Dockets: T-1409-04
T-1890-11
T-2300-05
Citation:
2017 FC 726
Ottawa, Ontario, July 26, 2017
PRESENT: The
Honourable Mr. Justice Barnes
Docket: T-1409-04
|
BETWEEN:
|
ASTRAZENECA
CANADA INC. AND AKTIEBOLAGET HÄSSLE
|
Plaintiffs
(Defendants
by Counterclaim)
|
and
|
APOTEX INC.
|
Defendant
(Plaintiff
by Counterclaim)
|
Docket: T-1890-11
|
AND BETWEEN:
|
ASTRAZENECA AB
AND AKTIEBOLAGET HÄSSLE
|
Plaintiffs
(Defendants
by Counterclaim)
|
and
|
APOTEX INC.
|
Defendant
(Plaintiff
by Counterclaim)
|
Docket: T-2300-05
|
AND BETWEEN:
|
APOTEX INC.
|
Plaintiff
|
and
|
ASTRAZENECA
CANADA INC.
|
Defendant
|
PUBLIC JUDGMENT AND REASONS
Table of Contents
I. During the Period of
Infringement of the 693 Patent, Did Apotex Have an Available Non-Infringing
Alternative 4
A. Are the Proposed NIAs
Bioequivalent to LOSEC?. 23
B. Could Apotex Have
Conducted Human Clinical Trials on its Proposed NIAs to Prove Bioequivalency? 38
C. Are the Proposed NIAs
Sufficiently Stable?. 41
D. Would the Proposed
NIAs Have Received Regulatory Approval?. 59
E. Would Apotex Have
Obtained and Used a Third-Party NIA?. 63
F. Conclusion on the
Availability of a NIA.. 79
II. How Should the Court
Reconcile the Section 8 Judgment in Favour of Apotex in Court Docket T-2300-05
with the Infringement Judgment in Favour of AstraZeneca in Court Dockets
T-1409-04 and T-1890-11. 79
III. With Respect to
Apotex’s Profits From the Infringement of the 693 Patent, What Allowance Should
be Made for Profits-On-Profits. 92
A. Tax Effects on
Profits-on-Profits. 94
IV. With Respect to the
Infringement of the 693 Patent, What Allowance is Required Having Regard to the
United States District Court Award For the Infringement of the United States
505 Patent And Apotex’s Satisfaction of That Award. 95
V. Disposition. 106
[1]
In these bifurcated proceedings, AstraZeneca
Canada Inc., Aktiebolaget Hässle and AstraZeneca AB [collectively AstraZeneca]
seek an accounting of the profits earned by Apotex Inc. [Apotex] from the
infringement of AstraZeneca’s Canadian Letters Patent No 1,292,693
[693 Patent]. In the liability phase of the infringement actions (Court
dockets T-1409-04 and T-1890-11), the Court found in favour of AstraZeneca:
see AstraZeneca v Apotex, 2015 FC 322, 134 CPR (4th) 1, aff’d
in part 2017 FCA 9, [2017] FCJ No 22 (QL). The commercial product covered by
the 693 Patent is an omeprazole formulation marketed by AstraZeneca in Canada
under the trade name LOSEC. The period of infringement by Apotex runs from
September 5, 2003 to December 3, 2008.
[2]
In Court docket T-2300-05 Apotex, in turn, seeks
an offset for its section 8 damages for being held out of the Canadian market
for its generic omeprazole formulation [Apo-Omeprazole] between January 3, 2002
and December 30, 2003 by reason of AstraZeneca’s failed Patented Medicines (Notice
of Compliance) [NOC] application concerning its 762 Patent.
[3]
All of these references were consolidated by a
case management Order dated December 11, 2013 and were tried together at
Toronto.
[4]
To their considerable credit, the parties have
resolved most of their quantification issues and have reduced their agreement
to writing [see Exhibit AZ 24]. They have also agreed that their respective
accounting experts will adjust their calculations as required by the streamlining
agreement and by any other issues resulting from the Court’s Judgment. Any
remaining points of disagreement between the accounting experts will be
referred to the Court for final resolution.
[5]
The parties have left with the Court the
following matters for determination:
(a)
During the period of infringement of the 693
Patent, did Apotex have an available non-infringing alternative [NIA];
(b)
How should the Court reconcile the
section 8 Judgment in favour of Apotex in Court docket T-2300-05 with the
infringement Judgment in favour of AstraZeneca in Court dockets T-1409-04 and
T-1890-11;
(c)
With respect to Apotex’s profits from the
infringement of the 693 Patent, what allowance should be made for
profits-on-profits; and
(d)
With respect to the infringement of the 693
Patent, what allowance is required having regard to the United States District
Court award for the infringement of the United States Patent No 4,786,505 [505
Patent] and Apotex’s satisfaction of that award.
[6]
The matter of costs is to be left pending
further submissions from the parties.
[7]
It is now well established in Canadian law that a
NIA defence is available to a patent infringer to potentially reduce an
innovator’s claim to damages or to the recovery of the infringer’s profits.
[8]
The onus rests on Apotex to prove that a NIA was
available and at what cost. This point was made in Reading & Bates
Construction Co v Baker Energy Resources Corp (1992), 44 CPR (3d) 93 at pp 106-107,
56 FTR 22 (FCTD), aff’d (1994) 58 CPR (3d) 359, 175 NR 225 (FCA), where Justice
Barry Strayer held:
I also agree with
the learned referee’s conclusions of law that the onus is on the defendant to
prove that an alternative non-infringing method existed and the costs of using
that method. Although the defendant cited several cases to the contrary, these were
cases from the Circuit Courts of the United States, one of which was over 100
years old and none of which were less than about 50 years old. On the other
hand, I believe that such Canadian jurisprudence as exists is consistent with
the burden being on the defendant to prove the alternative and its cost. It has
been held in this court, for example, that in accounting for profits the burden
is on the defendant to prove his costs, and thus establish the net profits from
his sales: [citations removed]. Consistently with this fundamental principle,
it is equally incumbent upon the defendant to prove his real net profits from
using the infringing method by establishing on a balance of probabilities what
his costs would have been had he used the most likely non-infringing
alternative method. Therefore, the learned referee was right in law in imposing
that burden on the defendant in this case.
Also see Apotex Inc v Merck & Co, 2015
FCA 171 at para 74, 387 DLR (4th) 552 [Lovastatin FCA], and Pfizer
v Teva, 2016 FCA 161 at paras 53-66, 400 DLR (4th) 723, where
the Court said: “[m]ere possibilities short of
probabilities do not suffice” [para 56].
[9]
The NIA defence was initially received in Canada
with some hesitation perhaps because of the qualified language used in Monsanto
Canada Inc v Schmeiser, 2004 SCC 34, [2004] 1 S.C.R. 902. More recently,
however, the Federal Court of Appeal has fully endorsed the defence, at least
in conceptual terms: see Lovastatin FCA and Apotex Inc v ADIR,
2017 FCA 23, [2017] FCJ No 110 (QL) [Perindopril FCA]. But, as with any
legal principle, the real challenge lies in its application to the evidence.
This case is no different.
[10]
Lovastatin FCA,
above, contains a useful discussion of the theory behind the NIA defence and
the method of applying it. At its root is the need for a causal link between
the infringement and the claimed recovery. Behind the application of the NIA
idea is said to lie “robust common sense” about
what would and could have happened “but for” the
infringement. The following passages from the decision are particularly
instructive:
[48] The difficulty with the Judge’s
approach is that if damages for lost profits are calculated never having regard
to an available non-infringing alternative, the patentee will sometimes be
better off than it would have been in the absence of infringement. This is so
for the following reason. Where a defendant can make and sell a non-infringing
alternative, the patent does not confer a complete monopoly on the patent
holder. Instead, the patent confers a share of market power upon the patentee.
In this circumstance, where, instead of using a non-infringing alternative, a
defendant infringes, it is a question of fact whether, “but for” the
infringement, the defendant would not have competed with it. The defendant’s
lawful competition in the “but for” world may have deprived the patentee of
some sales.
[49] Put another way, in cases where,
in the “but for” world, the infringer could and would have made and sold a
non-infringing alternative, these sales may well reduce the patent owner’s
sales. Awarding the patentee full damages for lost profits in every case will,
therefore, sometimes over-compensate the patentee.
[50] Perfect compensation requires
consideration of: (i) what, if any, non-infringing product the defendant or any
other competitors could and would have sold “but for” the infringement; and,
(ii) the extent lawful competition would have reduced the patentee’s sales.
…
[73] When considering the effect of
legitimate competition from a defendant marketing a non-infringing alternative,
a court is required to consider at least the following questions of fact:
i) Is the
alleged non-infringing alternative a true substitute and thus a real
alternative?
ii) Is the
alleged non-infringing alternative a true alternative in the sense of being
economically viable?
iii) At
the time of infringement, does the infringer have a sufficient supply of the non-infringing
alternative to replace the non-infringing sales? Another way of framing this
inquiry is could the infringer have sold the non-infringing alternative?
iv) Would
the infringer actually have sold the non-infringing alternative?
[74] As a matter of principle, the
burden lies on the defendant to establish the factual relevance of a
non-infringing alternative on a balance of probabilities. Indeed, Apotex
acknowledged in oral argument that it bears the persuasive burden, on a balance
of probabilities, to prove that it would have used the non-infringing
alternative. This is consistent with jurisprudence such as Rainbow
Industrial Caterers Ltd. v. Canadian National Railway Co., [1991] 3 S.C.R.
3, 84 D.L.R. (4th) 291.
[Emphasis in original.]
…
[89] While this is dispositive of the
appeal on this issue, I also find that Apotex failed to establish that it would
have replaced its infringing sales. I reach this conclusion on the following
basis.
[90] First, as Apotex conceded in oral
argument:
• The
real world informs our construction of the “but for” world.
• Conduct
in the real world is “very important” to what would have happened in the “but
for” world.
• Findings
of fact from the liability decision are relevant to constructing the “but for”
world.
• “Brazen”
infringement in the real world makes it very difficult to prove that the
defendant would have deployed the non-infringing alternative in the “but for”
world.
[91] In the liability phase, the Judge
found, at paragraph 309 of her reasons (reported at 2010 FC 1265), that if Blue
Treasure had been using the non-infringing process to ferment lovastatin, it
would have lost significant amounts of money for each kilogram of product it
shipped to AFI. However, Apotex knew that once Blue Treasure began to use the
allegedly non-infringing process it became profitable. The inference to be
drawn is that Apotex knew Blue Treasure was in fact using the infringing
process; yet Apotex used that bulk product to prepare and sell its lovastatin
tablets.
[92] In this circumstance it is
relevant to note that from January 1, 1997 to January 1, 2001 Apotex believed
Merck’s patent was invalid.
[93] Apotex’ evidence falls far short
of demonstrating that it would have sold the non-infringing product when one
considers: the scale of Apotex’ infringement; its likely knowledge that Blue
Treasure was supplying it with infringing lovastatin; its belief the Merck
patent was invalid; its failure to call a witness from AFI to support its
contention that, had it known the product was infringing, it would have
resurrected operations at AFI in Winnipeg; and the fact the Judge found that
the testimony of Apotex’ only fact witness was, albeit not on this point,
unsubstantiated and self-serving.
[11]
The NIA defence was more recently endorsed in Perindopril
FCA, above. There Apotex advanced the defence based on the asserted
availability of the patent-protected product from certain foreign sources for
sale into non-infringing markets. The Court expressly rejected the idea that a
NIA could not take the exact form of the patented product. Such an approach,
it said, would inappropriately extend the territorial reach of the Canadian
patent into non-infringing jurisdictions. The Court was also unperturbed by
the fact that, at the beginning of the infringing period, none of the
identified foreign third-party suppliers of Perindopril had the compound at
hand. The question posed was whether, in the hypothetical world, Apotex could
and would have obtained sufficient quantities of non-infringing product and
that it could and would have used that product [see para 41]. The Court
discussed this point in the following way:
[42] As this Court later explained in Pfizer
Canada Inc. v. Teva Canada Limited, 2016 FCA 161, 483 N.R. 275, (Effexor)
at paragraph 50, both the “could have” and “would have” requirements are
important. To prove “could have”, the defendant must demonstrate that it was
possible for it to secure non-infringing product. To prove “would have”, the
defendant must demonstrate “that events would transpire in such a way as to put
them in that position” (Effexor, paragraph 50). The importance of the
“would have” requirement is that by requiring a defendant to show that it would
have used a non-infringing alternative, the defendant shows that the value of
the patented invention is not such that reliance on alternatives is unlikely or
fanciful. Put another way, notwithstanding the availability of a non-infringing
alternative, the defendant must show that there are no impediments to its use.
[12]
AstraZeneca contends that the jurisprudence does
not support a NIA that is not perceived by the infringer to be non-infringing
at the point of the infringement. It also posits that a NIA must be “foreseeable” to the infringer at the relevant time.
Anything short of this is said to be speculative.
[13]
In support of the “knowledge”
requirement, AstraZeneca relies on the trial decision in Wellcome Foundation
Ltd v Apotex Inc (1998), 82 CPR (3d) 466 at paras 32-33, 151 FTR 250 (FCTD)
[Wellcome FC], aff’d [2001] 2 FCR 618, 11 CPR (4th) 218
(CA). AstraZeneca cites to Lovastatin FCA, above, at paras 93-95 on the
issue of foreseeability.
[14]
I do not read these decisions as broadly as
AstraZeneca suggests. In Wellcome FC, above, Justice MacKay did focus
on whether Apotex had actual knowledge that its proposed NIA was non-infringing,
but he also considered whether “it could have known”
[para 33]. Knowing whether or not a proposed NIA would infringe is, of course,
a factor in determining whether the infringer “would
have” employed it in place of the infringing product. But this falls
well short of making prior knowledge of non-infringement an absolute
pre-requisite to the assertion of a NIA.
[15]
I also place little significance on the stray
reference to “foreseeability” in Lovastatin FCA,
above. In the context of its use I take that reference to mean only that the
concept of a viable NIA would have been available to the infringer based on
what was known in the art at the time. If foreseeability meant that the
infringer must have the asserted NIA in mind at the time of the infringement,
it could potentially punish those who had no idea their product was infringing while
rewarding those who had an appreciation of the risk and courted it, but
nevertheless had a back-up, work-around solution available.
[16]
In its Closing Argument on NIA at para 56,
AstraZeneca cites two United States authorities (Grain Processing Corp v
American Maize-Products Co, 185 F 3d 1341 (Fed Cir 1999) [Grain
Processing], and Micro-Chemical Inc v Lextion Inc, 318 F 3d 1119
(Fed Cir 2003) [Micro-Chemicals]) for the idea that a NIA requiring the
infringer to “invent around the patented technology”
is not considered to be “available” to the
infringer. I do not agree with this interpretation and in oral argument
counsel retreated somewhat from the above proposition. Neither Micro-Chemical,
above, nor Grain Processing, above, stand for the idea that the
availability of a NIA is necessarily contingent on the amount of inventive
effort required to make it. The time and effort of coming up with a
non-infringing solution is certainly relevant to whether the infringer would
have pursued it, but they are not absolute barriers to the defence. That this was
all Judge Rader for the Court was saying in Micro-Chemical is clearly evident
from his statement at p 1123 that high costs and the complexity of the exercise
“to design or invent around the patented technology to
develop an alleged substitute weighs [sic] against a finding of
availability”. The Court in Grain Processing makes the same
point.
[17]
The American authorities cited by the parties also
do not, on my reading, support an argument for exclusion of a NIA that is not “on the market” at the time of infringement. In Grain
Processing, above, the Court was only concerned with the hypothetical
availability of a NIA “including but not limited to
products on the market” [p 1349]. Where the substitute was not on the
market at the relevant time, the Court observed that an inference of
unavailability could be drawn but not that it must be drawn. The Court went on
to say at p 1353 that “the trial court must proceed
with caution in assessing proof of the availability of substitutes not actually
sold during the period of infringement”. In that case, however, the
trial court had found that the asserted substitute could have been made by a
process that was known in the art. That finding was upheld on appeal. I can
see nothing in the Micro-Chemical decision that detracts from the above
view.
[18]
There is, of course, a difference between cases
like Perindopril FCA and this one. In Perindopril FCA the NIA
was known to exist at the time of infringement. The NIAs Apotex proposes in this
case were unknown and never made by anyone before or during the infringing
period let alone approved for use in Canada, the United States or elsewhere.
Notwithstanding this distinction, I accept Apotex’s point that in the
hypothetical, but for pharmaceutical world the infringer’s failure to produce a
viable NIA formulation in the real world is not a threshold bar to the use of
the NIA defence. In this context, the question is: Could the infringer have
made the product had it attempted to do so at the relevant time and would the
infringer have sold the product on some reasonable financial basis in
substitution for the infringing product?
[19]
I think this is the point being made by Justice
Eleanor R Dawson for the Court in Perindropril FCA, above, when she said
at para 62 “the fact that an event does not take place
in the real world does not necessarily mean that the event could not and would
not have taken place in the hypothetical world”. Added to this is the
recognition in Perindropril FCA that the availability of a NIA is not to
be foreclosed simply because it was not immediately available to the infringer,
i.e. on the eve of first infringement. The Court is still obliged “to consider whether at some later point in time a supplier
would and could have provided” a replacement product [see para 67].
This lends support to Apotex’s view that a viable NIA need not exist at the
exact time of infringement.
[20]
All of this is not to say that the
post-infringement development of a NIA does not present problems of proof for
the infringer asserting the defence. Indeed, as explained below, serious
problems of proof are manifest in this case.
[21]
One of the difficulties with an ex post facto
NIA solution was recently discussed in Bell Airbus Helicopters SAS v Bell
Helicopter Texteron Canada Limitée, 2017 FC 170 at para 295, 144 CPR (4th)
281 [Airbus]. There Justice Luc Martineau explained that the Court must
be very wary of hindsight bias when it considers the claimed ease with which an
after-the-fact NIA could be developed, tested, scaled-up and approved for use.
In a case where the use of a product carries considerable infringement risk,
one is left to wonder why the supposedly simple, non-infringing, equal cost
version was never attempted. The “could have and would
have” evidentiary concerns are also magnified when the proposed
hypothetical NIA(s) were never, at any time, submitted to the relevant
regulator for assessment and approval.
[22]
I do not, however, think that Justice
Martineau’s decision in Airbus, above, stands for the proposition that ex
post facto NIAs of the sort proposed in this case must be excluded from
consideration as a matter of law. Justice Martineau simply expressed
reservations about the dangers of relying on a NIA that was either unknown during
the period of infringement or had been previously discarded. He was
appropriately concerned about the reliability of this type of look-back evidence
and the risk of hindsight bias [see para 295].
[23]
I have similar concerns to those expressed by
Justice Martineau about the NIA evidence presented by Apotex in this case concerning
its recently developed in-house NIA formulations.
[24]
It is one thing to rely upon a NIA that is known
and available for use during the period of infringing activity. It is quite
another thing to propose a NIA made long after an infringement has taken place.
When a pharmaceutical NIA has been created and has obtained regulatory
approval, one is not left to wonder whether it “could”
have been available for use (assuming a capacity to obtain it in commercial
amounts). In this case, however, Apotex’s self-created NIAs were made in
non-commercial batches, without full stability, bioequivalency or clinical
studies, and without obtaining the required regulatory approvals for commercial
use. Indeed, Apotex had no intention of ever developing these formulations for
commercial exploitation. Many questions, therefore, remain about whether and,
if so, when any of the formulations could have been used successfully during
the period of infringement.
