly asserts that Genentech was obligated to seek clarification if it was confused by the scope of Phigenix’s infringement contentions.

As a threshold matter, we need not consider the prejudice to Genentech in evaluating whether the court abused its discretion. See O2 Micro, 467 F.3d at 1368 (“Having concluded that the district court could properly conclude that [the plaintiff] did not act diligently in moving to amend its infringement contentions, we see no need to consider the question of prejudice to [the defendant].”). In any event, Genentech adequately explained how it was prejudiced by Phigenix’s untimely narrowing of its infringement theory.

The consequences imposed on Phigenix, furthermore, are not beyond the discretion of the district court. Both the Ninth Circuit and this court have concluded that the exclusion of evidence is often an appropriate sanction for a party’s failure to comply with the patent local rules. O2 Micro, 467 F.3d at 1369 (first citing SanDisk, 415 F.3d at 1292; then citing Wong v. Regents of Univ. of Cal., 410 F.3d 1052, 1060 (9th Cir. 2005)). Thus, even if other courts might have chosen a less potent remedy in these circumstances, we assess only whether the district court abused its discretion in prescribing a harsher one. On these facts, we conclude that it did not.

ALLERGAN SALES, LLC v. SANDOZ, INC. (August 29, 2019)

Claim: A method of treating a patient with glaucoma or ocular hypertension comprising topically administering twice daily to an affected eye a single composition comprising 0.2% w/v brimonidine tartrate and 0.68% w/v timolol maleate, wherein the method is as effective as the administration of 0.2% w/v brimonidine tartrate monotherapy three times per day and wherein the method reduces the incidence of one o[r] more adverse events selected from the group consisting of conjunctival hyperemia, oral dryness, eye pruritus, allergic conjunctivitis, foreign body sensation, conjunctival folliculosis, and somnolence when compared to the administration of 0.2% w/v brimonidine tartrate monotherapy three times daily.

We are not persuaded by Sandoz’s primary counterarguments, including Sandoz’s reliance on Bristol–Myers, 246 F.3d 1368, In re Copaxone Consolid. Cases, 906 F.3d 1013 (Fed. Cir. 2018), and Georgetown Rail Equip. Co. v. Holland, L.P., 867 F.3d 1229 (Fed. Cir. 2017). In Bristol–Myers we expressly noted that the disputed claim terms “w[ere] voluntarily made after the examiner had already indicated . . . the claims were allowable” and such “unsolicited assertions of patentability made during prosecution do not create a material claim limitation.” 246 F.3d at 1375. Likewise, in Copaxone we held that the disputed claim terms were not “necessary or relevant to the examiner’s approval.” 906 F.3d at 1024. Finally, in Georgetown Rail we explained that the disputed claim terms were not relied upon during prosecution to “distinguish the patented invention from the prior art.” 867 F.3d at 1238. Here, however, both Allergan and the Examiner explicitly relied on the “wherein” clauses to distinguish the claimed methods over the prior art during prosecution. The “wherein” clauses were neither unnecessary nor irrelevant, but were instead material to the Examiner’s patentability determination.

AMGEN INC. v. COHERUS BIOSCIENCES INC. (July 29, 2019)

Claim 1: A process for purifying a protein on a hydrophobic interaction chromatography column such that the dynamic capacity of the column is increased for the protein comprising mixing a preparation containing the protein with a combination of a first salt and a second salt, loading the mixture onto a hydrophobic interaction chromatography column, and eluting the protein, wherein the first and second salts are selected from the group consisting of citrate and sulfate, citrate and acetate, and sulfate and acetate, respectively, and wherein the concentration of each of the first salt and the second salt the mixture is between about 0.1 M and about 1.0.

We hold that argument-based prosecution history estoppel applies here because Amgen clearly and unmistakably surrendered unclaimed salt combinations during prosecution. In its January 6, 2011 response, Amgen distinguished Holtz on the basis that Holtz did not teach or suggest the “particular combinations of salts” recited in Amgen’s claims. Indeed, Amgen emphasized “particular” and referred to its particular salts three times in the span of two pages. The Declaration attached to Amgen’s response also highlights and discusses the same particular combinations recited in Amgen’s claims. For example, the Declaration refers to sulfate/citrate and sulfate/acetate as “particular dual salt combination[s]” that resulted in increased dynamic capacity as compared to a single salt.  It also explains that using a sulfate/citrate, sulfate/acetate, or acetate/citrate 10 combination (the only claimed combinations) resulted in reduced commercial manufacturing costs as compared to using only a single salt. Notably, Amgen’s response to the examiner’s office action quotes the Declaration’s conclusion that “[u]se of this particular combination of salts greatly improves the cost-effectiveness of commercial manufacturing by reducing the number of cycles required for each harvest and reducing the processing time for each harvest.” . Amgen’s response and Declaration do not mention any salt combinations other than those claimed. Based on Amgen’s statements during prosecution, we agree with the district court’s conclusion that “a competitor would reasonably believe” that Amgen surrendered unclaimed salt combinations. 

