Tuesday, February 28, 2017

GENENTECH’S PATENT TO A METHOD FOR INCREASING YIELD SURVIVES IPR PETITION LACKING OBVIOUSNESS POSITION

           The PTAB (Patent Trial and Appeal Board) denied a petition to institute an Inter Partes Review (IPR2016-01608) against Genentech’s U.S. Patent No. 6,716,602, which claims a method for increasing the product yield of a properly folded polypeptide of interest produced by recombinant host cells:

1. A method for increasing the product yield of a properly folded polypeptide of interest produced by recombinant host cells, wherein expression of the polypeptide by the recombinant host cells is regulated by an inducible system, which method comprises
(a) culturing the recombinant host cells under conditions of high metabolic growth rate; and
(b) reducing the metabolic rate of the cultured recombinant host cells at the time of induction of polypeptide expression, wherein reducing the metabolic rate comprises reducing the feed rate of a carbon/energy source, or reducing the amount of available oxygen, or both, and wherein the reduction in metabolic rate result in increase yield of properly folded polypeptide.

The Petitioner (bioeq IP AG) relied on Seeger (17 Enzyme & Microbial Tech. 947–53 (1995)), in which cells were grown at 30°C in a fed-batch procedure, with a predetermined exponential feeding rate to ensure constant specific growth rates. Seeger explains that product formation was induced by shifting either the temperature from 30°C to 42°C, or by adding isopropyl-ß-D-thiogalacto-pyranoside (IPTG). Seeger observes acetic acid accumulation in response to temperature-induced product expression. To prevent that accumulation, the exponential feeding rate was reduced  after the temperature shift to 42°C.  Seeger determined that the temperature-induced production of bFGF (basic fibroblast growth factor)  “generated more total and more soluble bFGF compared to IPTG-induced cultures.”

            The PTAB disagreed that Seeger anticipated claim 1 since it did not teach reduction of metabolic rate.  In particular, “Petitioner relies upon the disclosure of the ’602 patent describing reducing metabolic rate in cells already in a reduced growth state by reducing the rates of oxygen uptake and the corresponding rates of uptake of a carbon/energy source. However, Petitioner has not addressed adequately how Seeger’s method of inducing expression by increasing temperature from 30 to 42°C may have affected the metabolic rate.” 

            Petitioner relied only on Seeger for anticipation of claim 1, and did not have a back-up obviousness position.  

Thursday, February 23, 2017

Dr. Reddy’s Infringes Aloxi (palonosetron) Patents

            On Feb. 14, 2017, a New Jersey Federal judge[1] found that Dr. Reddy’s proposed generic infringed the following three patents (US Patent 7,947,724, US Patent 8,729,094, and US Patent 9,066,980) covering the anti-nausea drug Aloxi.  This case arose from Dr. Reddy’s submission of a so-called “Paper NDA” under § 505(b)(2). There is separate and ongoing litigation regarding various Abbreviated New Drug Applications filed under 21 U.S.C. § 505(j) to manufacture generic versions of Helsinn’s branded palonosetron product.

            An example of an asserted claim is claim 1 of US Patent 9,066,980 , to which Dr. Reddy’s only raised an invalidity defense:

1. A pharmaceutical single-use, unit-dose formulation for intravenous administration to a human to reduce the likelihood of cancer chemotherapy-induced nausea and vomiting, comprising a 5 mL sterile aqueous solution, said solution comprising: a) palonosetron hydrochloride in an amount of 0.25 mg based on the weight of its free base, b) optionally a chelating agent, and c) a tonicifying agent in an amount sufficient to make said solution isotonic, wherein said formulation is stable at 24 months when stored at room temperature. 

The claims and defenses in the case were as follows:

’724 9 Non-Infringement
’094 22 Invalidity
’094 23 Invalidity
’094 24 Invalidity
’094 25 Invalidity
’094 27 Non-Infringement
’980 1 Invalidity
’980 2 Invalidity
’980 3 Invalidity
’980 4 Invalidity
’980 5 Invalidity
’980 6 Non-Infringement
’980 16 Invalidity

Below are excerpts from the Order on infringement and invalidity. 