[25]
Apotex attempts to explain away the evident
weaknesses in its testing evidence with the argument that AstraZeneca and its
experts misconceived Apotex’s NIA burden. Apotex puts the issue in the
following way, at para 113 of its Closing Submissions:
…The issue before the Court is whether one
or more of the NIA formulations could meet regulatory requirements had Apotex
manufactured them at a commercial scale and made the requisite regulatory
filings, not whether the data generated is sufficient to meet regulatory
standards.
•
Were Apotex to have done what Astra requires of it, millions of
capsules would have had to be manufactured and studied over the period of a
year. Moreover, hundreds of humans would have needlessly been subjected to
clinical studies.
[26]
The difficulty with the above idea is that,
without ever acquiring the data necessary to satisfy regulatory requirements
for its proposed NIAs, Apotex cannot directly establish that any of them would
have obtained that approval. Incomplete or inconclusive data is weak data.
The fact that Apotex began its stability testing too late to get it finished
before trial and did not conduct clinical bioequivalency research at all does
not make its case for NIA viability any stronger. The same can be said of the
experimental short-cuts and less-than-optimum testing protocols employed by
Apotex in the generation of its stability data. While these approaches may be
entirely appropriate for the purpose of making in-house formulation choices to
advance product development, they have diminished probative value where the
question is whether a particular formulation would have been sufficiently
viable to obtain regulatory approval on a balance of probabilities.
[27]
It is also of some significance that Apotex
unsuccessfully asserted a NIA defence in the damages-assessment phase of the United
States litigation. Apotex argued there that it could have made adjustments to
the infringing formulation, adopted an existing non-infringing formulation or
used a microtablet formulation. These arguments were wholly rejected by the
United States District Court for the Southern District of New York [District
Court] in AstraZeneca AB v Apotex Corp, 985 F Supp 2d 452 (2013). The
Court characterized Apotex’s proposed formulation adjustments in the following
way, at p 499:
As for Apotex's proposals for tinkering with
the ingredients in its pellets, it is pure speculation whether any of its
various proposals would create a stable, bioequivalent product that was
non-infringing. Apotex has never asked one of its many experts to try to create
the revised formulation, much less to create and test it. See SynQor, Inc. v.
Artesyn Techs., Inc., 709 F.3d 1365, 1382 (Fed.Cir.2013) (where an alleged
substitute is not on the market, “the accused infringer has the burden to
overcome the inference that the substitute was not ‘available’”) (citation
omitted).
There is a reason that Apotex chose the
ingredients that it did for its pellets following six years of research and
testing. Those ingredients created a successful product. This is no easy task
given the challenges of working with the omeprazole molecule and delivering it
sufficiently intact to the part of the body in which it is most effective.
[28]
In this case, Apotex belatedly attempted to
overcome the problem identified by the District Court by developing a set of
alternative formulations. However, Apotex has not adequately explained why it
waited until late 2015 to begin its stability testing when it knew or ought to
have known as of 2007 from the United States litigation that Apo-Omeprazole
infringed AstraZeneca’s formulation patents. Inexplicably, Apotex mounted a
purely theoretical NIA posture in the damages-assessment phase of the United
States proceeding and by the end of that case, the Court observed at p 449 that
it had “largely abandoned its argument that it could
have altered the infringing formulation successfully”. The rejection by
the District Court of Apotex’s NIA defence was based on a different and
presumably weaker evidentiary record than the record before me. Nevertheless,
I am left to wonder why Apotex failed to work-up its asserted alternative
formulations in this case long before the end of 2015. Its excuse that it
thought its formulation was non-infringing is undermined by the 2007 District
Court finding of infringement [see AstraZeneca AB v Mylan Labs Inc et al, 490
F Supp 2d 381 (2007)] which was subsequently upheld on appeal in 2008 in AstraZeneca
AB v Apotex Corp, 536 F 3d 1361 (Fed Cir). Apotex’s stability testing thus
commenced long after it knew or ought to have known that Apo-Omeprazole was
infringing.
[29]
Apotex’s failure to complete the testing of its
alternative formulations and to instead rely on extrapolations from its experts
in this case is an unacceptable approach. A recognition of this strategy would
potentially reward Apotex for its delay by excluding from consideration finished
stability test results – data that may well have established that the
alternative formulations would not work. It cannot be to Apotex’s advantage
that its delay in the initiation of obvious testing avoids the potential for
failed results. What Apotex is asking is that the Court predict a result that
it could have but failed to establish. On the evidentiary record before me, I
am not prepared to draw the inferences Apotex is seeking.
[30]
AstraZeneca relies heavily on the principle that
in the assessment of the but for world of NIAs the Court must look at what took
place in the real world including the behaviour and state of mind of the
infringer. Apotex does not deny this as a point of principle but argues for
its reduced significance.
[31]
Initially I did have reservations about the idea
that the availability of a NIA can be informed, in part, by the willfulness of
the infringement. But as I understand the decision of the Federal Court of
Appeal in Lovastatin FCA, the idea is no more than this: where an
infringer brazenly infringes a valid patent, or substantially courts the risk
of doing so, an inference may arise that no viable substitute was available.
If it were otherwise the rational choice would always be to employ the NIA and
not the infringing product.
[32]
It seems to me that what Apotex knew at the time
and what it did in response to that knowledge in the real world are important
considerations in the assessment of the hypothetical availability of its
after-the-fact NIAs. The suggestion today that the development and commercial
exploitation of the asserted NIAs would have been simple, cost-effective and
speedy is substantially belied by historical fact.
[33]
It is worth noting that it took Apotex many
years to develop and obtain regulatory approval for Apo-Omeprazole – a product
that Dr. Bernard Sherman apparently thought at the time would not infringe
the 693 Patent or the United States 505 Patent. This fact belies the argument
that any of the NIAs would have enjoyed an easier route to success if they were
developed from scratch and without the benefit of the development of Apo-Omeprazole.
Indeed, as I found in the liability phase, omeprazole is not an easy molecule
to formulate.
[34]
Dr. Sherman’s evidence that a work-around
NIA solution was a straight forward task is also belied by the experience of
producing the now-asserted NIA formulations. Initially Dr. Sherman
thought the solution lay in the removal of the alkaline reacting compound [ARC]
from the infringing formulation. Indeed, that was Dr. Sherman’s evidence
in the United States litigation. However when that approach was adopted for
this proceeding, it failed [see Exhibit APO 130, Chow Report #1 at
paras 84-85].
[35]
It is also noteworthy that none of the first 14
NIA formulations produced by Apotex were pursued in this litigation. This
supports an inference that each of them failed. Of those formulations that did
go forward to further testing, a number clearly failed to meet the necessary
stability or bioequivalency requirements. Of those formulations that Apotex
continues to assert, several were developed later in the selection process.
All of this undermines Apotex’s argument that numerous viable NIA options would
have been immediately obvious to a skilled formulator like Dr. Sherman.
[36]
Dr. Sherman’s excuse for not exploring his
NIA options during the infringing period was that he had no reason to think Apo-Omeprazole
was infringing. This evidence does not stand up to scrutiny. Indeed, as
discussed above, it either was or should have been increasingly obvious to
Dr. Sherman that Apo-Omeprazole was likely an infringing product.
Notwithstanding what Apotex knew or ought to have known, it persisted with its
use of Apo-Omeprazole. This continued infringing conduct was unreasonably
stubborn or dogmatic, if not wilfully blind to the consequences, and it contradicts
Dr. Sherman’s trial testimony that, had he known, Apotex would have immediately
searched for other options.
[37]
It is also of some significance that despite
increasing evidence of infringement Apotex chose not to examine Apo-Omeprazole
to determine if it incorporated an infringing subcoat. All of this conduct
undermines Dr. Sherman’s evidence that if he had only known Apo-Omeprazole
was infringing, he could easily have developed or purchased a NIA. The more
sustainable inference is that Apotex was prepared to run with Apo-Omeprazole
whatever the likely consequences and it is doubtful it would ever have pursued a
NIA option. That is particularly the case for pursuing a third-party NIA.
Dr. Sherman made it very clear that such an approach would not have been
considered unless and until he had exhausted his in-house options.
[38]
In assessing Dr. Sherman’s evidence about
what Apotex would have done in the hypothetical world it is necessary to
consider what he knew in the real world and what Apotex did or did not do with
that knowledge.
[39]
At least as early as 2000, Apotex knew that
AstraZeneca was asserting an infringement allegation based on an in situ
formed subcoat in connection with another generic omeprazole formulation. In AB
Hassle et al v Canada et al, 10 CPR (4th) 38, 102 ACWS (3d) 185
(FC), aff’d 2002 FCA 147, 18 CPR (4th) 558, Justice Daniele Tremblay-Lamer
made a finding of infringement on that basis.
[40]
In 2000, AstraZeneca made the same allegation
against Apotex and other generics in the infringement action in the United
States. In the first wave of that litigation, concluded in 2002, the District Court
found infringement on the part of a different defendant for an in situ formed
subcoat. At the conclusion of the second wave of cases in 2007, discussed
above, the same finding was made against Apotex.
[41]
In 2003, the Federal Court of Appeal construed
the 693 Patent claims to cover an in situ formed subcoat and rejected
Apotex’s arguments to the contrary: see AB Hassle v Apotex Inc, 2003
FCA 409, 29 CPR (4th) 23.
[42]
In 2004, AstraZeneca commenced the first of
these proceedings in Canada against Apotex for damages, alleging again that Apo-Omeprazole
infringed the 693 Patent on the basis of an in situ formed subcoat.
[43]
Notwithstanding the above history, Apotex took
no steps to pursue a NIA formulation or even to test whether Apo-Omeprazole
capsules contained an infringing subcoat layer.
[44]
On March 16, 2015, I rendered a Judgment in
these proceedings finding Apo-Omeprazole to be infringing of the 693 patent because
it incorporated a sub-coat layer formed in situ.
[45]
Having regard to the above background and to the
fact that no effort was made by Apotex until late 2015 to develop any NIA
formulations nor at any time or to pursue a third-party formulation,
considerable caution is warranted.
[46]
Apotex had no readily available NIA options at
any time during the infringing period and it had no back-up plan to develop or
purchase one. Instead it ran with Apo-Omeprazole to the end. Even now Apotex
produced late, incomplete and inconclusive stability and bioequivalency data
suggesting that it did not, and to this day does not, have a viable in-house
NIA option. Notwithstanding these concerns, I will proceed with an assessment
of Apotex’s evidence concerning its asserted NIAs to determine whether they
were available and true non-infringing substitutes for Apo-Omeprazole.
[47]
In that regard I can readily dispose of two
issues raised by AstraZeneca:
(a)
whether Apotex had the capacity to commercialize
one of its asserted NIA formulations (the could-have question); and
(b)
whether Apotex has failed to prove that each of
its asserted in-house NIAs is non-infringing.
[48]
While I accept that there would be manufacturing
challenges for Apotex during scale-up to commercial NIA production, I believe that,
with the exception of NIA formulation MR8620E1, these could be overcome by a
successful and sophisticated producer like Apotex. Enteric coatings have been
commercially used for many years and Apotex had considerable experience in
successfully applying them to its formulations. Dr. Davies identified a
number of production obstacles that Apotex may not have fully resolved in its
small-scale batches. However, I am left with the impression that Apotex could
and would have resolved most of these issues without the inordinate expenditure
of time or money and without compromising the dissolution profile of the
enteric coatings used in the NIA formulations.
[49]
I exclude from this finding formulation MR8620E1.
That formulation was designed to avoid an in situ subcoat by reducing
the water content of the MACP enteric coating dispersion. This change reduced
the potential for a reaction at the enteric coating/core interface.
[50]
I am not satisfied that MR8620E1 could have been
commercially developed because, as Dr. Davies explained, it failed to meet
the MACP manufacturer’s specification for solids content and this repeatedly
caused nozzle blockages [see Exhibit AZ 137 at paras 136-37]. In the
absence of persuasive evidence proving that this production problem could be
overcome at commercial production levels, I am not convinced that it would have
worked. Indeed, if it was as obvious a work-around as Apotex now suggests, one
is left to wonder why it was not attempted until well into Apotex’s NIA
development and why larger scale enteric coated batches were either not
attempted or were left undocumented.
[51]
I am also satisfied on the evidence provided by
the Apotex witnesses that it had ample in-house capacity to produce the
remaining NIA formulations, sufficient to match its infringing sales.
[52]
AstraZeneca contends that Apotex has failed to
prove that its proposed NIAs would not infringe the 693 Patent. Although
AstraZeneca has stipulated that Apotex’s proposed NIAs produced at batch scale
do not infringe [see Exhibit APO 69], it does not concede the same point for
any of the NIAs if produced at a commercial scale. I do not accept this
argument because it lacks direct evidentiary support.
[53]
If the NIAs are non-infringing at batch scale,
one would expect them to remain non-infringing on commercial scale-up. That
expectation might be rebuttable with cogent evidence that a production scale-up
would be likely to give rise to an infringing characteristic (e.g. an in situ
subcoat layer). No evidence directly on point was before me and I find that
the asserted NIA formulations at commercial scale would not infringe the 693
Patent.
[54]
Dr. Mario González is an expert in
pharmacokinetics, clinical pharmacology and biopharmaceutics [including the
development and application of in vitro–in vivo correlations and
relationship in predicting bioequivalence of formulations]. He provided expert
opinion evidence on behalf of Apotex as to whether, in the absence of in
vivo data, one could reasonably predict that any of the asserted NIAs would
be likely to be bioequivalent to LOSEC and, if so, how the prediction could be
made.
[55]
After advising Apotex that, in certain
circumstances, such predictions could be made, Apotex gave Dr. González
its pharmacokinetic/statistical clinical data comparing the bioequivalency of Apo-Omeprazole
and LOSEC along with its in vitro dissolution data and testing protocol.
From that information Dr. González was asked to provide an opinion “as to whether any of the [NIAs] would be expected to be
bioequivalent to Losec”.
[56]
Dr. González’s first report [see Exhibit APO
41] acknowledges that assessing bioequivalency for regulatory purposes between
two pharmaceutical compounds is carried out with randomized, cross-over human
clinical testing where blood plasma concentrations are measured over time and
compared. Acceptable clinical studies would require at least 12 human subjects
but more typically between 18 to 24 subjects “to gain
meaningful data”. The tested population needs to be large enough such
that the data are not unduly thrown-off by intra- and inter-subject
variability. Health Canada will accept two formulations as bioequivalent if
the comparative data meet minimum statistical standards [see para 32].
[57]
Dr. González’s first report states, at para
33:
where it is
undesirable or impractical to conduct a comparative bioavailability study to
determine whether two formulations are bioequivalent, it is, in certain
circumstances, possible to use alternative methods, such as an in vitro/in
vivo correlation (“IVIVC”) or an in vitro/in vivo relationship
(“IVIVR”), to provide a reasonable prediction that two formulations will be bioequivalent.
[58]
In this case an IVIVC could not be carried out
and Dr. González was limited to using the less robust IVIVR method – a
technique that he said “can be of great value during
formulation development” [see para 38].
[59]
Dr. González began his IVIVR bioequivalency
work-up by plotting the bioequivalency data for Apo-Omeprazole and LOSEC. He
observed them to be bioequivalent in the fasted state. According to Dr. González
any NIA formulation with an in vitro dissolution profile that fell
between those of Apo-Omeprazole and LOSEC “would be
expected to be bioequivalent to these formulations” [see para 52]. Such
a prediction could not be made with the same degree of confidence for a NIA with
an out-of-range profile. Of the fifteen NIA formulations Dr. González examined,
eight fell within the Apo-Omeprazole and LOSEC dissolution profiles. The
others were said to be “less likely to be bioequivalent
to Losec® and Apo-Omeprazole based on dissolution data” [see para 60]. Dr. González
then carried out a comparison using the f2 metric for similarity for
the remaining eight NIA formulations and observed them to be similar to either LOSEC
or Apo-Omeprazole. He concluded that all eight “would
be expected to be bioequivalent to Losec® and Apo-Omeprazole” [see para
65].
[60]
AstraZeneca countered Dr. González’s
evidence with the opinion of Dr. David Taft. He was qualified as an
expert in pharmaceutical sciences, including pharmacokinetics.
[61]
Dr. Taft was asked to advise if IVIVR was
an accepted and reliable technique for predicting bioequivalency generally and,
more specifically, for predicting the bioequivalency of the Apotex NIAs to LOSEC
or Apo-Omeprazole based on the available data.
[62]
Dr. Taft defined bioequivalency and its
regulatory significance in the following way in his responding report [Exhibit
AZ 160]:
37. Bioequivalence has been defined
as, “the absence of a significant difference in the rate and extent
to which the active ingredient or active moiety in pharmaceutical equivalents
or pharmaceutical alternatives becomes available at the site of drug action
when administered at the same molar dose under similar conditions in an
appropriately designed study.” (emphasis added)
38. Government agencies that regulate
marketed drugs, like the FDA and HC, set standards for how “absence of a
significant difference in the rate and extent” are to be determined. Cmax
and AUC are the parameters used to measure the rate and extent of absorption.
[Emphasis in original.] [Footnotes omitted.]
[63]
Dr. Taft expressed the opinion that the
bioequivalency of LOSEC and Apo-Omeprazole was not established by the data
relied upon by Dr. González. He was particularly concerned by the
exclusion of data from one of the test subjects (OM75). Had those data been
included, the results of the Apotex study would not have satisfied either the Health
Canada or the United States Food and Drug Administration [FDA] requirements for
bioequivalency. Notwithstanding Dr. Taft’s concern, the fact remains that
regulatory approval was obtained for Apo-Omeprazole and presumably the
exclusion of OM75 was accepted by the regulators. On the evidence presented,
including Dr. Taft’s evidence at Transcript p 4087 and Exhibit AZ 78, I am
not prepared to look behind that decision to reassess its scientific merit.
[64]
Dr. Taft was highly critical of Dr. González’s
analysis for other reasons. He challenged Dr. González’s use of mean
dissolution and absorption data obtained from the Apotex Apo-Omeprazole biostudy
based on the wide variability of the actual plasma concentrations for each of
the test subjects as compared to the mean. Figure 6 of his report, at p 31,
nicely depicts this variability. According to Dr. Taft this variability
weakens the IVIVR model as a foundation for comparing LOSEC and Apo-Omeprazole
to the NIAs.
[65]
According to Dr. Taft, the problem of
subject-to-subject variability in the Apotex biostudy data for Apo-Omeprazole
is compounded by the variability in the in vitro dissolution data for
the NIAs used by Dr. González. Because all of the dissolution data for
the NIAs exceeded the accepted margin of 10% for the IVIVC method, the data, he
thought, were “too variable to use for reliably
predicting bioequivalence” using the less robust IVIVR method [see para
116].
[66]
Dr. Taft was also critical of Dr. González’s
use of IVIVR as a predictive tool in proof of bioequivalence. He pointed out
that IVIVR is only useful as a tool for guiding formulation development.