Monday, January 28, 2019

Genentech's Herceptin Patents Asserted Against Samsung Bioepis

Genentech and the City of Hope filed a patent infringement lawsuit against Samsung Bioepis over Bioepis’ biosimilar version of Herceptin.  (1:18-cv-01363-CFC).  The plaintiffs allege that “Bioepis did not disclose all of the information relevant to establishing whether the manufacture of Bioepis’s aBLA product will infringe each of the patents identified on Genentech’s Apr. 23, 2018 list, despite Genentech’s request that Bioepis provide sufficient ‘other information that describes the process or processes used to manufacture” as required by 42 U.S.C. § 262(l)(A).  Bioepis’s failure to provide sufficient information under those circumstances supports Genentech’s contention that manufacturing Bioepis’s aBLA product will infringe such patents.’”  The plaintiffs assert the following patents in the first amended complaint filed on January 17, 2019:

Method for Making Humanized Antibodies
6,407,213
1. A humanized antibody variable domain comprising non-human Complementarity Determining Region (CDR) amino acid residues which bind an antigen incorporated into a human antibody variable domain, and further comprising a Framework Region (FR) amino acid substitution at a site selected from the group consisting of: 4L, 38L, 43L, 44L, 58L, 62L, 65L, 66L, 67L, 68L, 69L, 73L, 85L, 98L, 2H, 4H, 36H, 39H, 43H, 45H, 69H, 70H, 74H, and 92H, utilizing the numbering system set forth in Kabat.
The Combination Chemotherapy Patents
7,846,441
1. A method for the treatment of a human patient with a malignant progressing tumor or cancer characterized by overexpression of ErbB2 receptor, comprising administering a combination of an intact antibody which binds to epitope 4D5 within the ErbB2 extracellular domain sequence and a taxoid, in the absence of an anthracycline derivative, to the human patient in an amount effective to extend the time to disease progression in said human patient, without increase in overall severe adverse events. 
7,892,549
1. A method for the treatment of a human patient with breast cancer that overexpresses ErbB2 receptor, comprising administering a combination of an antibody that binds ErbB2, a taxoid, and a further growth inhibitory agent to the human patient in an amount effective to extend the time to disease progression in the human patient, wherein the antibody binds to epitope 4D5 within the ErbB2 extracellular domain sequence.
The Cabilly Patents
6,331,415
1. A process for producing an immunoglobulin molecule or an immunologically functional immunoglobulin fragment comprising at least the variable domains of the immunoglobulin heavy and light chains, in a single host cell, comprising the steps of:
(i) transforming said single host cell with a first DNA sequence encoding at least the variable domain of the immunoglobulin heavy chain and a second DNA sequence encoding at least the variable domain of the immunoglobulin light chain, and
(ii) independently expressing said first DNA sequence and said second DNA sequence so that said immunoglobulin heavy and light chains are produced as separate molecules in said transformed single host cell.
7,923,221
1. A method for making an antibody heavy chain or fragment thereof and an antibody light chain or fragment thereof each having specificity for a desired antigen, wherein the heavy chain or fragment thereof comprises a human constant region sequence and a variable region comprising non human mammalian variable region sequences, the method comprising culturing a recombinant host cell comprising DNA encoding the heavy chain or fragment thereof and the light chain or fragment thereof and recovering the heavy chain or fragment thereof and light chain or fragment thereof from the host cell culture.
The Method of Administration Patents
6,627,196
1. A method for the treatment of a human patient diagnosed with cancer characterized by overexpression of ErbB2 receptor, comprising administering an effective amount of an anti-ErbB2 antibody to the human patient, the method comprising: administering to the patient an initial dose of at least approximately 5 mg/kg of the anti-ErbB2 antibody; and administering to the patient a plurality of subsequent doses of the antibody in an amount that is approximately the same or less than the initial dose, wherein the subsequent doses are separated in time from each other by at least two weeks. 
7,371,379
1. A method for the treatment of a human patient diagnosed with cancer characterized by overexpression of ErbB2 receptor, comprising administering an effective amount of an anti-ErbB2 antibody to the human patient, the method comprising: administering to the patient an initial dose of at least approximately 5 mg/kg of the anti-ErbB2 antibody; and administering to the patient a plurality of subsequent doses of the antibody in an amount that is approximately the same or less than the initial dose, wherein the subsequent doses are separated in time from each other by at least two weeks; and further comprising administering an effective amount of a chemotherapeutic agent to the patient. 
10,160,811
1. A method for the treatment of a human patient diagnosed with breast cancer characterized by 2+ or 3+ overexpression of ErbB2 receptor as determined by immunohistochemistry or fluorescence in situ hybridization (FISH), comprising the steps of administering to the patient an initial dose of 8 mg/kg of anti-ErbB2 huMAb 4D5-8 antibody; and administering to the patient a plurality of subsequent doses of 6 mg/kg of the antibody, wherein all doses are separated in time from each other by three weeks.
The Acidic Variants Patents
6,339,142
1. A composition comprising a mixture of anti-HER2 antibody and one or more acidic variants thereof, wherein the amount of the acidic variant(s) is less than about 25%.
6,417,335
1. A method for purifying an antibody from a composition comprising the antibody and a contaminant, which method comprises:
(a) loading the composition onto a cation exchange resin, wherein the amount of antibody loaded onto the cation exchange resin is from about 20 mg to about 35 mg of the antibody per mL of cation exchange resin; and