Infringement
            A ligand is a molecule that can bind to a metal ion. (Dkt. 175 at 55.) Ligands can form different types of bonds with metal ions, such as unidentate, multidentate, and bridging bonds. (Id. at 52–53.) A multidentate ligand is a molecule that has at least two atoms which can simultaneously make a bond with a metal ion. (Id. at 55.) Bidentate bonds are a type of multidentate bond consisting of two bonds between a ligand and a single metal ion, forming a “chelate” ring structure. (Id. at 53.) A particular molecule may form a chelate bond in certain circumstances, but not others. (Dkt. 178 at 126.)
            DRL’s Accused Product contains sodium acetate trihydrate. When placed in aqueous solution, sodium acetate trihydrate dissociates into sodium, acetate, and water. (Dkt. 175 at 51.) Acetate is capable of forming a pair of simultaneous bonds with metal ions. (Id. at 61– 63.) Specifically, under certain circumstances, the two oxygen atoms in acetate are capable of simultaneously binding to a metal ion to form a four-membered chelate ring. (Id.)
            Numerous scholarly references from peer-reviewed journals support the conclusion that acetate can under some circumstances act as a multidentate ligand capable of forming chelate ring structures. See, e.g.: Ishioka (PTX-260); Sakohara (PTX-273); Bryant (PTX- 274); Schürmann (PTX-231); Weber (PTX-216); Favas (PTX-221); Martell (PTX-280); Jia (PTX-230); Deepa (PTX-228); Warthen (PTX-226); Li (PTX-264); Quilès (PTX-236); Jiang (PTX-272); Feldman (PTX-223); and Kakihana (PTX-237). These articles demonstrate that acetate can form chelate rings with metal ions under various experimental conditions, including in a variety of solvents and temperatures. Acetate-metal chelate rings have been demonstrated through a variety of experimental detection methods, including x-ray crystallography, extended x-ray absorption fine structure spectroscopy, infrared spectroscopy, and Raman spectroscopy. Further, these chelate rings were detected in various applications of acetate and with a variety of metal ions, including: (1) zinc chelation used in insulin formulations; (2) lead chelation used in treatment of lead poisoning; (3) iron chelation used in fertilizers); (4) gadolinium chelation used in medical imaging dyes; (5) lanthanum chelation used in medical imaging dyes; (6) ruthenium chelation used in industrial processes; (7) tungsten chelation used in preparing electrochromic films; and (8) copper chelation used in fertilizers and for contaminated pharmaceuticals. That acetate is a multidentate ligand capable of forming a ring structure with a metal ion under certain circumstances is further supported by expert testimony from both Helsinn and DRL.
            DRL submitted some evidence that acetate under some circumstances forms unidentate bonds. At best, we find that this evidence supports only the conclusion that acetate forms unidentate bonds in certain circumstances while forming multidentate bonds in other circumstances.
            We construed the term “chelating agent” to mean a “multidentate ligand that can form a ring structure by reacting with a metal ion.” Under the second step of the infringement analysis, Helsinn needed to prove by a preponderance of the evidence that DRL’s accused palonosetron product contains a chelating agent under our claim construction. Based on a review of all of the evidence presented at trial, and as reflected in the findings of fact above, we conclude that Helsinn proved by a preponderance of the evidence that the acetate present in DRL’s product is a multidentate ligand that can form a ring structure by reacting with a metal ion. Accordingly, the Accused Product contains a “chelating agent,” which the parties have stipulated is the determinative issue to establish infringement here. We therefore find that DRL’s Accused Product infringes claim 9 of the ’724 patent, claim 27 of the ’094 patent, and claim 6 of the ’980 patent.