According to Dr. Taft, there is nothing in the literature supporting its
reliability for predicting bioequivalence or indicating that it can be used as
a substitute for an IVIVC. Furthermore, Dr. González’s IVIVR method did
not meet the minimum standards required of an IVIVC. Dr. Taft concluded
this part of his report in the following way:
126. More fundamentally, Dr. González’s
“IVIVR” technique is incompatible with the underlying principles for an IVIVC.
An IVIVC is based on a formulation consistently demonstrating a relationship
between in vitro dissolution and in vivo absorption. Dr.
González’s “IVIVR” is based on an entirely different proposition. In particular,
Dr. González’s “IVIVR” technique contends that where different formulations
with different in vitro dissolution profiles (i.e.,
Apo-Omeprazole and LOSEC) have similar in vivo absorption profiles, any
test formulation (i.e., an AF) that has an in vitro dissolution
profile lying between the other two will also have the same in vivo
absorption profile (and hence be bioequivalent). That is, as he seems to
acknowledge, Dr. González’s “IVIVR” technique is not based on a
correlation between in vitro dissolution and in vivo absorption;
but rather on the assumption that such a correlation is irrelevant. However, if
dissolution is irrelevant to absorption, then it is not predictive of
absorption.
127. Based on the foregoing, in my view
the scientific community has not accepted and would be unwilling to accept Dr.
González’s “IVIVR” technique for establishing bioequivalence of the [NIAs] to
Apo-Omeprazole or LOSEC.
[Footnotes omitted.]
[67]
Dr. Taft had several other concerns about Dr. González’s
use of IVIVR to predict the bioequivalence of the NIAs to LOSEC and Apo-Omeprazole.
These included his failure to estimate the error rate, the lack of validation
as required by the FDA for IVIVCs and the lack of any data showing bioequivalence
in the fed state.
[68]
Dr. González’s reply report generally
addressed the points advanced by Dr. Taft [see Exhibit APO 43]. In
particular, he picked up on Dr. Taft’s point that LOSEC and Apo-Omeprazole
exhibited different dissolution rates for the first 40 minutes. When Dr. González
adjusted for the five minute time lag in dissolution between the two
formulations, the profiles were similar.
[69]
Dr. González commented on Dr. Taft’s
view that some in vivo absorption data for the NIAs was needed for a
valid IVIVR. Dr. Taft’s point was weakly countered with the following
statement, at para 45: “[h]owever, if there were in
vivo data available for these formulations, this would remove the need for
an IVIVR”. Dr. González’s substantive response was limited to the
observation that, as “minor” variations to Apo-Omeprazole,
the NIAs would be assumed to have the same linear relationship between
dissolution and absorption.
[70]
Dr. González accepted that “there may be some larger than expected variance” in
the dissolution data for the NIA formulations. This he attributed to the fact
that the NIAs came from pilot scale batches “as opposed
to optimized formulations” but “this does not
mean that the data are unsuitable for use in an IVIVR” [see para 57].
[71]
Dr. González answered Dr. Taft’s
concern about the IVIVR technique not being peer-reviewed or generally accepted
in the scientific community in the following way:
58. At paragraphs 118 to 127 of his
report, Dr. Taft provides the opinion that the IVIVR technique that I used in
my prior report was not peer-reviewed or generally accepted. However, I know
from my personal experience in the pharmaceutical industry that, while IVIVRs
may not be the subject of large numbers of journal articles, IVIVRs are
routinely used within the pharmaceutical industry, and have been used since the
early 1980s during formulation development to direct the modification of
formulations. An IVIVR allows a formulator to decide if a modified formulation
is likely to be successful in a bioavailability study, and thus whether to test
in a human population.
59. During formulation development, it
would be rare for an IVIVC to be available for an early formulation, and it is
not practical to conduct an in vivo bioavailability study for each
formulation prepared during the development of a product. Rather, in vivo
data will only be obtained for a select number of formulations, and an IVIVR
will be used to identify a bio-relevant dissolution test to identify additional
formulations that would be expected to be bioequivalent to, or to have a better
drug absorption profile, for example, a longer tmax than, a
reference formulation.
[72]
Dr. González’s reply also took issue with
Dr. Taft’s application to the IVIVR model of a 10% prediction error
threshold used in IVIVCs. His less than compelling response was that the IVIVR
model would be expected to throw off a higher error rate than an IVIVC such
that “a prediction error of 10-20% [would] be good for
an IVIVR”. He summed up the point in the following way:
73. For these reasons, it is not at
all surprising that an estimate of prediction errors with an IVIVR would differ
from those typically seen with a more rigorous and robust IVIVC. The intent of
an IVIVR is to provide assurance of the likely performance or bioequivalence of
a test formulation when compared to a reference formulation without requiring
the need for larger amounts of in vivo data, and also allowing for a
greater range of differences between formulations.
[73]
Dr. González responded to the absence of
NIA bioequivalence data in the fed state by pointing out that because
Apo-Omeprazole is bioequivalent to LOSEC in the fed state, and because the NIA
formulations were similar to LOSEC and Apo-Omeprazole, the NIAs would also be
expected to be bioequivalent in the fed state.
[74]
Under cross-examination Dr. González
accepted that the best means of establishing bioequivalence is through a
well-designed clinical study [Transcript p 1235]. Dr. González also
stated that one does not “run a bio study with some
little pilot formulation. You want to make sure that you have a really good
formulation before you get into a bio study” [Transcript p 1241].
[75]
Dr. González was questioned closely on his
view that, because the NIA formulations were similar to LOSEC and Apo-Omeprazole,
they would be expected to behave in the same way. He conceded that he was not
a drug formulator and he was clearly out of his depth with respect to this
issue, as can be seen from the following exchange at Transcript p 1307:
Q. Moving away from the compression
forces to the ingredients: In your view, these formulations are similar
because they have the same ingredients?
A. Yes. Omeprazole, mannitol,
magnesium hydroxide and povidone, yes.
Q. Let's look at the first
experimental formulation, 15-1214B. That doesn't have magnesium hydroxide. It
doesn't have povidone?
A. That is true.
Q. Let's look at the third one.
A. Qualitatively, that one is
different.
Q. The third one, that is different
too. Fifteen -- I think it should read "12"?
A. Povidone is missing.
Q. Next one, the ingredient is
missing?
A. "Magnesium hydroxide" is
missing. Right.
Q. Next one, an ingredient is
missing?
A. That is true.
Q. Next one, as well, 6108-288C. So
many of these formulations do not have the same ingredients. Correct?
A. That is true, but I am not using
any of these formulations to arrive at the IVIVR. I am using the dissolution
profiles from these formulations to see how well they fall within the other two
dissolution profiles.
Q. You just said, Dr. González, that
you were relying on the ingredients being the same to assume that they would
have the same relationship.
A. Yes, I did say that.
Q. So you can't make that assumption?
A. Not for all of these formulations,
no. On the next page, the other four all do have the same ingredients and, in
fact, even the same percentage, so I don't know how --
[76]
Dr. González was asked about the
differences between IVIVCs and IVIVRs. He referred to the IVIVC as “a predictive mathematical model” and conceded that
the IVIVR “doesn’t have such clout” [Transcript
p 1313]. The IVIVR is less reliable in predicting bioequivalence
[Transcript p 1315]. Unlike the quantitative information produced by an IVIVC,
the IVIVR shows only a “qualitative”
relationship [Transcript pp 1314, 1320-21]. Establishing an IVIVR would
not be accepted by a regulator as evidence of bioequivalence because it does
not produce the required quantitative data [Transcript p 1321]. Rather,
IVIVRs are typically used as a directional screening tools to guide formulation
development. They are not a replacement for biostudies [Transcript pp
1355-56].
[77]
The predictive value of an IVIVR was further
explained by Dr. González in the following exchange at Transcript p 1359:
Q. But you didn't have a biostudy for
a single one of the experimental batches?
A. No.
Q. Do you agree with Devane and
Butler that in vivo data for the experimental batches would permit a reality
check on your IVIVR model?
A. Well, I would agree that at some
point in time, Apotex would eventually run a biostudy on one or two of those
formulations. But that it needs to be done for them to select a formulation to
pursue, I don't think you have to run a biostudy at that stage.
[78]
In another exchange Dr. González refused to
accept a characterization by Devane and Butler that IVIVRs have “limited value”. He answered by saying that IVIVRs
produce “some predictive value, that your dissolution
shows a relationship to in vivo absorption” [Transcript p 1361].
Even in the absence of biostudy data for the NIA formulations, Dr. González
expressed the opinion that “there is a good chance that
they are bioequivalent” [Transcript p 1370] and “[s]ome of them appear to be bioequivalent on the basis of
IVIVR or should be bioequivalent on the basis – of the fact that their dissolution
falls within the dissolution of the product that we are testing” [Transcript
p 1371]. And further at Transcript pp 1372 and 1378, he stated:
We have a set of
two dissolution profiles. Now I have a bunch of formulations that have a
similar mechanism of release or at least what I assume is a similar mechanism
of release. There is a good chance those are going to fall -- that the ones to
fall within the two dissolution profiles will have a good chance for
bioequivalence. That is it.
…
That is exactly
what I did actually. I took two extreme batches that were bioequivalent, and
now I am hoping that these other ones will fall in there, that they are
going to be clinically relevant.
[Emphasis added.]
[79]
Generally speaking Dr. Taft was a better
witness than Dr. González and he provided more reliable evidence on the
central issue of the validity of Dr. González’s IVIVR bioequivalency
analysis. I would add to this that many of the central aspects of
Dr. Taft’s written opinion and his evidence in Chief were left
unchallenged on cross-examination. Furthermore, some of the issues addressed
in Dr. Taft’s responding report were not answered in Dr. González’s reply.
The failure to fully engage with Dr. Taft’s opinions supports an inference
that his unchallenged evidence was unimpeachable.
[80]
By way of example, Dr. Taft was concerned
about the high variability in the Apotex biostudy data for Apo-Omeprazole (what
he called a “wide dispersion” of data around the
mean result). According to Dr. Taft, data variability is an important
factor in the establishment of a valid IVIVR. He addressed this point at Transcript
p 4033:
Q. Going back in your report,
paragraph 106, given those features, what does that tell you about the
reliability of using that data to try to generate an IVIVR?
A. Again, as I have mentioned in
paragraph 105, according to Cardot and Davit, in that situation where the mean
curve does not reflect the individual behaviour, IVIVC is not recommended. In
that context and looking at what we just talked about of the relative data from
the OMCP10 that seems to match what is being described by Cardot and Davit, it
is my opinion that using an IVIVR based upon biostudy OMCP10 to predict
bioequivalence is unreliable.
JUSTICE BARNES: Does it matter that what
Dr. González was looking at was not an IVIVC but rather than IVIVR? Does that
change anything? And if so, how?
THE WITNESS: In my opinion -- I believe
this is the opinion the scientific community -- the only surrogate
bioequivalence is a level A IVIVC. You have asked me the question. Essentially,
what Dr. González, in my opinion, is attempting to do is to use an IVIVR as if
it was a level A IVIVC.
[81]
Dr. Taft also spoke to the problem of
variability of subject data and the corresponding potential for error in
relying on the mean to support an IVIVR. According to Dr. Taft the
variability of the data called into question the reliability of the mean as a
measure of absorption or dissolution [see Transcript p 4036].
[82]
On the issue of using IVIVR generally to predict
bioequivalence, Dr. Taft testified as follows at Transcript p 4044:
Q. Turning to the next point, this is
more broadly to the extent that that technique has been generally accepted in
the scientific community for establishing bioequivalence. What are your views
there?
A. In the first case, for example, if
it is not available in the peer-reviewed literature, you can certainly look
towards regulatory agencies and their opinions and views through guidances and
other things.
In the documents
that I have reviewed not only in this case but in my day-to-day professional
activities, I have never come across a guidance document that would support
using an IVIVR to establish bioequivalence of alternate formulations.
[83]
It is of some significance that in answer to
questions from me, Dr. Taft attributed some value to the IVIVR technique but
only as a rough screening tool [Transcript p 4045]:
JUSTICE BARNES: Before you go there, to run
this to ground a little bit, it strikes me from what I have heard so far that
IVIVR is a "recognized technique," if I could put it that way, in the
pharmacokinetic world. It has some value in some places.
THE WITNESS: Yes, Your Honour, it does.
JUSTICE BARNES: Yes, so it is recognized as
providing some scientific value to some sort of an analysis. Where does it fit
in the scheme of things then?
THE WITNESS: I believe that the Devane
paper provided some background on the use of IVIVR in formulation development.
Certainly formulators will attempt to rely upon in vitro data to establish or
to move products forward in screening. It is not unlike what I do in my
day-to-day activities, looking at in vitro pharmacokinetic parameters and
relating them to in vivo.
The issue really
becomes, in the case of using an IVIVR, to establish bioequivalence. That is,
in my view, a totally different application that essentially what you are
attempting to do is use the principals [sic] of a level A IVIVC to make that
determination, and that is my point.
[84]
This evidence is not materially different from
Dr. González’s ultimate trial testimony.
[85]
Dr. Taft went on to calculate the internal
prediction error rate associated with Dr. González’s IVIVR techniques – a
form of validation Dr. González failed to employ. Dr. Taft found a
prediction error range between 18.7% and 36.6% – well beyond the validation
threshold of 10 to 15%. This led Dr. Taft to reasonably conclude that
Dr. González’s IVIVR model, based on mean data, was not able to capture
the predicted profiles for the test subjects [Transcript p 4052].
[86]
Dr. Taft repeated his point that an IVIVR
model cannot be used to predict bioequivalency in the fasted state let alone
the fed state [Transcript pp 4052-53]. On this issue, I agree with
Dr. Taft that a prediction of bioequivalency in the fed state cannot be
extrapolated from data observed in the fasted state. Dr. González had no
data to support his opinion. That opinion rested only on an assumption that
the NIA formulations were sufficiently similar to LOSEC and to Apo-Omeprazole
that the ingestion of food would not make any difference to their
bioequivalency. The firmness of that view wavered under cross-examination
where Dr. González conceded the difficulty of predicting food-effects on
formulation bio-availability. He ended with the equivocal statement that one “may be able to predict how things are going to work out”
[Transcript p 1327].
[87]
The far better evidence on this point came from
Dr. Taft. He described the problem of predicting bioequivalency in the
absence of a biostudy in the fed state. The presence of food in the stomach,
he said, can “influence the absorption of a drug or
drug formulation, particularly for an enteric-coated product”
[Transcript p 4000; see also Transcript pp 4052-53]. This evidence was not
challenged under cross-examination.
[88]
Apotex’s failure or inability to conduct NIA
biostudies in the fed state represents a large gap in its case for bioequivalency.
That is so because, if Health Canada required bioequivalency data for any of
the NIAs, biostudies in the fed and fasted state would have been necessary just
as they were for Apo-Omeprazole [see Exhibit AZ 158 at p 1].
[89]
Dr. González failed to counter much of
Dr. Taft’s evidence about the validity of Dr. González’s methods. I accept
Dr. Taft’s concerns about the variability of the data employed by
Dr. González and the corresponding potential for error. Dr. González
should also have run a validity analysis of his own and I reject Apotex’s
criticisms of Dr. Taft’s approach to that issue. I also agree with
Dr. Taft that the error range for Dr. González’s analysis exceeded
acceptable levels.
[90]
The above-noted methodological problems are
sufficient on their own to wholly undermine Dr. González’s prediction of
NIA bioequivalency. But there is a more fundamental problem with Dr. González’s
use of IVIVR as a predictive tool – a problem that Dr. González
acknowledged up to a point. In his trial testimony Dr. González retreated
somewhat from his firmer written opinion.
[91]
In his first report Dr. González concluded
that eight of the Apotex NIA formulations “would be
expected to be bioequivalent to Losec® and Apo-Omeprazole” [Exhibit APO
42, para 65]. However under cross-examination, he conceded that IVIVR is a
directional or screening methodology that only showed the selected NIAs to “have a good chance for bioequivalence” [Transcript p
1372]. Given the inherent limitations of IVIVR as described by Dr. Taft,
I reject the suggestion that the IVIVR analysis carried out by Dr. González
supports a balance of probabilities finding of bioequivalency. Indeed, the
technique in its present form has only a limited value in the area of pre-formulation
selection or screening. It produces nothing of quantitative value and Apotex’s
attempt to extend its reach is unjustified. In these circumstances Dr. González’s
bioequivalency opinion does not rise above the level of speculation. It is
certainly a wholly inadequate proxy for the kind of data required for
establishing NIA bioequivalency necessary for regulatory approval. Furthermore,
it is an unreliable platform for drawing an inference of bioequivalency. In
the result, I find that Apotex has failed to establish that the NIA
formulations assessed by Dr. González are, or would be seen by a regulator
to be, bioequivalent to either LOSEC or Apo-Omeprazole.
[92]
Apotex contends that it could not conduct human
clinical trials for its proposed NIAs on ethical grounds. According to this
view, because the NIAs were only developed for litigation and no potential
health benefits would result, the risks to human subjects would always outweigh
the parochial objectives of the work. AstraZeneca disagrees and argues that
human clinical trials could have been conducted. AstraZeneca also says that
Apotex should, at a minimum, have sought permission from its research ethics
board to conduct these studies. By failing to even ask, Apotex failed to
produce definitive evidence on the point and should not be the beneficiary of
any lingering doubt. Whatever the outcome, the issue, AstraZeneca says, would
be conclusively resolved, rendering any ex post facto ethics opinion on
the point effectively moot.
[93]
Each party called an eminent ethicist in support
of its position. Apotex relied on the evidence of Dr. Michael McDonald and
AstraZeneca presented evidence from Dr. Charles Weijer. Dr. McDonald
testified that a properly qualified research ethics board would not have
authorized human clinical studies in these circumstances. Dr. Weijer came to
the opposite conclusion.
[94]
There is some attractiveness to AstraZeneca’s
argument that opinion evidence on this issue ought to be excluded in the
absence of a research ethics board ruling. I am not convinced, however, that
AstraZeneca’s suggested approach would necessarily have resolved the matter –
at least if the answer provided by the Apotex research ethics board was in the
negative. In that event it would remain open to AstraZeneca to argue that the
decision was self-serving or wrong. In short, a fact-based negative ruling was
unlikely to be a complete and final answer to the question of the propriety of
conducting human trials. That is not to say, however, that the approach
asserted by AstraZeneca would not have been helpful in determining whether the
administration of the Apotex NIA formulations to humans would be unethical and
would never be authorized. An actual decision on-point would certainly have
provided relevant and likely probative evidence.
[95]
I do not accept Dr. McDonald’s point that
Apotex would have been ethically constrained from even asking its research
ethics board if a biostudy could be carried out. Dr. McDonald testified
that the circumstances of this case were novel and he posed the question: “how does one deal with a novel case?” [Transcript p
2517]. In the absence of clear guidelines and where there are opposing views
about the standards of ethical review to be applied, it cannot be the case that
Apotex is entitled to make a pre-emptive and potentially self-serving decision
not to approach its research ethics board for direction. Apotex could and
should have sought direction from its research ethics board in this case.
[96]
It does, however, seem doubtful that Apotex
would have been authorized to conduct human biostudies in connection with its
hypothetical NIAs, given that the sole purpose of the experiments would be to
advance Apotex’s litigation interests.
[97]
For this, I accept Dr. McDonald’s views
over those expressed by Dr. Weijer. In particular, I accept
Dr. McDonald’s evidence concerning the risk-benefit ratio and the ethical
requirement that the importance of a research objective outweigh the risks
faced by the research subjects. In the case of the Apotex NIAs the risks may
have been slight but they were not unworthy of consideration and concern.