(b) eluting the antibody from the cation exchange resin.
9,249,218
1. A therapeutic composition comprising a mixture of anti-HER2 antibody and one or more acidic variants thereof, wherein the amount of the acidic variant(s) is less than about 25%, and wherein the acidic variant(s) are predominantly deamidated variants wherein one or more asparagine residues of the anti-HER2 antibody have been deamidated, and wherein the anti-HER2 antibody is humMAb4D5-8, and wherein the deamidated variants have Asn30 in CDR1 of either or both V.sub.L regions of humMAb4D5-8 converted to aspartate, and a pharmaceutically acceptable carrier.
HER2 Diagnostic Patents
7,993,834
1. A method for increasing likelihood of effectiveness of breast cancer treatment with humanized anti-ErbB2 antibody huMAb4D5-8, which method comprises administering a cancer treating dose of said antibody to a human subject diagnosed with breast cancer, wherein an erbB2 gene amplification in breast cancer cells in a tissue sample from the subject has been detected, and wherein the breast cancer cells from the human subject have been found to have a 0 or 1+ score of ErbB2 protein expression by immunohistochemistry.
8,076,066
1. A method of identifying and treating a breast cancer patient disposed to respond favorably to a HER2 antibody, huMAb4D5-8, which method comprises detecting her2 gene amplification in cancer cells in a breast tissue sample from the patient and treating the patient with her2 gene amplification with the HER2 antibody in an amount effective to treat the breast cancer, wherein the patient's cancer cells express HER2 at a 0 or 1+ level by immunohistochemistry. 
8,440,402
1. A method for identifying and treating a patient disposed to respond favorably to a HER2 antibody, huMAb4D5-8, for treating breast cancer, which method comprises detecting her2 gene amplification in tumor cells in a tissue sample from the patient and treating the patient with her2 gene amplification with the HER2 antibody in an amount effective to treat the breast cancer, wherein the patient's tumor cells express HER2 at a 0 or 1+ level by immunohistochemistry.
Cell Culture, Purification, and Antibody Manufacturing Patents
6,610,516
1. A process for producing a human glycoprotein having multiple glycoforms, comprising culturing Chinese hamster ovary cells expressing nucleic acid encoding said glycoprotein in a serum-free medium in a production phase at a temperature of about 30C. to 35C. and in the presence of about 0 to 2 mM of a butyrate salt, wherein the process produces an increased percentage of glycoprotein molecules having one glycoform relative to an identical process performed at 37C. in the absence of butyrate.
7,390,660
1. A method for reducing glucose consumption during cultivation of CHO, myeloma, or hybridoma cells, comprising cultivating CHO, myeloma, or hybridoma cells in culture medium in the presence of citric acid or citrate wherein said citric acid or citrate is maintained at a concentration of about 1 to 50 mmol/l during cultivation and wherein said citric acid or citrate is not bound in a chelate complex with iron or another transition metal ion.
7,485,704
1. A method of purifying a protein which comprises a CH2/CH3 region comprising: a. subjecting a composition comprising said protein to protein A affinity chromatography to provide a recovered composition and measuring leached protein A in said recovered composition; and b. if greater than about 20 ng protein A per mg of said protein is measured in said recovered composition, then performing subsequent purification of compositions comprising said protein by protein A affinity chromatography at a temperature in the range from about 3C. to about 18C., such that protein A leaching is reduced.
7,807,799
1. A method of purifying a protein which comprises a CH2/CH3 region, comprising subjecting a composition comprising said protein to protein A affinity chromatography at a temperature in the range from about 10C. to about 18C.
8,512,983
1. A process for producing a polypeptide in a mammalian host cell expressing said polypeptide, comprising culturing the mammalian host cell in a production phase of the culture in a glutamine-free production culture medium containing asparagine, wherein the asparagine is added at a concentration in the range of 7.5 mM to 15 mM.
8,574,869
1. A method for the prevention of the reduction of a disulfide bond in an antibody expressed in a recombinant host cell, comprising, following fermentation, sparging the pre-harvest or harvested culture fluid of said recombinant host cell with air, wherein the amount of dissolved oxygen (dO2) in the pre-harvest or harvested culture fluid is at least 10%.
9,714,293
1. A process for producing a polypeptide in a host cell expressing said polypeptide, comprising culturing the host cell in a production phase of the culture in a glutamine-free production culture medium containing asparagine and aspartic acid, wherein the asparagine is added at a concentration in the range of 7.5 mM to 15 mM and wherein the aspartic acid is added at a concentration in the range of 1 mM to 10 mM.