Enablement
            Weighing the relevant Wands factors, we conclude that the totality of this evidence does not support a finding that undue experimentation would be necessary to practice the asserted claims. See Wands, 858 F.2d at 737. With respect to factor 4, the nature of the invention requires shelf-life stability at 18 months (claim 16 of the ’980 patent) and 24 months (claims 22, 23, 24, and 25 of the ’094 patent and claims 1, 2, 3, 4, and 5 of the ’980 patent). The asserted claims are broad in scope as they cover a range of formulations (factor 8). But the ’351 application and Example 4 provide sufficient direction and guidance to teach a POSA to practice the full scope of the formulations (factors 2 and 3). The ’351 application discloses stable 0.05 mg/mL palonosetron formulations. The ’351 application teaches that formulations with this optimal palonosetron concentration have the claimed 18-month and 24-month shelf stability. The ’351 application also teaches that specific categories of excipients may be used to improve upon that stability. Thus, based on the ’351 application, we find that a POSA would have been able to practice the full scope of the claimed inventions without extensive experimentation (factor 1). A POSA may need to perform routine stability studies to confirm that the formulations possessed the requisite 18-month or 24-month shelf-life stability but such testing does not rise to the level of undue experimentation.

Written Description
            We have considered the arguments and evidence presented by DRL and the countervailing arguments and evidence by Helsinn, and find that Helsinn’s evidence is more persuasive on the matter of written description. The ’351 application discloses to a POSA a range of 0.05 mg/mL palonosetron formulations that are stable at room temperature for 18 and 24 months. These formulations include stable embodiments containing 0.05 mg/mL palonosetron and a tonicifying agent, and 0.05 mg/mL palonosetron and mannitol with the formulation adjusted to the optimal pH range. We conclude that DRL has failed to meet its burden of demonstrating, by clear and convincing evidence, that the asserted claims lack adequate written description of the claimed stability limitation






[1] Mary L Cooper.  The case is Helsinn Healthcare SA et al. v. Dr. Reddy’s Laboratories Ltd. et al., case number 3:12-cv-02867.

Wednesday, February 22, 2017

PTAB Grants Petition for Inter Partes Review of Amgen's Neulasta Protein Refolding Patent

                The PTAB (Patent Trial & Appeal Board) granted Apotex’s petition for an inter partes review (IPR2016-01542) of Amgen’s U.S. Patent 8,952,138, which covers a method of refolding proteins.   Amgen has asserted the same patent in a civil case against Apotex, accusing Apotex’s Neulasta biosimilar of infringing the ‘138 Patent.

            Claim 1 of the ‘138 Patent claims a method for refolding a protein:

1. A method of refolding a protein expressed in a non-mammalian expression system and present in a volume at a concentration of 2.0 g/L or greater comprising: (a) contacting the protein with a refold buffer comprising a redox component comprising a final thiol-pair ratio having a range of 0.001 to 100 and a redox buffer strength of 2 mM or greater and one or more of: (i) a denaturant; (ii) an aggregation suppressor; and (iii) a protein stabilizer; to form a refold mixture; (b) incubating the refold mixture; and (c) isolating the protein from the refold mixture.

            Apotex's obviousness position relies mostly on two references, Schlegl (US 2007/0238860) and Hevehan (Biotechnology and Bioengineering, 1996, 54(3):221-230).  Apotex asserted that Schlegl discloses contacting bovine α-lactalbumin with a refold buffer comprising a redox component as part of the dilution refold method of Schlegl to form a refold mixture.  Apotex further asserted that a person of ordinary skill in the art would understand that the addition of cystine and cysteine in Schlegl serves as the redox system or redox component for bovine α- lactalbumin.  According to Apotex, this redox component has a thiolpair ratio of 2 and a redox buffer strength of 6 mM.  Regarding the second reference, Apotex asserted that Hevehan describes contacting a hen egg white lysozyme with a refold buffer comprising a redox component to form a refold mixture.  According to Apotex, the redox component had a thiol-pair ratio of between 0.3 and 9 and a redox buffer strength of 5 mM to 19 mM, the optimum being between 10-16 mM.

            In granting the IPR petition, the PTAB disagreed with Amgen that claim 1 requires a separate solution of “redox component” having a discrete volume and the “one or more of” components (i), (ii), and (iii) having a discrete volume. Rather, the PTAB stated that the broadest reasonable reading of the redox “component” is as a component portion of the refold buffer overall.