Where the only benefit of the trials would be to further Apotex’s business
interests (in contrast to a public good), it is doubtful that ethical approval
could have been obtained.
[98]
I also disagree with Dr. Weijer that a
biostudy involving the human ingestion of an unapproved drug carrying
anticipated risks and side-effects and done solely for private commercial or
litigation purposes ought to the subject of less rigorous standards or some
form of diminished risk-benefit analysis. His idea that there is an underlying
public interest in the outcome of private litigation has no appeal. Courts of
law decide cases on the basis of available evidence and, in many cases, the
evidentiary record is incomplete. It is inconceivable that any Court could or
would ever order a litigant to conduct human testing to answer a question
relevant to the outcome of a case. At most, an adverse inference can be drawn
where a party fails to advance evidence that is potentially available to it.
[99]
The fact that Apotex probably could not ethically
conduct human bioequivalency studies to prove the efficacy of its unapproved
NIAs does not, however, assist it in advancing its substantive case. At most
this barrier to human testing prevents the Court from drawing an adverse
inference. Human bioequivalency studies are an important means to prove the
viability of a NIA and are often required for obtaining regulatory approval.
The indisputable fact remains that those tests were not done, leaving a
significant gap in the evidence as to whether any of Apotex’s NIAs could have
been shown to be bioequivalent to LOSEC or Apo-Omeprazole and approved for sale
in Canada or the United States.
[100] Apotex contends that its testing data are sufficient to meet its
burden of establishing that the asserted NIAs were commercially viable
substitutes for Apo-Omeprazole. One of the key requirements for proving that
viability concerns the stability of those formulations (i.e., did they have an
acceptable shelf-life both from a regulatory and commercial standpoint?).
[101] Apotex’s stability case was based on data it obtained from in-house
stability testing commenced in late 2015, as interpreted by Dr. Kwok
Chow. Dr. Chow was accepted as an expert in pharmaceutical product
development and drug delivery systems including the design, execution and
management of formulation screening, including with respect to the physical and
chemical properties of drug substances and excipients, including, specifically,
stability thereof.
[102] Dr. Chow was asked by Apotex to review its in-house stability
test data for fifteen NIA formulations to determine if they would be expected
to have sufficient stability to be useful as pharmaceutical products.
[103] Dr. Chow confirmed in his first report dated September 16, 2016
[Exhibit APO 130] that in-house stability testing of the sort conducted by
Apotex is usually done under the research conditions recommended by the
appropriate regulatory bodies and/or the International Council for
Harmonisation of Technical Requirements for Registration of Pharmaceuticals for
Human Use [ICH] guidelines. Those guidelines set out the minimum data
requirements for a new drug submission under long-term (12 months),
intermediate (six months) and accelerated (six months) storage with varying temperature
and humidity conditions. Throughout the assessment period the formulations are
tested for assay values and impurity levels [see Chow Report #1, Exhibit 1].
[104] Dr. Chow's first report was based on stability data for three
different formulation groups (A, B and C). Group A formulations (four
formulations) had data covering at least eight weeks. Group B (nine
formulations) had some four-week data. Group C (two formulations) had only the
initial assay data.
[105] Dr. Chow reviewed Apotex's testing protocols and concluded they
met ICH guidelines.
[106] Notwithstanding the preliminary nature of the data, Dr. Chow
professed in his first report to be able to predict the stability outcomes for
some of the formulations. According to Dr. Chow two of the Group A
formulations "are expected to meet the acceptance
criteria at the end of the stability program" for total impurities
[see para 86]. For one of those formulations, he predicted a target shelf-life
of two years. For the others, the data was insufficient to support a
definitive two-year shelf-life.
[107] For four of the Group B formulations, Dr. Chow expressed the
view that they "may well meet acceptance criteria
at the end of the stability program and achieve a target shelf-life of two
years" [see para 115(a)]. For the other four Group B formulations,
Dr. Chow expressed uncertainty related to the interim impurity data
obtained. The last Group B formulation failed to meet the required dissolution
criteria.
[108] According to Dr. Chow both of the Group C formulations "meet the specification" [see para 124]. Because
of the compositional similarities to Apo-Omeprazole, Dr. Chow expressed
the view that Group C formulation MR8620E1 "would
be expected to have sufficient stability since Apo-omeprazole is an approved
product" [see para 125].
[109] Dr. Chow concluded his first report by stating that all of the
NIA formulations would be readily scalable to commercial quantities.
[110] Dr. Chow updated his stability opinion in his report of January
6, 2017 by reviewing the stability data developed since his first report
[Exhibit APO 131].
[111] For the two remaining Group A formulations, Dr. Chow dismissed
an assay anomaly for one (despite not knowing the cause) and higher than
expected impurity levels at 24 weeks under accelerated conditions for both. However,
he did reduce his shelf-life expectation for both formulations to 18 months.
He concluded that there was "a high probability
that both formulations will meet acceptance criteria for at least an 18-month
shelf-life" [see para 30].
[112] Of the Group B formulations, two were predicted to meet acceptance
criteria for a two-year shelf-life [see para 47(a)], three had higher impurity
levels and a predicted shelf-life of 18 months [see para 47(b)] and three did
not meet specification criteria under accelerated conditions; nevertheless, one
of those was predicted by Dr. Chow to have "a
high probability" of an 18-month shelf-life while the other two
were "less likely to meet specification
criteria" [see para 47(c)].
[113] Dr. Chow reviewed the 16-week data for the Group C
formulations. He dismissed an assay anomaly for one as being likely caused by
poor sample preparation. He concluded that both were expected to continue to
meet the requirements for drug release and "there
is a high probability that both formulations will meet acceptance criteria for
a 24-month shelf-life" [see para 60].
[114] AstraZeneca responded to Dr. Chow's opinions through
Dr. Martin Davies. Dr. Davies had testified in the liability phase
of this case and he was qualified to testify as an expert in physical
chemistry, pharmaceutical stability, drug delivery and biomedical surface
chemistry and in the development, testing and analysis and characterization of
pharmaceutical formulations.
[115] Dr. Davies expressed the view that Dr. Chow's stability
predictions were unsound because the supporting data were incomplete and, with
one exception, the small-scale test batches were unsuitable to predict the
stability of large scale commercially-produced products. Dr. Davies
identified a number of problems in the scale-up of production that might be
barriers to success and he identified testing procedures that failed to meet
the applicable stability guidelines. These included the failure to test more
than one batch (to reduce the effect of batch variability) and the general
failure to use pilot scale test batches (one-tenth commercial scale). Only one
formulation met the pilot scale standard.
[116] Dr. Davies pointed out that Dr. Chow's first report was
based on incomplete stability testing and, without completed testing, "there is insufficient data to reliably predict
shelf-life" [Exhibit AZ 137, para 79]. He was particularly
critical of Dr. Chow's extrapolation for Group C formulations for which
only early data existed. He also criticized Dr. Chow for assuming linear
degradation kinetics in the absence of empirical support and he challenged
Dr. Chow's treatment of data anomalies.
[117] Dr. Chow's reply report addressed Dr. Davies' comments
about the sufficiency of the incomplete stability data in the following way:
62. Many of the differences in the
opinions of Dr. Davies and me appear to come down to the different perspectives
from which we have looked at the stability studies conducted by Apotex. While I
have looked at the stability studies to assess whether or not the formulations
being studied would likely exhibit sufficient stability to be useful as a drug
product, Dr. Davies appears to have focused on whether the studies, standing
alone, could be presented to a regulatory agency, such as Health Canada, as
part of a drug submission. I do not disagree with Dr. Davies that the Apotex
studies in their present form would not be complete for filing with a
regulatory authority as part of a drug submission. However, this does not mean
that the studies fail to show whether the formulations are likely to have
sufficient stability for use as a drug product. In general, by focusing on
regulatory filings, Dr. Davies has failed to consider the realities of
formulation development, and how stability studies are conducted and used in
the pharmaceutical industry.
63. In light of Dr. Davies’ opinions,
it should also be understood that, as part of the formulation development
process, formulations are initially prepared on a laboratory or pilot scale.
Based on the test data obtained for these formulations, which will include
abbreviated (as opposed to full-length) stability studies, formulators make
informed predictions as to whether formulations are expected to possess the
properties that are desired for the final dosage form. Scale-up of a formulation
will only be conducted for formulations that are expected to be successful
based on this initial testing of material prepared on a small-scale. For
formulations that show sufficient stability in the lab or pilot scale, the
formulator’s expectation is that the formulation will also be stable when made
on a larger scale using the same manufacturing steps.
64. Based on my experience in the
pharmaceutical industry, I cannot recall an instance where the volume of
information that has been collected for the Apotex formulations would not be
considered to be suitable for deciding which formulation or formulations would
be expected to be successfully scaled-up.
[Footnotes omitted.]
[118] Dr. Chow accordingly accepted Dr. Davies’ point that the
standards and methods employed by Apotex would not meet regulatory
requirements. He said that, from a product development perspective, multiple
batch testing was not required. He made the same point concerning the small
lab tests batches. This, he said, was “a regulatory
concern and does not relate to whether or not a formulation is stable, or
expected to be stable” [see para 67].
[119] In response to Dr. Davies’ criticism that Apotex’s preliminary
data were incomplete and insufficient to support a stability prediction,
Dr. Chow stated, at para 73:
… At paragraph 60 of his Expert Report, Dr.
Davies comments on the fact that, at the time of my September 6, 2016 Expert
Report, the stability studies for the Group A, B and C formulations were not
complete. As discussed above, this issue appears to be directed more to whether
the studies could be submitted to a regulatory agency at that time, not whether
the available information could be used to determine whether a formulation was
likely to be stable throughout the duration of the stability program. However,
as noted above (see paragraph 63), it is standard practice to use preliminary
data from stability studies in order to determine which batch or batches to
scale up.
…
74. In his Expert Report, Dr. Davies
takes issue with my extrapolation of stability data, noting that the ICH Q1A
stability guideline provides specific conditions for the extrapolation of
stability. Again, Dr. Davies is confusing the regulatory guidelines for
submitting stability data as part of a drug market application with the ability
to predict whether a formulation is likely to be stable for the duration of a
stability program. For a regulatory submission to Health Canada, Dr. Davies is
correct that complete studies should be submitted, and that extrapolated data
can only be relied upon in certain circumstances. However, I know from my own
personal experience in the pharmaceutical industry that, during the development
of a formulation, a portion of, or abbreviated, stability studies are routinely
used to predict whether a given formulation is likely to have sufficient
stability in long-term testing. During drug development, the time requirement
to await the completion of a 6-month accelerated stability study is generally
not available to a formulator, which makes predictions on less data a necessity
to determine which formulations to progress.
[Footnotes omitted.]
[120] Dr. Davies’ report dated March 23, 2017 took issue with
Dr. Chow’s updated stability opinions [see Exhibit AZ 138].
Dr. Davies maintained that the test data continued to be incomplete and
the test methods continued to be deficient. He also noted that some of
Dr. Chow’s initial stability predictions proved, on further testing, to be
unsound.
[121] Dr. Davies also noted a further testing anomaly in the form of
progressive modifications to Apotex’s assay methods which, over time, increased
the assay results. These changes to sample agitation times indicated to
Dr. Davies two potential problems: either the omeprazole in the samples
was not, in the early testing, fully extracted or the formulations were
undergoing physical changes making extraction more difficult over time (or a
combination of both).
[122] Dr. Davies also drew attention to Dr. Chow’s initial
stability predictions for two of the Group B NIA formulations which, on further
testing, proved to be unsound. He also pointed to decreased assay values and
questionable impurity levels for several of the remaining formulations. He
challenged Dr. Chow’s 18-month shelf-life predictions for several of the
formulations as speculation.
[123] In reply Dr. Chow agreed in theory with the need to validate
assay testing methods but said in formulation development it is usually not
done [see Transcript p 3107]. Full validation, he said, was required
later in the process [Transcript p 3108]:
For example, you
want to sell the product. It is a regulatory requirement to validate the
method, so that you can sell the product -- make, test and sell the product.
Q. Surely you are validating for a
reason beyond just regulatory requirements?
A. Certainly, because a product needs
to be tested, and you want to have methods standardized, and you want to make
sure that the method works.
He also accepted that assay testing methods
should be kept constant “[a]s much as possible”
although “[i]n development, we tend to revise methods
from time to time” [Transcript p 3109].
[124] When questioned about Apotex’s initial low assay values,
Dr. Chow had “an idea about what could cause it”
[Transcript p 3113] and was capable of designing an experiment to identify
the exact cause [Transcript p 3114]. Nevertheless, he was not asked by
Apotex to determine the cause.
[125] Dr. Chow also acknowledged the following points:
(a)
he never calculated the degree of experimental
error in the Apotex testing but he had an “idea”
[Transcript p 3115];
(b)
he was unable to measure batch-to-batch
variations among NIA batches because only one batch of each was ever made
[Transcript p 3116]. Single batch testing did not meet the regulatory
standard [Transcript pp 3117-18];
(c)
batch-to-batch variation will have an impact on
shelf-life [Transcript pp 3118-19] and affects the degree of confidence “on production batches” [Transcript p 3119]. “[I]f you want to estimate shelf life accurately, you need
multiple batches” [Transcript p 3122];
(d)
when asked about his level of confidence in the
absence of empirical data, he gave the following answers [Transcript p 3123]:
Q. When you are referring to
"confidence," though, you are referring to it in a qualitative sense?
A. It is looking at the trend, yes. You
can say that it is somewhat eyeballing and trending.
Q. So the answer to my question is
"yes"?
A. Yes.
Q. You didn't quantify the confidence
level of your predictions?
A. The term "quantify"
means what?
Q. That you know the degree, the
percentage of confidence in a statistical way?
A. From a statistical, 95 per cent
confidence interval, I did not do it.
Q. You didn't do it for any
confidence interval?
A. No.
(e)
Dr. Chow made a “judgment
call” about the linearity of degradation kinetics for the NIA formulations
[Transcript pp 3185-3186];
(f)
Dr. Chow admitted being “a bit optimistic” about some of his initial stability
predictions [Transcript p 3192]. When challenged about his initial
comparative assessment for two formulations that were similar, he gave the following
answers [Transcript p 3193]:
Q. So your predictions based on
linear degradation kinetics for all three of these formulations were incorrect?
A. Based on these three, yes, I was
too optimistic. Those who fail continue to fail. Some of those that work
still work. It is only one impurity I failed to address that, and that I can
agree.
Q. In your opinion, experimental
batch 151218A is expected to have similar stability to experimental batch
151215A?
A. I said in my report that it would
have a similar stability profile and, also, in my report I -- responding report
based on the 16-week data, accelerated condition, 218 is performing better than
the 215.
Q. At the time of your first report,
you were prepared to predict stability of 151218A based on 151215A?
A. Let me check my wording before I
--
Q. Second sentence, paragraph 125, of
your first report.
A. I state it would have similar
stability.
Q. Now we know that 151215A no longer
meets all the stability requirements?
A. It did not meet the stability requirement
for one impurity, which is unknown impurity. It could be characterized later
on.
Q. You are no longer using that batch
to predict 151218A?
A. Well, I use that batch as a
baseline, the 215A, for predicting 218A, because of the similarity in terms of
composition.
Q. It turned out they don't have
similar stability?
A. It is difficult to say what is
"similar." One is slightly better than the other. Does this say
"similar"? I will say it is still very similar. They all got the
good side in terms of all studies -- all tests tested, except that the 218A is
slightly better in terms of impurity.
Q. Well, 151215A is failing one of
the specification requirements.
A. Yes, but if you look at the whole
picture or the whole profile, both lots are performing very well, except for
the one unknown impurity, and the unknown impurity level is still at a very low
level. I agree that they are not making the acceptance criteria.
It does not
necessarily mean that product is not useful as a pharmaceutical product. My
mandate was to provide opinion whether it is a useful, potentially useful
pharmaceutical product. I know that I may be more optimistic about stating the
shelf life of that.
Passing the
acceptance criteria that I would suggest, I would feel that it would likely
make 24 months, but not making that unidentified impurity would not necessarily
negate the point that it may still make a pharmaceutical product.
(g)
all of the Group A formulations ultimately
failed to meet one or more of Apotex’s specification criteria [Transcript
pp 3195-96];
(h)
he agreed that an increase of 20 minutes in the
sonification of test samples is a significant change to the assay test method
and a 5% increase or more in assay results was a significant change [Transcript
p 3259]. Without data the significance could not be determined
quantitatively [Transcript p 3269]. A possible explanation for an
increase in extraction time could be physical changes to the sample [Transcript
p 3278];
(i)
under Good Manufacturing Practice [GMP]
protocols, the assay method cannot be changed [Transcript p 3281];
(j)
different assay extraction methods required for Apo-Omeprazole
and the NIA formulations “could” reflect the
difference in behaviour [Transcript p 3282]; and
(k)
Apotex did selective retesting of its NIA
samples based on the “noticeably lower” assay
results [Transcript p 3283]. This may not be an ideal approach but it is
not uncommon for suspected samples [Transcript p 3285].
[126] Dr. Davies was cross-examined at length on his two reports.
When asked about the propriety of overlooking an anomaly in accelerated
stability data in favour of acceptable intermediate and long-term data, he gave
the following responses [Transcript p 3538]:
Q. If you look at the guideline I
have just given you -- let's try to expedite it. You accept it, too, provides
that predictions can be made from intermediate conditions when there are
significant changes in accelerated conditions?
A. I accept that when it is
justified, when you can provide evidence through investigation why you
would overlook the accelerated data and rely on the intermediate and long-term
data.
[Emphasis added.] [See also Transcript p 3549.]
[127] Dr. Davies’ concern was that Dr. Chow had provided no
evidence to support such an approach with respect to the NIA formulations that
presented in this way. Dr. Davies did accept that extrapolations of the
sort made by Dr. Chow can be useful in the early stages of drug
development and for selecting the best candidates [Transcript p 3546].
[128] Dr. Davies also challenged Dr. Chow’s assumption that all
of the NIA formulations had linear degradation kinetics. Dr. Davies
observed that at least two of the formulations had non-linear degradation
profiles [Transcript p 3568] and for others it was not possible to say
without complete data [Transcript pp 3573-74]. He also pointed to prior
art that showed omeprazole formulations “are prone to
accelerated degradation” [Transcript p 3577].
[129] A significant issue of concern for Dr. Davies was the discovery
that Apotex had made a series of changes to its testing protocols for the NIA
samples.
Because of apparent problems in obtaining complete dissolution of those
samples, Apotex substantially increased the agitation and sonification times
from those employed in its earliest assays.
[130] In the face of those changes and in the absence of validation of the
extraction method for the NIA formulations, Dr. Davies considered the data
unreliable. The problem as Dr. Davies saw it was explained in the
following exchange [Transcript p 3621-23]:
Q. With respect to the increased --
let's just first step back. The purpose of the assay is to accurately
determine the amount of omeprazole in the pellets. Correct?
A. That would be the purpose of an
assay. That would be what you would hope the assay would do.
Q. That is what Apotex was aiming to
do in its study?
A. Sure. In the context of this, if
you are changing your extraction method because you don't like the look of the
results, if you are changing the assay method because you think the results are
too low, that suggests a number of things. One, that the sample is changing,
that it is becoming more difficult to extract. Or two, there is a problem with
that extraction itself. That is just the least of them.