Wednesday, January 23, 2019

Patent term adjustment went beyond the period during which the applicant failed to undertake reasonable efforts

SUPERNUS PHARMACEUTICALS, INC v. IANCU

A plain reading of the statute shows that Congress imposed two limitations on the amount of time that the USPTO can use as applicant delay for purposes of reducing PTA. First, the statute expressly requires that any reduction to PTA be equal to the period of time during which an applicant fails to engage in reasonable efforts. Second, the statute expressly ties reduction of the PTA to the specific time period during which the applicant failed to engage in reasonable efforts.

The PTA statute requires that any PTA reduction be equal to the period of time during which an applicant fails to engage in reasonable efforts. The word “equal” is widely understood to mean “the same in amount, number, or size.” E.g., Equal, Cambridge Dictionary of American English 287 (2000). Stated differently, PTA reduction cannot exceed the period of time during which an applicant failed to engage in reasonable efforts. Thus, if there is no period of time during which the applicant could have but failed to engage in reasonable efforts, there can be no reduction to the PTA.





http://www.cafc.uscourts.gov/sites/default/files/opinions-orders/17-1357.Opinion.1-23-2019.pdf

Tuesday, January 22, 2019

Supreme Court: AIA Did Not Change Meaning Of On-Sale Bar

HELSINN HEALTHCARE S. A. v. TEVA PHARMACEUTICALS USA, INC.

Every patent statute since 1836 has included an on-sale bar. The patent statute in force immediately before the AIA prevented a person from receiving a patent if, “more than one year prior to the date of the application for patent in the United States,” “the invention was . . . on sale” in the United States. 35 U. S. C. §102(b) (2006 ed., Supp. IV). The AIA, as relevant here, retained the on-sale bar and added the catchall phrase “or otherwise available to the public.” §102(a)(1) (2012 ed.) (“A person shall be entitled to a patent unless” the “claimed invention was . . . in public use, on sale, or otherwise available to the public . . . ”). We must decide whether these changes altered the meaning of the “on sale” bar. We hold that they did not.

https://www.supremecourt.gov/opinions/18pdf/17-1229_2co3.pdf

Monday, January 21, 2019

A generic pharma company had standing to appeal PTAB's decision in an IPR

Federal Circuit:

We agree with Amerigen that it has standing to appeal from the Board’s decision because the launch of its tentatively approved drug is blocked by the ’650 patent, and invalidation of the patent would advance its drug’s launch. The ’650 patent is listed in the FDA’s “Orange Book”12 entry for Toviaz®. Amerigen has a Paragraph III certification for the ’650 patent,13 which means that the FDA will only approve Amerigen’s ANDA after the ’650 patent has expired. 21 U.S.C. § 355(j)(5)(B)(ii). However, if the ’650 patent is held unpatentable through reversal of the Board’s decision, then the New Drug Application (“NDA”) holder14 must “promptly notify” the FDA that the patent “no longer meet[s] the statutory requirements for listing.” 21 C.F.R. § 314.53(f)(2)(i). And § 314.53 expressly states that a patent does not meet the requirements for listing “if there has been a judicial finding of invalidity for a listed patent, from which no appeal has been or can be taken.” Id. After a notification from the NDA holder that a patent may no longer be listed, the FDA “will remove a patent . . . from the list if there is no first applicant eligible for 180–day exclusivity based on a paragraph IV certification to that patent or after the 180–day exclusivity period of a first applicant based on that patent has expired or has been extinguished.” Id.


http://www.cafc.uscourts.gov/sites/default/files/opinions-orders/17-2596.Opinion.1-11-2019.pdf