            Accordingly, the PTAB found that Apotex had demonstrated a reasonable likelihood of prevailing on its assertion that the challenged claims are obvious.  (Claims 1-11 and 13-24 under 35 U.S.C. § 103(a) as unpatentable over Schlegl and Hevehan and Claim 12 under 35 U.S.C. § 103(a) as unpatentable over Schlegl, Hevehan, and Hakim (mAbs, 1:3, 281-287)).



Friday, February 17, 2017

GENENTECH: AMGEN NOT DISCLOSING MANUFACTURING PROCESS IN AVASTIN BIOSIMILAR PATENT DANCE

           On February 15, 2017, Genentech filed suit against Amgen accusing Amgen of failing to adequately disclose its manufacturing process to Genentech despite agreeing to engage in the biosimilar "patent dance.” After the FDA accepted Amgen’s aBLA for a biosimilar version of Avastin®, Amgen gave notice of the acceptance to Genentech within the 20 day statutory period.  Amgen further produced only its aBLA, and announced that doing so “satisfie[d] Amgen’s production obligations under 42 U.S.C. § 262(l)(2)(A),” and took the position that the sixty-day countdown for Genentech to prepare its list of patents under § 262(l)(3)(A) had begun to run.  In the Complaint, Genentech states that it is unable to prepare a list of patents that Amgen infringes without additional information from Amgen, particularly “information that describes the process or processes used to manufacture the biological product that is the subject of such application.” 42 U.S.C. §262(l)(2)(A).  Genentech seeks immediate relief from the court since “if Genentech fails to list a patent, it could be barred permanently from asserting that patent against Amgen’s biosimilar Avastin®.”

            Genentech further states that it identified for Amgen the following categories of information relevant to antibody manufacturing patents in Genentech’s portfolio, and provided Amgen with exemplary Genentech patents that were potentially implicated:

• Characterization of the complete genome and phenotype of host cells used to manufacture bevacizumab;
• Composition of all cell culture media, including the amounts of each component of the media;
• For each attempt by Amgen to culture cells transformed with DNA encoding bevacizumab, all information concerning the extent and nature of glycosylation of bevacizumab (for example, relative percentages of different glycoforms of bevacizumab);
• Parameters monitored during any attempt by Amgen to culture cells transformed with DNA encoding bevacizumab;
• All information concerning any sparging of the pre-harvest or harvested culture fluid;
• Protein A chromatography parameters, including the compositions and properties of all buffers used in the process;
• From each attempt by Amgen to purify bevacizumab using Protein A chromatography, information concerning the temperature of the material loaded onto the column and the temperature of the column;
• Cation exchange chromatography parameters, including the compositions and properties of all buffers used in the process, the amount of antibody loaded onto the cation exchange resin, the volume of the cation exchange resin, and column regeneration procedures;
• Anion exchange chromatography parameters, including the compositions and properties of all buffers used in the process;
• From each attempt by Amgen to purify bevacizumab, measurements of the amount of bevacizumab monomer and amounts of bevacizumab dimers and multimers, before and after cation or anion exchange chromatography;
• All parameters concerning any viral inactivation steps or protocols, including all information concerning the effects of such processes on the stability of bevacizumab;
• All information concerning the ABP 215 formulation and its development, including any experiments performed with excipients other than those found in ABP 215;
• All information concerning the filling of vials to manufacture the ABP 215 drug product; and
• All information concerning the use of tangential flow filtration, including the processes used to adjust buffer concentrations.

Genentech seeks information from Amgen regarding the above manufacturing processes before it provides Amgen a list of patents that Amgen possibly infringes. 

            According to Genentech, the issue in this case is different from  Amgen Inc. v. Sandoz Inc., 794 F.3d 1347 (Fed. Cir. 2015), cert. granted, ___ U.S. ___, 2017 WL 125662 (Jan. 13, 2017), where the Supreme Court will decide whether a biosimilar applicant can opt out of the BPCIA information exchanges altogether, and if so what are the consequences.





[1] Case No. 1:17-cv-00165, D. Delaware.