Q. Sample is changing or there is a
problem with the extraction. I got you. Let's park that for a second and walk
through this slowly with me, please. The purpose of an assay test is to --
that is the aim, to accurately determine the amount of omeprazole in the
pellet?
A. I wouldn't disagree with that.
Q. That is to say you would agree
with that?
A. That is the purpose in this case,
is try to measure the amount of omeprazole that is present within the
alternative -- the batch of the alternative formulations.
Q. You say that if you have failed to
fully extract the omeprazole from the assay, then you are unable to reliably
and accurately measure the omeprazole content?
A. I would agree with that.
Q. The purpose of the agitation in
the assay is to try and get the omeprazole to properly dissolve within the
dissolution media?
A. Properly dissolve in the solvent
for extraction.
Q. Right. If it doesn't properly
dissolve, you don't have all of the omeprazole. Therefore, you are not going
to get an accurate result?
A. That is true. You don't know.
That is true if you -- you don't know.
Q. Your objective in conducting an
assay is to get the omeprazole dissolved so that you can ensure you have closer
to the real result?
A. It is, but usually across -- if
you are going to compare samples across time, you would use the same assay, a
validated approach. You wouldn't need to change the assay over time. That is
what begs the question: What if you use 90/60 on those initial results? What
would we [sic] the value of those? We don't know. That is why you can't
assume -- to your point -- that they have measured accurately in the initial
results the value of omeprazole. We don't know.
[131] According to Dr. Davies, the increases in agitation and
sonification times created the possibility that Apotex’s earlier sample assays
were under-reporting the levels of omeprazole. With that uncertainty it was
not possible to do a valid comparison with later assays where optimum
dissolution was obtained [Transcript p 3624].
[132] When it was then put to Dr. Davies that another possible
explanation for the need for longer sample preparation time was that the
samples were changing over time, he gave the following answers [Transcript p
3631]:
Q. The other possibility which you
also raise is that you may have a problem with your assay. Correct?
A. It is possible.
Q. If the sample is changing over
time, that doesn't mean you are actually losing omeprazole through the process,
that you are not catching it. It means you have a sample that is changing over
time that requires different agitation to get the omeprazole out?
A. They are not mutually exclusive.
In the context of the sample changing over time, as a formulation scientist,
you would be very concerned why that was occurring.
[See also Transcript pp 3632-36]
[133] The underlying problem with under-reported omeprazole assay values
in the initial tests is that it has the potential to mask the amount of lost
omeprazole as determined by subsequent assays. According to Dr. Davies
this cannot be known from the extant data [Transcript p 3642]. This issue
was more fully explained in the following exchange under direct examination [Transcript
p 3398-3400]:
Q. Looking at the next set of
comments you make, starting on paragraph 37, you say that:
"Apotex's
investigations cast serious doubt on the reliability and accuracy of its assay
method for measuring the omeprazole content of the alternate
formulations." (As read.)
Could you explain
why that is?
A. Yes, because these results
revealed that extending the agitation times increases the assay results. We
don't know the reason for that. It could be that the original assay time
itself was insufficient to fully extract the omeprazole from the alternative
formulations. Therefore, that calls into question all those initial results.
In a stability
program, you are going to be comparing results against each other. The
assumption is that you are analyzing each of them in the same way. But if your
actual analytical method has been changed so it can increase the amount drug
that is present, then that means you -- it makes it very difficult to compare
results produced by different assay methods.
Q. Looking at your last set of
comments on paragraph 39, you say that:
"Apotex's
modifications to the assay method during the course of the stability program
could have obscured 5 per cent or greater reductions in omeprazole content from
the initial values." (As read.)
Could you explain
how modifications could obscure such reductions?
A. Yes. The point I am making here
is because you are not treating the initial results with the same analytical
procedure, you are using less agitation than was used later in the testing,
because we know 5 per cent or greater is a significant result.
If Apotex had
tested those first set of data based on the information that we have, those results
are likely to be higher. Therefore, failing to retest -- they can't retest
because you can't go back. The very fact that they are retesting the later
results could obscure a 5 per cent or greater reduction in omeprazole content.
That is the point I am making.
JUSTICE BARNES:
Just to clarify that, if you have an artificially low initial assay result and
a true assay result later on, just clarify for me how that could obscure
degradation or a "poor stability outcome," if I can put it that way.
THE WITNESS: I
don't think we know what the true result later on is, Your Honour, because
whether it is September or October or November, you are changing the assay as
compared to the first result.
To say, for
example, that you increase the results in September by 10 per cent, but you
have done nothing to the initial results -- so say you have gone up from 90 per
cent to 100 per cent in September. It was originally 90 per cent but you have
increased it by 10 per cent. The value at the beginning could be, say, 95 per
cent.
Well, you could
increase -- if you use the same agitation time for the initial results, based
on the data here, it seems to indicate you have better extraction, so that 95
result may become 105 result per cent. So now instead of having 95 and 100 per
cent -- 95 for the initial and 100 per cent for the retested sample in October
or September -- you now have the retested initial result, 105 per cent,
compared to 100 per cent. So you now have, potentially, a 5 per cent or more
change, but Your Honour will never know.
Because if you are
changing -- if you are trying to compare results all the way through when your
analytical method is changing because you recognize you are not extracting all
the omeprazole, what about the results at the beginning, which is your
baseline, where you start from? That is what makes it difficult.
[134] The fundamental difference between Dr. Chow and Dr. Davies
concerns the scientific weight that should be attributed to the stability data
produced by Apotex’s in-house testing. On this issue, I prefer the evidence of
Dr. Davies to that of Dr. Chow.
[135] Dr. Chow was forced to make predictions based on incomplete
data. The initial predictions he made were based on very early data and, not
surprisingly, some were later shown to be unsound. But Dr. Chow’s
apparent willingness to rely on preliminary and inconclusive data says
something about his credibility – albeit within the context of an early stage
drug development scenario.
[136] Like Dr. González’s use of IVIVR to predict bioequivalency,
Dr. Chow approached the problem of NIA stability as though it was a
formulation screening exercise. Although Dr. Chow acknowledged some of
the issues raised by Dr. Davies as having potential relevance to the
process for achieving regulatory approval, he felt that, for his more limited
purposes, the methods he employed and the data he reviewed were sufficient to
make some reliable predictions about the stability of the tested NIAs. My
reservations about this approach as they applied to Dr. González’s opinions
apply equally to Dr. Chow’s predictions. Early and incomplete data are
just that. They do not support an inference of regulatory and commercial
viability simply because a formulator might find them useful for internal
screening purposes.
[137] Dr. Davies approached the test data from a far more rigorous
perspective – a perspective that was generally in keeping with the standards
that would be required to commercialize the asserted NIAs. This was the better
approach because it was commensurate with the ultimate burden of proof.
[138] Apotex has attempted to justify its stability testing shortcuts and
incomplete test data largely on the strength of its claimed entitlement to a
standard of proof that is lower than the applicable regulatory requirements.
In short, it says the Court should draw an inference that at least some of its
NIA formulations would have been more likely than not to be sufficiently stable
to obtain regulatory approval and commercial success.
[139] On the evidence presented, I am not prepared to draw that inference.
[140] Although Dr. Chow’s stability analysis rests, in part, on
empirical data, the results were incomplete.
[141] To the extent that Dr. Chow attempted to minimize the
shortcomings of Apotex’s stability testing methods and the significance of the
observed data anomalies noted by Dr. Davies, I reject Dr. Chow’s
views. The concerns raised by Dr. Davies about batch variability, batch
size and the lack of validation were valid and cast doubt on the reliability of
the obtained test data.
[142] The numerous changes Apotex made to its NIA sample preparation and
its selective retesting also undermine the value of the initial assay data. I
accept Dr. Davies’ point that it is not possible to fully understand the
significance of those changes, but it was Apotex’s burden to complete its
stability testing in a way that would eliminate this type of avoidable
uncertainty. It seems inescapable to me that where the baseline assay data are
suspect, downstream extrapolations from those data are also rendered suspect.
Apotex’s attempts to rehabilitate those data by reference to the observed
impurity and mass balance data were insufficient and unconvincing.
[143] Dr. Davies is correct that Apotex’s methodological changes and
selective retesting introduced an unacceptable degree of error to the process.
The initial assay values were rendered almost worthless for comparison purposes
– at least far from a level of certainty required for my purposes. I accept
Dr. Chow’s point (concurred in by Dr. Davies) that shortcuts like the
ones employed by Apotex may be acceptable in the early stages of product
selection, but they are entirely unacceptable in the context of proving the
viability of a NIA for the purpose of this case.
[144] Apotex contends that, for regulatory purposes, it could have
piggy-backed its NIAs on the approval it received for Apo-Omeprazole. Under
this hypothesis, once Apo-Omeprazole obtained regulatory approval, it would be
a routine and simple exercise to obtain an approval for each of the NIAs.
According to this argument, Health Canada would have accepted any of the seven
now-asserted NIAs as minor variants to Apo-Omeprazole such that robust
bioequivalency testing would not have been required for approval. This idea
is, of course, wholly belied by what Apotex actually did in the face of its
knowledge that Apo-Omeprazole was likely an infringing product and it is
inconsistent with what the regulators would have accepted. Indeed, the theory
is based on an apparent assumption that the regulators would not have followed
applicable guidances nor applied robust due diligence to the approval of the
submitted NIAs.
[145] Each of the parties led evidence from a qualified witness with
expertise in Canadian regulatory affairs. Apotex called Ms. Sue Wehner
and AstraZeneca called Ms. Anne Tomalin. The fundamental differences
between these witnesses concerned the likely willingness of Health Canada to
deviate from its own guidances and whether bioequivalency testing would have
been required for Apotex’s seven remaining hypothetical NIAs.
[146] Where they differ, I prefer the evidence of Ms. Tomalin over
the evidence of Ms. Wehner. I do not accept that all of the NIAs involved
minor changes to the formulation for Apo-Omeprazole and that, with the
exception of Formulation MR8620E1, Health Canada would have accepted notifiable
change requests [NCs] for the proposed NIAs.
[147] Ms. Wehner’s analysis depended largely on her characterization
of Apo-Omeprazole as an immediate release drug, as opposed to modified release,
and on her reliance on Dr. González’s unsound bioequivalency data.
Ms. Tomalin described Ms. Wehner’s approach as follows [Transcript p 3841]:
As I understand what Ms. Wehner did, she
started with her understanding of omeprazole and that omeprazole, in her view,
was an uncomplicated product. She looked at the data supporting the changes and
felt that the data confirmed that the changes being made were not significant
changes, and from there, went to a conclusion that the type of submission that
would be required would be a notifiable change.
[148] In contrast, Ms. Tomalin’s analysis depended on Health Canada’s
published policies and guidance documents at the relevant time. While they may
not technically have the force of law, the real question is whether they were
used by Health Canada at the relevant time to interpret the applicable
statutory and regulatory requirements. I accept Ms. Tomalin’s evidence
that Health Canada did strictly follow its guidance documents over the relevant
period.
[149] I also accept Ms. Tomalin’s evidence that Apo-Omeprazole is a
modified release product, as that term is understood by Health Canada. There
is ample support for this conclusion in the guidance documents. For example,
the 1996 Bioavailability and Bioequivalence Studies document clearly
defines a modified release product as including formulations designed to delay
absorption, such as enteric-coated forms [see Exhibit E-2 to Wehner Report #1, Exhibit
APO 50, p 2]. This approach was confirmed in the later Post-NOC
Changes document which defined modified release solid oral dosage forms as
including “both delayed and extended release drug
products” [see Exhibit E-5 to Wehner Report #1, p 229].
[150] I do not accept Ms. Wehner’s evidence that a qualitative change
to either an excipient or to an enteric coating would be treated by Health
Canada as insignificant. The better evidence came from Mr. Len Arsenault
based on his experience working at Health Canada where he was manager of the
oral products division of the Bureau of Pharmaceutical Sciences.
Mr. Arsenault – now vice president of scientific affairs with Sandoz - was
called by Apotex to speak to the history of Sandoz’s omeprazole product. He
testified that Health Canada considered an enteric coated formulation to be a
modified release product for which an alteration typically required the
submission of bioequivalency data [Transcript pp 1735]. Like Ms. Tomalin,
he also testified that the switch from one kind of polymer to a different
enteric coating polymer or a qualitative change to an excipient would require
proof of bioequivalency [see Transcript pp 1742, 1744-45].
[151] I also do not accept Ms. Wehner’s evidence that a change in the solvent
used in the coating process, from water to organic solvents, would not be
considered a significant change. Ms. Wehner’s conclusion is inconsistent
with a 1997 letter from Health Canada advising that “[i]n
the case of delayed-release or other modified-release preparations whose
coatings affect drug release,” a supplement is required when a change is
made to the tablet coating [see Schedule D-5 to Tomalin Report #1, Exhibit AZ
155, p 3]. I prefer Ms. Tomalin’s evidence that these requirements would
apply because the enteric coating of Apo-Omeprazole affects drug release by delaying
the release of omeprazole until it reaches the small intestine. I also accept
that this document would be applied equally to both tablets and pellets.
[152] Finally, I do not accept Ms. Wehner’s conclusion that changes
from wet to dry granulation would not likely be considered significant. This
conclusion was based primarily on Ms. Wehner’s opinion that Apo-Omeprazole
was considered to be an immediate release drug with rapid dissolution, which I
do not accept. I prefer the evidence of Ms. Tomlin who stated that Health
Canada would have viewed the change as one requiring supporting bioequivalence
data and the submission of a supplement because it is a change that could have
an impact on bioequivalence. This was eventually expressly reflected in the
2007 guidance, which Ms. Wehner acknowledged [see Transcript p 1561].
[153] Dr. Sherman testified that he could and would have obtained a
NIA from a third-party source, but only if he was unable to develop an in-house
formulation. From this evidence I can only conclude that if a third-party
option was required, it would only have been pursued after Apotex had tried and
failed to produce and commercialize its own formulation. At best this option
would only have been pursued well into the infringing period and would have
required Canadian regulatory approval.
[154] I do accept that, in theory, there were NIAs potentially available
to Apotex from two sources. Those sources were Kremers Urban Development
Company [Kudco] and Estevé.
[155] Evidence concerning the Estevé omeprazole formulation was given by
Ms. Cinta Lacasa Pujadó. Ms. Lacasa holds a senior management
position with Estevé and is responsible for regulatory affairs, intellectual
property, pharmacokinetics and product development.
[156] Estevé is a Spanish-based pharmaceutical company. It produces both
innovative and generic products and has business relationships with both Apotex
and AstraZeneca.
[157] According to Ms. Lacasa, Estevé began to develop an omeprazole
formulation in the late 1980s. By 1994 or 1995 it had a marketable and
patented omeprazole formulation. At that time Estevé’s business model was
limited to the sale of its own innovative drugs. It was not equipped to sell
generic formulations and, thus, it supplied its omeprazole formulation only to
interested third parties. In the United States Estevé entered into a
contractual relationship to supply its omeprazole product in pellet form to Mylan.
Mylan encapsulated the pellets for sale into the United States. Mylan was also
responsible for obtaining regulatory approval in the United States, albeit with
technical support coming from Estevé. It was not until mid-2003 that Mylan
obtained United States regulatory approval.
[158] After Mylan launched the product in the United States in August
2003, Mylan and Estevé were sued for infringement. That suit was successfully
defended on the basis that the Estevé pellets did not infringe AstraZeneca’s
United States patent. After obtaining Canadian regulatory approval, Mylan began
to sell the same omeprazole formulation in Canada in 2009. Estevé assisted
Mylan with its Canadian regulatory filings in 2007 and later.
[159] When Ms. Lacasa was asked about Estevé’s hypothetical interest
in supplying its omeprazole formulation to Apotex, she said it would have been “[v]ery much interested” and would have provided the
necessary technical support for a Canadian regulatory submission [see
Transcript p 2940].
[160] Ms. Lacasa went on to say that Estevé would have needed Mylan’s
approval to sell its omeprazole product to Apotex. Mylan’s agreement was
required because the governing contract with Estevé gave Mylan exclusive
distribution rights in the United States, Canada and Mexico.
[161] Before Mylan entered the market, neither Apotex nor any other party had
approached Estevé about supplying the Canadian market. However, in the case of
Mexico, Estevé was approached by a third party in 2003 or 2004 to supply its
omeprazole product and Mylan unconditionally waived its exclusivity rights in
that country.
[162] According to Ms. Lacasa, Estevé had more than ample production
capacity to fully meet Apotex’s supply requirements. Its usual terms of sale
for similar third-party sales of its omeprazole were in the range of 25 to 35%
of net sales depending on the specific details of each case [see Transcript p
2953].
[163] Over an objection from AstraZeneca’s counsel, I allowed
Ms. Lacasa to speak to the issue of Mylan’s likely willingness to waive
its Canadian rights. She answered that Mylan had done so in Mexico and, at the
same time, it had no commercial presence in Canada. She concluded with the
statement: “[t]here could have been additional
business also for Mylan, depending on the type of arrangement that we could
have created” [Transcript p 2956]. Later she said “[e]veryone needs to gain something” [Transcript p 2994].
[164] Under cross-examination, Ms. Lacasa confirmed that Estevé’s
development of a non-infringing omeprazole formulation for supply to Mylan took
from 1995 to the point of filing a United States ANDS in 2000. It was only in
2003 that the product was actually launched into the United States. During the
five years of product development, a number of formulations were tested
involving “a lot of work” [Transcript p 2973]. Ms. Lacasa
also acknowledged that “[o]meprazole has been a very
difficult product” and a number of its bioequivalence studies failed
[Transcript p 2975].
[165] It is of some significance that, when Ms. Lacasa was asked if
Mylan would have permitted Estevé to supply its omeprazole formulation to
Apotex for sale into the United States, she observed that those companies
competed in that market. She concluded with the statement that “[t]his is something we should ask Mylan. I don’t know”
[Transcript p 2990].
[166] Based on Ms. Lacasa’s testimony, I am satisfied that Estevé had
the manufacturing capacity to supply its non-infringing formulation to Apotex
in sufficient quantities to substitute for Apotex’s sales of Apo-Omeprazole.
[167] I do not, however, accept that the evidence before me is sufficient
to establish that Apotex would have been able to obtain a supply agreement from
Estevé.
[168] It is very apparent that Mylan would never have allowed Apotex entry
to the United States market with Estevé’s product. Mylan had invested heavily
in the further development and approval of Estevé’s omeprazole formulation for
sales into the United States market. According to Ms. Lacasa, Mylan had
captured a significant portion of that generic market with that formulation.
In those circumstances, it is inconceivable that Mylan would have waived its
exclusive rights in favour of a generic competitor.
[169] Notwithstanding Mylan’s North America rights, it did allow Estevé to
sell the formulation to a third party in Mexico. Mylan had no commercial
presence in Mexico and one can presume it was commercially indifferent to that
market. I do not agree, though, that this experience can be extended to the
Canadian market in the hypothetical world. In the real world Mylan did
enter the Canadian market with the Estevé formulation in 2009 and Estevé
assisted with that process beginning in 2007. Presumably, Mylan began to
contemplate an entry to the Canadian market even earlier than 2007. The
likelihood that Mylan would have waived its Canadian rights in favour of a
significant competitor seems very remote. As Ms. Lacasa noted, businesses
are not in the business of doing significant favours for their competitors.
Rather, they exploit their commercial advantages.
[170] In order to establish the conditions under which Mylan may have
considered a waiver of its exclusive distribution rights, Apotex should have
called a Mylan witness. In the absence of that evidence, I am not persuaded
that Apotex could and would have obtained a NIA from Estevé.
[171] Given the evidence of Dr. Sherman, I also doubt that Apotex would
have come to terms with either Mylan or Estevé under some form of licensing
agreement, even if the opportunity presented itself.
[172] Apotex also led evidence about the hypothetical availability of a
non-infringing omeprazole formulation to be supplied by Kudco. Kudco is the
United States subsidiary of the German pharmaceutical company Schwartz Pharma and
it held the United States rights to a non-infringing omeprazole formulation
under license from a French company, Pharma Pass. Kudco sought regulatory approval
in the United States for its capsule product in mid-1998 by way of an ANDS. It
received a conditional approval in 2001 subject to the resolution of
outstanding infringement proceedings with AstraZeneca. Final regulatory
approval was granted in 2002 when the Kudco product was found to be non-infringing.
Until August 2003, Kudco was the only generic entrant in the United States
market.
[173] Kudco’s Chief Financial Officer at the relevant time was Jon Thiel.
Mr. Thiel was called on behalf of Apotex.
[174] Mr. Thiel testified that in the period of 2001 to 2003 Kudco
did not have a business presence in Canada and did not supply its omeprazole
product to the Canadian market. Nevertheless, he was asked if Kudco would have,
at the time, been interested in entering into a business relationship with
Apotex for sales into Canada. In response, he gave the following testimony [Transcript
p 1615]:
A. We were a company that tried to be
very opportunistic in terms of creating additional revenue sources. Canada
was, as I stated, not a market that we were in. With the proper capacity and
obviously the proper financial terms, it would have been attractive to us.
It was an easy way
to get into Canada. Assuming that Apotex would have done the regulatory stuff
within Canada, it would have been an easy way for us to get in the market and
gain additional profits.
Q. Would Kremers have had to have
sought permission from Schwarz or anyone to enter into such an arrangement?
A. When you say "Schwarz"?
Q. Schwarz Pharma.
A. U.S., Germany? There are quite a
few of us.
Q. Let's say the German operation.
A. For something like this, no.
Q. Was that a decision that would
have been made locally within your group at Kremers KUDCo?
A. Yes, the small team that ran the
brand and generic in the U.S. would have made that decision.
Q. You were a part of that team?
A. Yes, I was.
Q. Would the omeprazole product have
been any different from the product that was sold in the United States?
A. No, it would not have been.
Q. Would Kremers or KUDCo have had
the capacity to supply Apotex with omeprazole capsules?
A. Over time, yes. As soon as we
launched, we started increasing capacity daily. We made significant
investments in our Seymour facility, both in terms of machinery and equipment,
people, increased shifts.
So as soon as we
had approval from the FDA to launch, I think immediately we knew that if we
increased capacity, it would pay off -- I don't remember anymore. It was
days. We substantially increased capacity every day.
Q. You mentioned that you took some
steps. What year did you take those steps in?
A. As soon as we got approval, so
that would -- I would have to -- I don't recall at the time of launch, anymore,
what our capacity was. But as soon as we got into November of 2002 when we
knew we were alone in the market, that is when we started increasing capacity
over and above what we already had.
[175] AstraZeneca objected to this line of questioning on the basis that
it called for an inadmissible lay opinion. I allowed the evidence
provisionally.
[176] I take AstraZeneca’s point that there are clear dangers with asking
a witness directly about what he or, more to the point, his employer would have
done in the context of a hypothetical environment. Questions about the extant
factual conditions that would have been relevant to the decision are
appropriate. Answering the bare question of “what
would you have done” carries very little, if any, weight. In this case
the answers provided by Mr. Thiel are factually relevant and admissible.
[177] Mr. Thiel was asked about the terms that Kudco would have
required to do business with Apotex. This line of enquiry is appropriate
insofar as it has a factual foundation rooted in the business terms that
typically applied to similar arrangements. Mr. Thiel was also the appropriate
witness to speak to this issue because he was the manager who was, at the time,
involved in these types of negotiations. His answers are instructive of the
terms that Kudco would likely have sought and accepted, at least with respect
to sales into the Canadian market [Transcript p 1617]:
Q. Mr. Thiel, let's assume for a
moment that Apotex and Kremers or KUDCo decided to enter into a supply
agreement. How would Kremers have priced its capsules for purchase by Apotex?
A. What we typically did is we
charged our customers our cost of goods.
Q. Would there be any additional
remuneration that would be required in addition to your cost of goods?
A. Yes. We would have required some
type of a profit share, gross profit share, usually anywhere from 25 to upwards
-- we go as high as we could, quite honestly. We would try to get 50 per cent
if we could.
Q. All right, so somewhere in that
range. Where do you think you ultimately would have ended up?
A. It is hard to say, obviously,
because we are kind of in the hypothetical world. Obviously, we would have
asked for 50 and they would have said, "How about five or 10" or
whatever. And 30 or 40 per cent, I would guess.
Q. Can you tell me, is that similar
to the range that other agreements were entered into with other companies?
A. I don't specifically recall
"yes." Again, having done this now, obviously with Schwarz as well
as County Line, my experience as a pharmaceutical finance person, yes, that is
kind of the range -- is 25 to 50 per cent these days.
Q. Assuming that financial terms were
agreed upon, do you believe it likely that such an agreement would have been
entered into with Apotex?
A. Yes.
Q. We have been talking about
Canadian sales. Would Kremers have been willing to do a deal for sale of an
Apotex product into the United States after competition you had already gone
and received generic competition in the U.S. market?
A. Would we have considered it? Was
that your question?
Q. Yes.
A. Yes, I believe we would have.
Again, as I said, we were very opportunistic. We were a relatively small
pharmaceutical company for the U.S. standards and we were very aggressive as --
I guess "aggressive" is the right word -- as it came to creating
profit opportunities.
Certainly if the
conditions were right in terms of if there was so much competition, we
understood that at some point, to make money, more volume helps. When you own
a manufacturing facility, you bring your costs down by running more volume
through it.
It is free money,
at some point. So yes, we absolutely would have considered it, again, with the
right conditions in terms of competition and everything else to run -- to sell
to a competitor, yes.
Q. That includes a competitor even in
the same jurisdiction where were you [sic] operating?
A. Yes.
[178] Under cross-examination Mr. Thiel confirmed that between 2001
and 2008, Apotex made no enquiries about obtaining the Kudco formulation.
During the same period, Kudco neither sold its formulation to any other generic
company nor did it approach another company with a view to doing business.
[179] Mr. Thiel was unable to recall if Kudco had the legal right to
sell its omeprazole formulation to Apotex under the terms of its assignment
agreement with Pharma Pass and that agreement was not put into evidence.
[180] Mr. Thiel also acknowledged that he was being paid $500 (USD)
per hour for his preparation and trial testimony.
[181] Further evidence concerning the hypothetical availability of the
Kudco omeprazole formulation was provided by Mr. Tom Lewis.
Mr. Lewis started working as Kudco’s Plant Controller in 1999. In that
role he was responsible for the financial activities of the business and
strategic and capacity analysis. He reported to Mr. Thiel.
[182] Mr. Lewis testified that under the terms of the licensing
agreement with Pharma Pass and, with the exception of Europe, there were no
restrictions on Kudco’s right to sell its omeprazole formulation to a
third-party.
[183] According to Mr. Lewis, the licensing agreement could not be
produced because of business confidentiality concerns. Nevertheless, he
professed to be intimately aware of its terms.
[184] Mr. Lewis described Kudco’s manufacturing capacities in considerable
detail in terms of producing its omeprazole formulation, including the efforts
it made to increase capacity in response to demand. He summed up that evidence
in the following way [Transcript p 1670]:
Q. Let me ask you that question
again. If, in the period 2001 to 2003, Apotex approached Kremers and asked
whether it would have been interested in entering into a supply agreement for
the sale of Kremers' omeprazole product in Canada, would this have been
something Kremers would have been capable of doing?
A. Yes.
Q. Why do you say that?
A. At that same time, there were
multiple other third parties that were approaching us to do additional
production for them in our facility, and we accepted the majority of those and
started manufacturing for those companies.
Q. I want to ask you specifically if
Apotex had needed one million 20 milligram capsules in 2003, 26 million
capsules in 2004, 63 million capsules in 2005, 115 million capsules in 2006,
150 million capsules in 2007, and 133 million capsules in 2008, are any of
those numbers I have given you on a yearly basis beyond the capability or
within the constraints of Kremers?
A. We would have easily been able to
do that.
Q. Would a supply deal with Apotex
have been something Kremers would have been willing to consider?
A. We would have.
Q. Why do you say that?
A. As I mentioned earlier, we had
multiple third-party companies that were approaching us to do business for
them, to manufacture for them, and we were doing that because we had excess
capacity to lend to them.
[185] Under cross-examination Mr. Lewis was asked what Kudco typically
expected as a royalty under its third-party supply contracts. Like
Mr. Thiel he said the negotiated rate could be as high as 50% but “probably 20 to 50 percent, maybe 30 to 50 percent” [Transcript
p 1692]. He also confirmed that Kudco did not historically supply its
products to other generic competitors for the United States market [Transcript
p 1696].
[186] Dr. Sherman also gave evidence about what Apotex would have
done to obtain a NIA formulation from either Estevé/Mylan or Kudco and on what
terms. When asked about his willingness to pay a royalty rate as high as 50%,
he used the word “exorbitant” [Transcript p 1959].
In direct examination he went on to describe a likely hypothetical negotiation
in the following terms [Transcript p 1960]:
Q. Let's say they insisted on cost
plus a profit share, what would your reaction be?
A. I would say fine if it is going to
be cost plus profit instead of markup, it should be your net cost plus some
modest percentage of the profit.
Q. What if they had insisted on 40 or
50 percent?
A. If they had insisted and I had no
option, I would have to take it I suppose, but that is unlikely because they
wouldn't know that I would have no option. It is first of all not likely to
happen, but secondly, they wouldn't know I had no other option. It is most
unlikely.
If it were, in
fact, the case I had no option and they knew I had no option, then I would
accept it. It is not likely that would end up that way. It is more likely
they would say, "Fine. Let's do it at cost plus 20 percent or a 10
percent royalty or some compromise." That would be the most likely
outcome.
[187] Under cross-examination, Dr. Sherman conceded that no attempt
was ever made to obtain a non-infringing third-party omeprazole formulation or
to reflect on the terms of any such arrangement [Transcript p 2192]:
Q. During the life of the 693 Patent,
Apotex never asked Mylan, KUDCo, or Lek to supply omeprazole product to Apotex?
A. No. Of course not. There was no
reason to do so. If there had been reason to do so, I would have.
Q. During the life of the 693 Patent,
Apotex never knew what it would cost for anyone to supply it with an omeprazole
product using the KUDCo, Lek, or Mylan processes?
A. I would have to discuss it with
them and negotiate to find out.
Q. The answer would be you didn't
know?
A. Of course not. I could have guessed.
Q. Am I correct that Apotex never
sought or obtained a license from anyone to make, use, or sell any omeprazole
formulations using the KUDCo, Mylan, or Lek process during the life of the 693
Patent?
A. Sorry. You are contemplating I
would make it using their process when I didn't know the details?
Q. You never sought or obtained a
license from anyone to make, use, or sell any omeprazole formulations using the
KUDCo, Lek, or Mylan processes?
A. Of course not. I wouldn't need a
license. If I wanted to make a product without an ARC, I wouldn't need a
license. I could do it far better and design a formulation in a matter of
minutes that is better than that used by any of the three of them according to
what is in their patent.
[188] Dr. Sherman’s explanation for this was given in the following
way [Transcript p 2115]:
Q. Let me explore that. You were
sued in the United States?
A. Yes.
Q. In 2000, thereabouts, when the
action started in the U.S.?
A. Yes, 2000-2001. I am not sure.
Q. You never took any steps to
mitigate or to investigate the existence of a subcoating in your products?
A. Now you are getting back to the
same issue, the one about semantics. We are going to have this with every
question unless I can give you my understanding now.
Q. You never took any steps to
develop a non-infringing alternative after you were sued in the United States;
correct, until 2001?
A. We had a non-infringing product,
and I couldn't predict the basis upon which a court would find otherwise.
Q. You never took any steps after you
lost, in the United States litigation in 2007, to switch to a non-infringing
alternative. Correct?
A. Again, there is a premise in there
that the product was infringing. It was found to be infringing in the United
States. That doesn't mean that it was in fact infringing, nor it would be held
infringing in Canada.
[189] The above evidence establishes that there was some potential
for Apotex to license the Kudco NIA on some basis at some point.
Mr. Thiel and Mr. Lewis testified that Kudco was opportunistic and
would likely have sought a royalty of 50% of the gross profits earned from
Apotex’s Canadian sales and might have settled for something in the range of
30% to 40%. Kudco also had no direct means to access the Canadian market and
it had excess manufacturing capacity.
[190] A major problem for Apotex, though, is that no deal would have been
available to it until Kudco had established that its omeprazole formulation was
non-infringing. That finding was made by the District Court in late 2002 and
affirmed on appeal a year later. It was only in this time-frame that Apotex
could have realistically sought regulatory approval from Health Canada for the
Kudco product. It is also noteworthy that the Kudco product had been approved
in the United States with Prilosec (AstraZeneca’s omeprazole product for sales
into the United States) and not LOSEC (the Canadian product) as the reference
product. This is significant because when Apotex sought approval for Apo-Omeprazole
one of the major hurdles it faced in Canada involved its use of Prilosec as the
reference product. Furthermore, given Dr. Sherman’s evidence that Apotex
would only have considered a third-party source if its in-house approach
failed, I do not think this NIA strategy could have been realistically pursued
until well into the infringing period.
[191] An even larger problem with this theory is the weakness of the
evidence tendered by Apotex concerning the commercial terms that could have
been expected in reaching a licensing agreement with Kudco. I do not accept
that reaching such an agreement would have been as routine as Dr. Sherman
suggested. Dr. Sherman expected to do a deal for a “modest” royalty in a likely range of 10 to 20%. In
the United States District Court proceeding, Apotex took the position that a
license fee of no more than 7% was warranted [see AstraZeneca AB v Apotex
Corp, 985 F Supp 2d 452 (2013)]. Kudco on the other hand typically
negotiated a higher rate of return of 30 to 40%.
[192] This kind of evidence is not very helpful. Evidence in the form of
an economic model would have been more persuasive than this kind of
hypothetical anecdotalism. I am not, therefore, satisfied that Apotex is more
likely than not to have entered into some form of licensing agreement with
Kudco during the infringing period. If I am wrong about the standard of proof
that applies such that the test in Athey v Leonati, [1996] 3 S.C.R. 458,
140 DLR (4th) 235, is applicable, I would fix the possibility of reaching
an agreement with Kudco to supply the Canadian market by the beginning of the
infringing period at 15% and at a royalty rate of 35% on Apotex’s net sales.
[193] I also do not accept the evidence of the Kudco witnesses insofar as
they suggested Kudco would have considered licensing Apotex to sell into the
United States market. I think it is highly unlikely that Kudco, as a leader in
the generic market for omeprazole, would have seriously considered an
arrangement to further divide the United States market – at least on terms
Apotex would have entertained. The evidence from Mr. Thiel and Mr. Lewis
about the contractual basis for allowing entry to Apotex was simply too vague
to support such a finding. Indeed, this is an inherent weakness in promoting a
NIA theory on the strength of assumptions that have little or no historical
precedent.
[194] I would add one more concern about the evidence of Mr. Thiel.
He acknowledged under cross-examination that he was receiving $500 (USD) per
hour from Apotex for time spent in preparation and in giving his trial
testimony. The practice of paying fact witnesses substantial sums to testify
about the likelihood of hypothetical events is suspect, at least when it goes
beyond an indemnity for lost income. The hindsight nature of the enquiry
coupled with the incentive to provide an answer the payor is looking for – or
to colour the answer on favourable terms – makes the practice particularly
questionable. To the extent Mr. Thiel expressed an optimistic view of the
hypothetical prospects of doing business with Apotex, I discount that evidence.
[195] Dr. Sherman’s testimony that it would have been a simple,
straight-forward exercise to develop a commercially viable NIA to
Apo-Omeprazole is belied by history. That was not Apotex’s experience with
Apo-Omeprazole and it was not Estevé’s experience with its non-infringing
formulation. That was also not Apotex’s experience with the numerous NIA
formulations it tried, most of which, by its own acknowledgement, failed. In
the end, Apotex has asserted seven alternative formulations but, as noted
above, not one of them has been shown to be approvable and commercially
viable. For the reasons given above, I also do not accept that Apotex would
have obtained a NIA from a third party.
[196] Apotex has, therefore, failed to prove on a balance of probabilities
that it would have had an available NIA omeprazole formulation at any time
during the infringing period in any of its markets.
III.
How Should the Court Reconcile the Section 8
Judgment in Favour of Apotex in Court Docket T-2300-05 with the Infringement
Judgment in Favour of AstraZeneca in Court Dockets T-1409-04 and T-1890-11
[197] The parties have agreed on the quantum of Apotex’s claim in the
section 8 reference. The only questions left for my determination are whether
Apotex is entitled to recover that amount having regard to a) its corresponding
infringement of the 693 Patent and to b) its NIA defence to recovery by
AstraZeneca for that infringement. As discussed above, I have found that
Apotex did not and would not have had an available NIA during the infringing period.
The only remaining issue concerns the interplay between the section 8 Judgment
favouring Apotex in Court file T-2300-05 and my Judgment finding for
AstraZeneca in Court files T-1409-04 and T-1890-11.
[198] According to Apotex its entitlement to damages is fixed and final
and cannot be redetermined. Apotex relies on a series of judgments rendered in
its favour by this Court and by the Federal Court of Appeal which, based on
principles of res judicata and abuse of process, it contends prevent the
issue of its section 8 entitlement from being relitigated.
[199] AstraZeneca has a different view of the significance of the earlier
litigation history and says it is not estopped from making a case for a nil recovery
by Apotex. According to AstraZeneca’s argument, Apotex has not suffered any
financial loss but only a lost opportunity to unlawfully infringe the 693
Patent.