Wednesday, February 15, 2017

COHERUS FILES FOUR IPR PETITIONS AGAINST HUMIRA BUFFER-LESS PATENT

            Coherus BioSciences, Inc. filed four IPR petitions against a single Humira patent, U.S. Patent No. 9,085,619.  (IPR2017-00822, IPR2017-00823, IPR2017-00826, and IPR2017-00827).  At first glance, one might think that the ‘619 Patent is a crucial patent since Coherus filed four IPR petitions against the ‘619 Patent.  However, the ‘619 patent is not on the list of Humira patents that Abbvie alleged Amgen’s Humira biosimilar (Amjevita) infringed. 

            Independent claim 16 of the ’619 Patent covers formulations of adalimumab (Humira) in water without a “buffering system.”  According to Coherus, it was known for decades that a protein, by itself, can provide buffer capacity and that a protein’s buffer capacity comes from the acidic or basic side chains of certain of its constituent amino acids.

            Coherus relies on a different ground of invalidity in each IPR petition.  In IPR2017-00822, Coherus argues that Gokarn PCT (WO 2006/138181) teaches the use of adalimumab in self buffering formulations—i.e., “in the absence of other buffers.”  According to Coherus, the Gokarn PCT also teaches that in a most preferred embodiment, the protein (e.g., adalimumab) provides “at least approximately 99% of the buffer capacity of the composition.”  Additional prior art relied on by Coherus in its petitions include the 2003 Humira® Label, Fransson (J. Pharm. Pharmacol., 48:1012-1015 (1996)), the 2005 Gamimune® Label, Gokarn provisional patent application (No. 60/690,582) (relied on as a 102e references), and Gokarn U.S. 2016/0319011.

            In IPR2017-00822, Coherus provides the following claim chart to prove that the claims of the ‘619 Patent are anticipated in view of the Gokarn PCT:

U.S. Patent No. 9,085,619
Gokarn PCT (WO 2006/138181)

Claim 16. An aqueous pharmaceutical formulation comprising:       
The preamble is non-limiting.
"[T]he invention provides self-buffering pharmaceutical protein formulations that are suitable for veterinary and human medical use."
(a) an anti-tumor necrosis factor alpha antibody comprising a light chain variable region (LCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO:3, a CDR2 domain comprising the amino acid sequence of SEQ ID NO:5, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 7, and a heavy chain variable region (HCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO:4, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 6, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO:8,
"wherein the protein is selected from the group consisting of ... HUMIRA (adalimumab) .... Adalimumab comprises these sequences. This is further demonstrated by claim 18, which is dependent on claim 16 and recites adalimumab.

wherein the concentration of the antibody is 50 to 200 mg/ml; and
"HUMIRA (adalimumab)" discloses 50 mg/mL. "[W]herein the concentration of the protein is between approximately ... 20 and 200 ... mg/ml."

"[W]herein the concentration of the protein is between approximately 20 and 400 mg/ml.



(b) water;
"[F]ormulations of self-buffering proteins comprise a protein and a carrier… In preferred embodiments ... the carrier is a liquid.... Liquid carriers may be organic or non-organic. Preferably they are aqueous, most preferably are largely or entirely comprised of pure water."







wherein the formulation does not comprise a buffering system.
"The self-buffering protein formulations are substantially free of other buffering agents  ...
"Self-buffering' means the capacity of a substance, such as a pharmaceutical protein, to resist change in pH sufficient for a given application, in the absence of other buffers.
"the protein provides ... very highly especially particularly preferably at least approximately 99% of the buffer capacity of the composition"
 Claim 5 refers to "pH maintained by the buffering action of the protein .... "

Claim 17. The formulation of claim 16, wherein the antibody comprises a LCVR comprising the amino acid sequence set forth in SEQ ID NO: 1, and a HCVR comprising the amino acid sequence set forth in SEQ ID NO: 2.
"Wherein the protein is selected from the group consisting of ... HUMIRA (adalimumab) .... Adalimumab comprises these sequences. This is further demonstrated by claim 18, which is dependent on claim 17 and recites adalimumab.