[200] As a starting point to this part of the analysis it is important to consider
some of the underlying principles to a claim to section 8 damages. Section 8
of the Patented Medicines (Notice of Compliance) Regulations, SOR/93-133
[NOC Regulations] provides:
8 (1) If an
application made under subsection 6(1) is withdrawn or discontinued by the
first person or is dismissed by the court hearing the application or if an
order preventing the Minister from issuing a notice of compliance, made
pursuant to that subsection, is reversed on appeal, the first person is
liable to the second person for any loss suffered during the period
|
8 (1) Si la
demande présentée aux termes du paragraphe 6(1) est retirée ou fait l’objet
d’un désistement par la première personne ou est rejetée par le tribunal qui
en est saisi, ou si l’ordonnance interdisant au ministre de délivrer un avis
de conformité, rendue aux termes de ce paragraphe, est annulée lors d’un
appel, la première personne est responsable envers la seconde personne de
toute perte subie au cours de la période :
|
(a) beginning on the date, as certified by the Minister, on which
a notice of compliance would have been issued in the absence of these
Regulations, unless the court concludes that
|
a) débutant à la date, attestée par le ministre, à laquelle un
avis de conformité aurait été délivré en l’absence du présent règlement, sauf
si le tribunal conclut :
|
(i) the certified date was, by the operation of An Act to amend
the Patent Act and the Food and Drugs Act (The Jean Chrétien Pledge to
Africa), chapter 23 of the Statutes of Canada, 2004, earlier than it would
otherwise have been and therefore a date later than the certified date is
more appropriate, or
|
(i) soit que la date attestée est devancée en raison de
l’application de la Loi modifiant la Loi sur les brevets et la Loi sur les
aliments et drogues (engagement de Jean Chrétien envers l’Afrique), chapitre
23 des Lois du Canada (2004), et qu’en conséquence une date postérieure à
celle-ci est plus appropriée,
|
(ii) a date other than the certified date is more appropriate; and
|
(ii) soit qu’une date autre que la date attestée est plus
appropriée;
|
(b) ending on the date of the withdrawal, the discontinuance, the
dismissal or the reversal.
|
b) se terminant à la date du retrait, du désistement ou du rejet
de la demande ou de l’annulation de l’ordonnance.
|
(2) A second
person may, by action against a first person, apply to the court for an order
requiring the first person to compensate the second person for the loss
referred to in subsection (1).
|
(2) La seconde
personne peut, par voie d’action contre la première personne, demander au tribunal
de rendre une ordonnance enjoignant à cette dernière de lui verser une
indemnité pour la perte visée au paragraphe (1).
|
(3) The court may
make an order under this section without regard to whether the first person
has commenced an action for the infringement of a patent that is the subject
matter of the application.
|
(3) Le tribunal
peut rendre une ordonnance aux termes du présent article sans tenir compte du
fait que la première personne a institué ou non une action en contrefaçon du
brevet visé par la demande.
|
(4) If a court
orders a first person to compensate a second person under subsection (1), the
court may, in respect of any loss referred to in that subsection, make any
order for relief by way of damages that the circumstances require.
|
(4) Lorsque le
tribunal enjoint à la première personne de verser à la seconde personne une
indemnité pour la perte visée au paragraphe (1), il peut rendre l’ordonnance
qu’il juge indiquée pour accorder réparation par recouvrement de
dommages-intérêts à l’égard de cette perte.
|
(5) In assessing
the amount of compensation the court shall take into account all matters that
it considers relevant to the assessment of the amount, including any conduct
of the first or second person which contributed to delay the disposition of
the application under subsection 6(1).
|
(5) Pour
déterminer le montant de l’indemnité à accorder, le tribunal tient compte des
facteurs qu’il juge pertinents à cette fin, y compris, le cas échéant, la
conduite de la première personne ou de la seconde personne qui a contribué à
retarder le règlement de la demande visée au paragraphe 6(1).
|
(6) The Minister
is not liable for damages under this section.
|
(6) Le ministre
ne peut être tenu pour responsable des dommages-intérêts au titre du présent
article.
|
[201] The above provision was considered in somewhat similar circumstances
in Apotex Inc v Merck & Co, Inc, 2011 FCA 364, 430 NR 74 [Lovastatin
2011]. In that case Merck pleaded ex turpi causa actio non oritur
in the context of Apotex’s claim under section 8. Merck’s argument was that
Apotex could not claim a loss that arose in connection with an act of
infringement. In allowing the appeal Justice John Evans had this to say:
[36] I do not
accept Merck’s submission that the Court should read into this provision
limiting words to the effect, “unless the second person’s claim is based on the
loss that is has suffered by being prevented from infringing the first person’s
patent earlier.” The presumption against reading words into a statutory text
may be rebutted when demanded by context and legislative objective. In my
view, it is not necessary to read an ex turpi causa exception into
subsection 8(1) in order to prevent patent infringers from unjustly recovering
compensation from a first person.
[37] This is
because subsection 8(5) confers a broad discretion on the court when assessing
the amount of compensation that the second person must pay. It provides that
the court “shall take into account all matters that it considers relevant to
the assessment of the amount,” including any conduct by either party that
contributed to the delay in the disposition of the first person’s application
for prohibition. In my view, this provision enables the Court to determine in
its discretion whether, and to what extent, a second person’s claim for
compensation should be reduced, or eliminated.
[38] The
Court’s broad discretion under subsection 8(5) allows it, when considering
arguments based on ex turpi causa, to have regard to the factual
situation in its entirety, including its nuances. In the present case, one
such nuance is that not all the tablets sold by Apotex were found in the
infringement action to contain lovastatin made by the infringing process. A
court is likely to find it easier to apply the ex turpi causa principle
through an exercise of judicial discretion than through the definition of
liability. Discretion enables the court to assess the appropriate amount of
compensation payable (including nil) in a manner that properly takes account of
all the relevant facts.
[Emphasis added.]
[202] The discretion conferred upon the Court by subsection 8(5) must accordingly
be exercised with due regard to all of the circumstances bearing on the claim.
[203] Apotex’s claim to section 8 damages arose from a NOC proceeding
heard initially by Justice John A O'Keefe in connection with AstraZeneca’s
762 Patent. That patent covered a combination medication for omeprazole and an
anti-bacterial agent. Justice O’Keefe dismissed AstraZeneca’s application
on the basis that it had failed to establish that the Apotex product would
infringe: see AstraZeneca AB v Apotex Inc, 2004 FC 313, 33
CPR (4th) 97. It was from Justice O’Keefe’s Judgment that Apotex then brought
its action seeking section 8 losses in the proceeding now before me in Court file
T-2300-05. That proceeding was first heard by Justice Roger Hughes
in the early part of 2012 and his Judgment can be found at Apotex Inc v
AstraZeneca Canada Inc, 2012 FC 559, 410 FTR 168.
[204] When Justice Hughes heard the matter he was aware of AstraZeneca’s
outstanding action against Apotex in Court file T-1409-04, alleging an infringement
of the 693 Patent. Indeed, one of the issues presented jointly to Justice
Hughes was the following:
5. Whether the alleged
infringement of the ‘693 Patent is relevant in law, including whether it is
relevant as a defence, to the section 8 claim of Apotex (including possible
set-off of damages) (and if so, see para. 4 of Order of October 4, 2011)?
[205] In dealing with this issue Justice Hughes cited the Federal Court of
Appeal decision in Lovastatin 2011, quoted above, and the United Kingdom
High Court of Justice, Chancery Division, Patents Court decision in Les
Laboratoires Servier v Apotex Inc, [2011] EWHC 730 [Servier trial]. Justice
Hughes neatly summed up the trial holding in Servier in the following
way:
[143] The question of ex turpi causa
came before the United Kingdom High Court of Justice, Chancery Division,
Patents Court in Les Laboratoires Servier v Apotex Inc, [2011] EWHC 730
(Pat), a decision given by Justice Arnold. In that case, Apotex had been
prevented from selling perindopril in the United Kingdom by an interlocutory
injunction given pending trial. Servier, who obtained the injunction, had
given an undertaking as to damages. Apotex prevailed at trial and sought
damages pursuant to the undertaking. Servier argued that Apotex could not have
made and sold the product, in any event, since the product would have been made
in Canada. The Federal Court of Canada (Snider J.,) had held that Apotex’s
product would infringe a valid Servier Canadian patent, hence it would be
unlawful for Apotex to make and export the product from Canada (Laboratoires
Servier v Apotex Inc, 2008 FC 825). Justice Arnold made an extensive
review of the law of ex turpi causa and concluded that the unlawfulness
as proven must be sufficiently serious before a person engages the ex turpi
causa rule, and that such unlawfulness must be an activity personal to the
claimant, not vicarious.
[206] Although noting that the Servier trial decision was overturned
on appeal, Justice Hughes observed that Apotex made a concession in that case
that “played a critical role in the reversal”. By
that concession Apotex agreed that its recovery of damages in the English
proceeding must be reduced by any damages awarded to Servier in its Canadian
infringement action (as an additional cost of manufacture). This allowance was
said to serve the interests of comity and to ensure that Apotex was not
overcompensated. Justice Hughes said that the Servier solution “accords with what may properly be done in the present
situation”. He then went on to say, at para 148:
… A Court hearing the pending infringement
action, if it concludes that the patent is valid and has been infringed by
Apotex in making the omeprazole drug that is the subject of these proceedings, can
at that time craft a remedy that is appropriate, having in mind any
compensation awarded in these proceedings. It would be unconscionable for
the present proceedings to come to a halt or for this Court to refuse to award
compensation simply because another action on another patent was pending. To
do so would be simply to encourage such actions to be brought. The best way to
deal with the matter is as I have set out above.
[Emphasis added.]
[207] Justice Hughes summarized his finding on this issue in the following
way:
FINDING: In the circumstances of this
case, the ex turpi causa rule is not engaged; the future possibility
of a finding of infringement is insufficient to engage that rule. The
infringement action is not material to a determination under subsection 8(1) in
this case.
[Emphasis added.]
[208] His Judgment was then stated as follows:
1. Apotex is entitled to be compensated for loss suffered by
it by reason of the proceedings taken by AstraZeneca in T-2311-01 for the
period from January 3, 2002 until December 30, 2003 under the provisions of
subsection 8(1) of the NOC Regulations;
2. There
is no basis for an exercise of judicial discretion under subsection 8(5) of the
NOC Regulations to reduce or refuse an award of such compensation;
…
[209] AstraZeneca appealed Justice Hughes’ Judgment raising, inter alia,
the following issue:
1. Is it relevant to the section 8 claim that Astrazeneca has
sued Apotex for infringement of the patent in issue and the infringement trial
has not yet been completed?
[210] The Court of Appeal upheld Justice Hughes’ Judgment on this point for
the following reasons [see Astrazeneca Canada Inc v Apotex Inc, 2013 FCA
77, 444 NR 254]:
[4] The first question arose when
Astrazeneca asked Justice Hughes to delay the determination of section 8
damages in this case because its claim against Apotex for damages for
infringement has not yet been determined. We note the jurisprudence to the
effect that in assessing section 8 damages, the judge has the discretion under
subsection 8(5) to reduce the damages based on an argument of ex turpi causa
which could include an infringement claim (Apotex Inc. v. Merck & Co.
Inc., 2011 FCA 364, at paragraphs 36 to 38). In this case, however, there
has been no judicial determination that Apotex has infringed the patent, or
would have done so but for the mandatory statutory stay during the prohibition
proceedings.
[5] Justice Hughes had the discretion
to refuse the request of Astrazeneca to delay the proceedings, and he did so. We
have not been persuaded that the record discloses any basis upon which this
Court should intervene.
[6] The fundamental reason for
Justice Hughes’ decision on this point is stated as follows at paragraph 148 of
his reasons:
A Court hearing the pending infringement
action, if it concludes that the patent is valid and has been infringed by
Apotex in making the omeprazole drug that is the subject of these proceedings,
can at that time craft a remedy that is appropriate, having in mind any
compensation awarded in these proceedings.
[7] We agree with this statement. It
will be for the judge trying the infringement action to ensure that overall,
taking both proceedings together, a party is compensated for its provable loss,
if any, on proper principles, no more and no less.
[Emphasis added.]
[211] The above disposition did not end the matter. When this Court did
make a finding of infringement against Apotex in Court files T-1409-04 and
T-1890-11, AstraZeneca brought the matter back to Justice Hughes in Court file
T-2300-05 on a motion to reconsider. What AstraZeneca was seeking was a
qualification to Justice Hughes’ initial Judgment directing the reference judge
to have regard to the infringement finding in Court files T-1409-04 and T-1890-11
“to reduce or refuse an award of […] compensation”.
[212] Justice Hughes dismissed AstraZeneca’s motion both because my Judgment
was still on appeal and because the requested relief amounted to an
inappropriate request under Rule 399 of Federal Courts Rules,
SOR/98-106, to reverse the effect of his original Judgment.
[213] Once again AstraZeneca appealed and once again the appeal was
dismissed [see Astrazeneca Canada Inc v Apotex Inc, 2016 FCA 194, [2016]
FCJ No 759 (QL)]. In considering the effect of my intervening infringement
finding on Apotex’s claim under section 8 of the NOC Regulations,
the Court of Appeal said this:
[9] One of the errors asserted on
appeal by AstraZeneca was that Justice Hughes had erred in finding the pending
infringement action to be irrelevant to the claim for damages under section 8
of the Regulations. This Court rejected AstraZeneca’s argument, expressly
affirming the correctness of the passage quoted above at paragraph 6 and
characterizing the passage to have been the fundamental reason for Justice
Hughes’ decision. The Court went on to state that “[i]t will be for the judge
trying the infringement action to ensure that overall, taking both proceedings
together, a party is compensated for its provable loss, if any, on proper
principles, no more and no less”.
[10] It is in this context that
AstraZeneca moved for variation of the May 11, 2012 judgment. The basis for
its motion was a finding by Justice Barnes of the Federal Court in an action
for patent infringement that Apotex had infringed certain claims of the ‘693
Patent (2015 FC 322 and 2015 FC 671). This finding was said by AstraZeneca to
be a new matter that arose after the judgment of Justice Hughes in the section
8 proceeding.
[11] Two variations to the judgment
were sought by AstraZeneca on the motion. The first variation sought to add a
provision that when determining Apotex’ entitlement to damages, the reference
Judge may have regard to the judgment of Justice Barnes. The second variation
would have reversed Justice Hughes’ conclusion that there was no basis for an
exercise of discretion under subsection 8(5) of the Regulations to reduce or
refuse compensation to Apotex. Instead, the judgment would provide that the
reference Judge might have regard to the judgment of Justice Barnes when
exercising discretion under subsection 8(5) of the Regulations.
[12] As noted above, Justice Hughes
dismissed the motion for variation. He gave a number of reasons for his
decision, only two of which need be dealt with on this appeal.
[13] Justice Hughes’ principal reason
for dismissing the motion was that in his original decision he had expressly
considered the scenario where Apotex might later be found to have infringed
another patent. Further, this Court had agreed with his conclusion that it
would be for the Judge in the infringement action to ensure a party is neither
over nor under compensated for its loss. Thus, Justice Hughes wrote the “only
thing that has now happened is that the ‘might happen’ scenario considered by
me and the Court of Appeal has become a reality. That makes no difference. The
‘reality’ has already been considered and a determination made. Nothing
changes.”
[14] I agree with Justice Hughes for
the reason that he gave. Justice Barnes’ finding of infringement of the ‘693
Patent is not a matter that arose or was discovered after Justice Hughes’
judgment in the section 8 proceeding within the contemplation of Rule 399(2)(a).
This finding is dispositive of the appeal.
[…]
[24] A final note. This appeal and
motion highlight the difficulties that ensue when inconsistent findings are
made in parallel infringement and section 8 proceedings. I can only repeat
Justice Sharlow’s admonition on the prior appeal to the effect that it will be
for the Judge hearing “the infringement action to ensure that overall, taking
both proceedings together, a party is compensated for its provable loss, if
any, on proper principles, no more and no less.”
[214] What I take from the above-noted statements and particularly those
of the Federal Court of Appeal is that, as the section 8 reference Judge, I
have the discretion to take into account the intervening infringement finding,
among other relevant facts, and to craft an appropriate remedy.
[215] This is also consistent with the broad parameters of the discretion
discussed in Lovastatin 2011, above.
[216] I do not accept that AstraZeneca’s right to raise this issue against
Apotex ought to be precluded because of the order in which these matters
initially came before this Court. If Apotex would have been required to
account for its infringement of the 693 Patent had that finding been made ahead
of Justice Hughes’ section 8 Judgment, there is no principled basis for
foreclosing that result because the order of the outcomes was reversed.
[217] What I am left with is a situation where, in order to recover its “losses” from being barred from selling Apo-Omeprazole
between January 3, 2002 to December 30, 2003 in the face of AstraZeneca’s 762
Patent, Apotex necessarily had to infringe AstraZeneca’s 693 Patent. Apotex’s
claim to section 8 losses is accordingly offset by the costs of the
corresponding infringement or, as AstraZeneca put it – if Apotex had entered
the market during the section 8 liability period, it would have been required
to disgorge its profits to AstraZeneca.
[218] As the Federal Court of Appeal observed in Lovastatin 2011,
above, it is unnecessary to apply the theory of illegality to resolve this
issue. Indeed, the strict application of that principle could, in some cases,
leave a party undercompensated. This was essentially the point being made by
Lord Toulson of the United Kingdom Supreme Court in the ultimate appeal of the Servier
trial decision (Les Laboratoires Servier v Apotex Inc, [2014]
UKSC 55 [Servier UKSC]) in the following passages:
52. The
order made by the Court of Appeal accords with Lord Diplock’s method of
assessment. As Etherton LJ explained in his judgment at para 88, its effect is
to place Apotex in precisely the position in which it would have been if there
had been no UK interlocutory injunction, and it does not offend comity with
Canada. Apotex will recover whatever sum may be left after deducting, from the
proceeds of the lost sales, both the costs of the sales and the amount for
which it would have had to account to Servier in the Canadian proceedings by
way of damages for patent infringement. The result, Etherton LJ said, would
neither be offensive to comity with Canada nor infringe English public policy.
53. By
contrast, the order sought by Servier would potentially place it in a better
position than if it had not obtained the English injunction for which it gave a
cross-undertaking. I use the word potentially, because it remains to be seen
how the Canadian court will calculate damages for the infringement which led to
UK sales by Apotex. It will be a simple matter to apply the same approach to
the lost sales as the Canadian court will apply in relation to actual sales
made by Apotex. The result may be that Apotex will be unable to establish any
loss, after deduction of the damages which it would have had to pay in Canada,
but that will depend on the outcome of the Canadian proceedings.
[…]
63. Cross-undertakings
are a standard and valuable feature of litigation, particularly but not only in
commercial litigation. There is a public interest in their enforceability in
bona fide disputes. It saves the court from having to make a more detailed –
and therefore time consuming and expensive – assessment of the merits at an
interlocutory stage than might otherwise be necessary, since the
cross-undertaking is designed to protect the defendant against the applicant
gaining a financial advantage from obtaining an injunction which is later set
aside on the claim failing. I cannot see a good public policy reason why
Servier should be put in a better position than if the English injunction had
not been granted, or why Apotex should be required to give greater credit to
Servier on account of its breach of the Canadian patent than the amount
assessed by the Canadian court as properly reflecting that breach.
[219] It follows that Apotex is not entitled to recover under section 8 of
the NOC Regulations because it suffered no loss by being kept out of the
marketplace between January 3, 2002 and December 30, 2003.
[220] The parties do not disagree that a profits-on-profits allowance
should be applied to AstraZeneca’s profits entitlement. They differ only as to
the amount and method of calculating the rate. AstraZeneca seeks prime plus
two percent compounded annually. Apotex argues for simple interest at the bank
rate.
[221] The burden on this issue rests with Apotex: see Reading &
Bates Construction Co v Baker Energy Resources Corp (1994), 58 CPR
(3d) 359 at p 375, 175 NR 225 (FCA) [Reading & Bates FCA].
[222] An award of interest on profits is not a matter of complete
discretion. Such a recovery represents an accounting for the additional profit
the infringer made from the use of the wrongfully acquired funds: see Teledyne
Industries, Inc v Lido Industrial Products Ltd (1982), 68 CPR (2d) 204 at p 226,
[1982] FCJ No 1024 (QL) (FCTD).