Claim 18. The formulation of claim 17, wherein the antibody is adalimumab.
"Wherein the protein is selected from the group consisting of ... HUMIRA (adalimumab)


Claim 19. The formulation of claim 16, wherein the formulation further comprises a non-ionizable excipient.
"One or more of sorbitol, mannitol, sucrose .... "  "polysorbate 80."
Claim 23, which depends from claim 9 ("requiring one or more pharmaceutically acceptable polyols").  As evidenced by Claim 10, such polyols include mannitol and sucrose.

Claim 24. The formulation of claim 16, wherein the pH of the formulation is from 4 to 8.
"HUMIRA (adalimumab)" discloses pH 5.2. Claim 23, which depends from claim 5 (requiring "the pH maintained by the buffering action of the protein is between approximately  3.5 to 8.")

Claim 25. The formulation of claim 16, wherein the pH of the formulation is from 4 to 6.
See above for claim 24. Further, "the pH maintained by the buffering action of the protein is between approximately ... 4.0 to 6.0…..


Claim 26. The formulation of claim 16, wherein the pH of the formulation is from 5 to 6.
See above for claims 24-25. Further, the Gokam PCT demonstrates that a variety of antibodies possess significant buffering capacity in the pH range 5.0 to 6.0.

Claim 27. The formulation of claim 18, wherein the pH of the formulation is from 4 to 8.
"HUMlRA (adalimumab)" discloses pH 5.2. Claim 23, which depends from claim 5 (requiring "the pH maintained by the buffering action of the protein is between approximately…4.0 to 6.0...



Thursday, February 9, 2017

The Federal Circuit Stays Injunction in Sanofi/Amgen Patent Fight over PCSK9 Inhibitor

http://bit.ly/2kAA3bP

HOSPIRA'S NEW HERCEPTIN IPRS (INTER PARTES REVIEW)

On January 30th, 2017, Hospira filed two petitions for inter partes review of Genentech’s U.S. Patent 7,371,379 (IPR2017-00805) and U.S. Patent 6,627,196 (IPR2017-00804). These patents claim methods of treating cancers characterized by overexpression of ErbB2 receptor with trastuzumab (Herceptin®).

U.S. Patent 7,371,379 claims:
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.

U.S. Patent 6,627,196 Claims
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.

Horpira’s position is that the claims of these patents are Obvious over the Herceptin Label in View of Baselga ’96[1], Pegram ’98[2], and the Knowledge of a Person of Ordinary Skill in the Art:
  • The Herceptin Label teaches treatment of a human patient diagnosed with cancer characterized by over expression of ErbB2 by administering an anti-ErbB2 antibody. For example, the Herceptin Label teaches that rhuMAb HER2, which is an anti-ErbB2 antibody, is “administ[ered]” in “dosage[s]” “for the treatment of [human] patients with metastatic breast cancer whose tumors over express the HER2 protein.
  •  Baselga ʼ96 reports the results of a phase II clinical trial in which patients with ErbB2-over expressing metastatic breast cancer were treated with rhuMAb HER2. The pharmacokinetic goal of the trial “was to achieve rhuMAb HER2 trough serum concentrations greater than 10 µg/mL, a level associated with optimal inhibition of cell grown in the preclinical model.” …Baselga ’96 also teaches that in preclinical studies (both in vitro and in xenografts), rhuMAb HER2 “markedly potentiated the antitumor effects of several chemotherapist agents, including cisplatin, doxorubicin, and paclitaxel, without increasing their toxicity.”
  •   Pegram ʼ98 reports that a rhuMAb HER2 “target trough serum concentration of 10 to 20 µg/mL” was used. Pegram ’98 also reports a toxicity profile of the combination that paralleled the toxicity of cisplatin alone. This led to the conclusion that rhuMAb HER2 did not increase toxicity.
Hospira’s previously filed Herceptin related IPR petitions can be accessed here: <http://biopharmapatent.blogspot.com/2016/12/hospira-has-filed-petition-for-inter.html>



[1] 14(3) J. CLIN. ONCOL. 737–44 (1996)
[2] 16(8) J. CLIN. ONCOL. 2659–71 (1998)