[223] Where it is not possible to know precisely how the infringer put its
profits to use, it will be assumed to have made “the
most beneficial use of them”: see Reading & Bates FCA at p 376
and Adir v Apotex Inc, 2015 FC 721 at para 146, 482 FTR 276 [Perindropril
FC]. In that situation the Court will estimate the return based on
relevant investment or borrowing proxies. Compounded interest is the presumptive
approach: see Reading & Bates FCA at p 374.
[224] As with the situation in Perindropril FC, above, it is not
possible on the evidence before me to know exactly how Apotex made use of the
profits generated from the sales of Apo-Omeprazole. Apotex co-mingled the sales
proceeds from all of its products and used those funds in the day-to-day
operation of its business. Money also moved back and forth on a daily basis
between Apotex and Apotex Pharmachem Holdings Inc.
[225] Neither Ms. Paula Frederick nor Mr. Howard Rosen provided much help
in establishing a rate of return and each adopted a rate that favoured the
party retaining them. There was some evidence produced about Apotex’s cost of
borrowing from third-party lenders, |||||||||||||||||||||||||| ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||.
Paying down those loans appears not to have been a priority for Apotex.
[226] It is apparent from Apotex’s balance sheets that a lot of its
capital expenditures and research & development costs were paid for from
cash generated from operations. The overall impression is that Apotex
typically used its profits to successfully build the business over the relevant
period of infringement.
[227] To the extent Apotex relies on financial interactions with related
companies as proof of its return, I reject that approach. I do not accept that
non-arms-length transactions between related businesses are useful in the
assessment of an appropriate rate of return. That is so because the terms of
such arrangements can be easily manipulated to favour one party over the other.
For example, one party could easily underutilize its profits by transferring
them to a related party at low or zero interest. Alternatively, one party
could borrow money from the other at an exorbitant rate and artificially
inflate its costs.
[228] The decision of my colleague Justice Jocelyne Gagné in Perindropril
FC, above, provides useful guidance about how to best apply the authorities
to a very similar set of facts. As with the case at hand, Justice Gagné could
not trace the profits earned by Apotex from its sales of perindopril. She
observed that most of the relevant authorities have used prime plus one or two
percent as proxies for a return on profits. She also observed that Apotex
operates in a highly profitable environment. In the result, she awarded the
compounded prime rate against Apotex. She applied slightly higher rates to the
award against Pharmachem, consistent with its higher costs of borrowing (prime
plus one-half and prime plus one, both compounded).
[229] In my view the benchmark rate for profits-on-profits in cases like
this one has consistently been set at the prime rate or slightly higher,
compounded annually. On the other hand, there is very little recent authority
utilizing a rate as high as prime plus two percent. In this case, I fix the
rate at prime compounded annually.
[230] Ms. Frederick acknowledged in her report that a deduction for income
tax would be warranted on her profits-on-profits assessment. Nevertheless, she
made no adjustment for tax because Apotex did not disclose its income tax
returns for the relevant period.
[231] Under cross-examination it was suggested to Ms. Frederick that she
could have applied a stipulated corporate tax rate to obtain an appropriate
adjustment. She disagreed, saying that Apotex operates in a complex tax
environment where generous research and development tax credits are available
and where assumptions are unwarranted [see Transcript pp 306-8].
[232] On this issue, I agree with Ms. Frederick. Apotex could have
avoided any uncertainty by producing its tax returns and it declined to do so.
This is not the first time Apotex has faced this problem. In Wellcome Foundation
Ltd v Apotex Inc, [2001] 2 FC 618, 11 CPR (4th) 218 (CA), the
Court observed at para 30 that Apotex “did not tender
its income tax returns as evidence of tax paid”. Apotex’s financial
statements were described as “merely projections”
and inadequate substitutes. The Court went on to observe that, despite being
aware of its potential liability, Apotex “had not
maintained financial records in a way that made it clear what expenses could be
attributed for its various product lines” (para 31). The same point
applies to some of Apotex’s records produced in this case.
[233] In the result, I allow nothing as a tax adjustment on the award of
profits on profits.
[234] A remaining point of disagreement between the parties concerns the
appropriate treatment of the District Court award to AstraZeneca for Apotex’s infringement
of the United States 505 Patent. The parties have resolved the accounting
issues in relation to this issue but disagree as to the legal effect of the
United States Judgment on AstraZeneca’s recovery in these proceedings. The
parties have presented four options for the Court’s consideration, ranging from
a disgorgement of all Apotex’s United States export profits less the amount
already paid in satisfaction of the United States Judgment to a nil recovery
(beyond the amount already paid in the United States proceeding). The four
recovery scenarios are the following:
•
Scenario A – Apotex disgorges all U.S. export
profits, less its portion of the U.S. Judgment.
•
Scenario B – Apotex disgorges US export profits
made after expiry of the U.S. patent, April 20, 2007.
•
Scenario C – Apotex disgorges U.S. export
profits made after expiry of the pediatric extension, October 20, 2007.
•
Scenario D – Apotex does not disgorge any U.S.
export profits.
[235] AstraZeneca seeks recovery under Scenario A while Apotex maintains
that Scenario D is the legally correct approach. Scenarios B and C are put
forward by Apotex only in the alternative to D. The parties have agreed to the
amounts to be paid (if any) depending on the Court’s determination of legal
entitlement.
[236] Apotex’s fundamental objection to paying further compensation for
its United States sales is based on principles of cause of action estoppel
(including the doctrine of election), issue estoppel and abuse of process.
AstraZeneca contends that it is not legally precluded from obtaining a further
recovery in these proceedings provided full credit is given for Apotex’s
payment in satisfaction of the United States Judgment (thereby eliminating any
form of double recovery).
[237] The legal authorities cited by the parties do not clearly resolve
the question of how this Court ought to treat the District Court award beyond
the recognition that full credit is due for Apotex’s payment in satisfaction of
the United States Judgment. On that point, the parties are agreed. Apotex
says that, having elected to pursue compensation in the United States,
AstraZeneca cannot claim any shortfall in these proceedings. AstraZeneca
asserts that there are no legal barriers to a full recovery for Apotex’s
infringement of the 693 Patent.
[238] Perhaps the closest Canadian authority touching on Apotex’s res
judicata argument is Apotex Inc v Sanofi–Aventis, 2011 FC 1486, 101 CPR
(4th) 1. In that case Apotex asserted a settlement agreement of a
United States action as a bar to recovery in an infringement action between the
parties in Canada. Justice Richard Boivin rejected Apotex’s contention
that the settlement agreement was intended to apply to Canada. He also dealt
with Apotex’s res judicata and abuse of process arguments in the
following way:
[284] Finally, the Court further recalls
that Apotex also raised the defences of estoppel and abuse of process in
reference to the Settlement Agreements.
[285] With respect to estoppel, Apotex
submits that, under this principle, Sanofi is precluded from pursuing in this
action what is, according to Apotex, a second claim for compensation in respect
of the very same manufacture and sale of the U.S. APO-clopidogrel.
[286] Moreover, Apotex argues that the
monetary judgment in the U.S. Clopidogrel Action was secured on the basis of a
contractual arrangement between the parties pursuant to which they stipulated
as to what is essentially a factual matter (the measure of Sanofi’s “actual
damages” in the event of a launch at risk by Apotex in the U.S. and subsequent
finding that the ‘265 Patent was valid and infringed). It follows, says Apotex,
that Sanofi would be claiming damages in Canada on the same pills that were
sold in the U.S. and were the subject of a damages award by Justice Stein of
the United States District Court – Southern District of New York.
[287] Apotex also submits that if Sanofi
is able to obtain an accounting of profits, they will be able to recoup the 50%
that they negotiated away in the March and May 2006 Agreements. Because Apotex
claims to have proceeded to act in reliance of that stipulation, Apotex argues
that Sanofi should be estopped from attempting to circumvent that stipulation.
[288] An estoppel defence operates to
preclude a party from relitigating the same cause of action twice (Danyluk v
Ainsworth Technologies Inc., 2001 SCC 44, [2001] 2 S.C.R. 460, at paras 18 and
54). In Toronto (City) v Canadian Union of Public Employees (C.U.P.E.),
Local 79, 2003 SCC 63, [2003] 3 S.C.R. 77, at para 23, the Supreme Court of
Canada held that three (3) preconditions must be met for estoppel to be
successfully invoked:
[23] …(1) the issue must be the
same as the one decided in the prior decision; (2) the prior judicial decision
must have been final; and (3) the parties to both proceedings must be the same,
or their privies (Danyluk v Ainsworth Tech., 2001 SCC 44, [2001] 2 SCR
460, 2001 SCC 44, at para 25, per Binnie J.) The final requirement,
known as “mutuality”, has been largely abandoned in the United States and has
been the subject of much academic and judicial debate there as well as in the
United Kingdom and, to some extent, in this country. (See G.D. Watson,
“Duplicative Litigation: Issue Estoppel, Abuse of Process and the Death of
Mutuality” (1990), 69 Can. Bar. Rev. 623 at pp. 648-51.) …
[289] Res judicata is essentially
premised on the notion that a matter has already been adjudged and is founded
on the principles that a party shall not be vexed twice for the same complaint
and that there is a societal value in the finality and conclusiveness of
judicial decisions (see Angle v Canada (Minister of National Revenue),
[1975] 2 S.C.R. 248, at para 267; CPU Options, Inc. v Milton (2006), 79 OR
(3d) 365, at para 15 (SCJ)).
[290] Against this background, the
Court is not able to accede to Apotex’ alleged estoppel defence because the
U.S. litigation and the Agreements simply did not deal with infringement or the
validity of the ‘777 Patent. It is therefore not open for the Court to conclude
that the issue is the same as the one decided in the Agreements. The Court
accordingly agrees with Sanofi that, where the legal rights upon which a cause
of action is based were not adjudicated in the previous proceeding, the
estoppel principle does not apply.
[291] Apotex has also raised the issue of
abuse of process. In common law, judges have an inherent and residual
discretion to prevent an abuse of the Court’s process (CUPE, above, at
para 35). However, the Court has not been convinced that this case boils down
to a question of abuse of process. On the basis of the evidence and for the
reasons mentioned above, the Court remains unpersuaded that Sanofi is using the
courts for an improper use and that the integrity of the court’s process is at
issue in this case.
[Emphasis added.]
[239] On appeal the Court declined to resolve the issue of whether the
losses from the infringement of the United States and Canadian patents were the
same and therefore barred from recovery by the rule against double recovery [see
Sanofi-Aventis v Apotex Inc, 2013 FCA 186, 114 CPR (4th) 1]:
[115] I would agree that the equitable
rule against double recovery would prevent Sanofi from recovering the same loss
twice. To the extent that the sale of clopidogrel in the U.S. in breach of the
‘265 patent is the same loss as that incurred by Sanofi from Apotex’s
exportation of clopidogrel to the U.S. for sale there, Sanofi could only
recover that loss once. I point out, however, that it has not been established
to this point that the infringement of the ‘265 and ‘777 Patent by the
exportation of clopidogrel to the U.S. are, in fact or in law, the same loss.
Since the matter must be returned to the Trial Judge on the question of
remedies, I will say no more about that question.
[240] Apotex’s res judicata argument is not based on an assertion
of any misconduct. Rather, it contends that a public policy principle is at
stake such that AstraZeneca ought to be estopped from pursuing the same
essential recovery in two places. In simple terms, Apotex says that by opting
to claim a recovery in the United States AstraZeneca must now accept that award
as full satisfaction of its entitlement from the infringement of the 693 Patent
for Apotex’s sales into the United States.
[241] Apotex relies for its argument on the holding of the British
Columbia Supreme Court in JRT Nurseries Inc v 0843374 BC Ltd, 2016 BCSC
501, [2016] BCJ No 578 (QL) [JRT]. That case involved proceedings
brought in Oregon and in British Columbia for the recovery of damages arising
from a common set of facts. The case was first tried in Oregon where product
liability damages were recovered. The plaintiff was not satisfied with the
Oregon results, including the inability in that jurisdiction to recover
pre-judgment interest, and it sought to retry the case in British Columbia claiming
under the Sale of Goods Act, RSBC 1996, c 410. The British Columbia
litigation was halted on the basis of cause of action estoppel. The Court
determined that the litigation could not be salvaged by recasting the liability
theory or by reference to juridical differences to recovery between Oregon and
British Columbia. The Court concluded as follows:
[51] … In this case, the plaintiffs
claimed their entire loss in the Oregon proceedings. Consequently, this is not
like Cuttell v Bentz (1985), 65 BCLR 273 (CA) where the loss claimed in
the first proceeding was not the full loss suffered by the plaintiffs (see
p 289). Here, the claim was for all of the damages the plaintiff suffered
from the conduct of the defendants, including Terralink as agent of Sun-Gro.
That claim was adjudicated, and the judgment was satisfied. There was no
shortfall within the proper meaning of that term, whether arising from a
rethinking of the claim, the denial of prejudgment interest, or compromise.
[242] Apotex argues that the same approach is required in this case. It
says AstraZeneca is seeking to recover essentially the same amounts that were
the subject of the United States litigation and for which full United States
compensation has been awarded and paid. Like JRT, above, it should not
be entitled to recast the claim in these subsequent proceedings.
[243] I am not convinced, however, that JRT, above, applies to the
situation before me. That case involved a common set of facts and issues that
were fully triable in either Oregon or British Columbia. The Court easily saw
through the plaintiff’s attempt to recast the cause of action to recover a
perceived shortfall in the Oregon proceeding.
[244] In these proceedings the circumstances are different. Here, the causes
of action in the two proceedings arose under different patents, involved
distinct acts of infringement and were tried in jurisdictions where different
substantive legal principles applied. It is particularly noteworthy that the
approach to recovery in the United States involved a hypothetical license and
the fixing of a reasonable royalty. In that country, a claim to the
infringer’s profits has been unavailable since 1946: see Allied Signal Inc
v Du Pont Canada Inc (1995), 61 CPR (3d) 417 at p 445 fn 11, 184 NR 113
(FCA). The amount awarded to AstraZeneca by the District Court under its
approach also fell well short of AstraZeneca’s entitlement in Canada to
Apotex’s profits from its United States sales.
[245] I would add that there is not a perfect temporal correspondence
between the acts of infringement in issue in the United States litigation and
those in play in these proceedings. AstraZeneca’s United States claim arose
from the infringement of the 505 Patent – a claim that necessarily ended with
the expiry of that patent on April 20, 2007. After that date Apotex was no
longer infringing the United States patent and no viable claim to further
royalties could be determined by the United States courts. But Apotex
continued to infringe the 693 Patent by making Apo-Omeprazole in Canada and by
exporting that product into the United States at a considerable profit. That
conduct constituted a separate cause of action for which compensation is
payable in these proceedings. It cannot be the case that, by proceeding first
in the United States, AstraZeneca should be taken to have abandoned its claim
for ongoing Canadian infringement post-dating the expiry of the United States
patent.
[246] It seems to me that Apotex’s argument in this case is much the same
as Servier’s argument in Servier, above. The issue presented by that
case was whether Apotex should be barred from recovering damages in the United
Kingdom because of its corresponding infringement in Canada of a Canadian
patent. Damages were claimed by Apotex in the United Kingdom proceeding under
an undertaking by Servier in support of the grant of an interlocutory
injunction and where the European patent was later held invalid. As above, Servier
argued that Apotex could not recover damages based on the principle of illegality
– that is to say, that Apotex’s lawful sales in the United Kingdom were the
result of its infringement of a Canadian patent. The effect of a finding of illegality
would be to preclude any recovery by Apotex under Servier’s undertaking,
regardless of the amount otherwise due in the United Kingdom proceeding.
[247] By the time the case was heard, Apotex had conceded that its claim
to damages in the United Kingdom should be reduced by the amount it was
ultimately required to pay in satisfaction of its Canadian liability. If that
amount was less than its United Kingdom damages, Apotex sought to make up the
difference.
[248] On appeal, both the English Court of Appeal and the United Kingdom
Supreme Court declined to apply the doctrine of illegality based, at least in
part, on a concern that the result might leave Servier with a windfall at the
expense of Apotex.
[249] This point is made in the following passages from the respective
Judgments of Lord Sumption and Lord Toulson in Servier UKSC:
30.
In my opinion, the illegality defence is not engaged by the consideration
that Apotex’s lost profits would have been made by selling product manufactured
in Canada in breach of Servier’s Canadian patent. A patent is of course a
public grant of the state. But it does not follow that the public interest is
engaged by a breach of the patentee’s rights. The effect of the grant is simply
to give rise to private rights of a character no different in principle from
contractual rights or rights founded on breaches of statutory duty or other
torts. The only relevant interest affected is that of the patentee, and that is
sufficiently vindicated by the availability of damages for the infringements in
Canada, which will be deducted from any recovery under Servier’s undertaking in
England. There is no public policy which could justify in addition the
forfeiture of Apotex’s rights.
[per Lord Sumption]
…
53.
By contrast, the order sought by Servier would potentially place it in a
better position than if it had not obtained the English injunction for which it
gave a cross-undertaking. I use the word potentially, because it remains to be
seen how the Canadian court will calculate damages for the infringement which
led to UK sales by Apotex. It will be a simple matter to apply the same
approach to the lost sales as the Canadian court will apply in relation to
actual sales made by Apotex. The result may be that Apotex will be unable to
establish any loss, after deduction of the damages which it would have had to
pay in Canada, but that will depend on the outcome of the Canadian proceedings.
…
63.
Cross-undertakings are a standard and valuable feature of litigation,
particularly but not only in commercial litigation. There is a public interest
in their enforceability in bona fide disputes. It saves the court from having
to make a more detailed – and therefore time consuming and expensive –
assessment of the merits at an interlocutory stage than might otherwise be
necessary, since the cross-undertaking is designed to protect the defendant
against the applicant gaining a financial advantage from obtaining an
injunction which is later set aside on the claim failing. I cannot see a good
public policy reason why Servier should be put in a better position than if the
English injunction had not been granted, or why Apotex should be required to give
greater credit to Servier on account of its breach of the Canadian patent than
the amount assessed by the Canadian court as properly reflecting that breach.
[per Lord Toulson]
[250] One other relevant consideration applied by the English Court of
Appeal, Civil Division, concerned the territorial reach of a patent [see Les
Laboratoires Servier v Apotex Inc, [2012] EWCA Civ 593]. At paragraph 83
the Court noted that a patent has limited territorial reach for liability
purposes; but that does not mean that a court cannot examine matters arising
beyond the border insofar as they concerned compensation:
[83] Thirdly, it was common ground
before us that sales made in the United Kingdom from goods manufactured in
breach of the Canadian Patent were not and (but for the injunction) would not
have been unlawful under either Canadian or United Kingdom law. That is because
patents are territorial. In the words of Kitchin LJ in the course of
submissions, the tort comes to an end at the border: comp. the Patents Act
1977s 60. That is reflected in the final injunction granted by the Canadian court
in the Canadian proceedings, which prohibited Apotex from manufacturing,
selling, offering for sale or otherwise dealing in prindopril [sic] products in
Canada [emphasis added]. Whether or not damages flowing from infringement,
or an account of profits for infringement, would involve any investigation of
matters that occurred outside the jurisdiction is a quite separate issue. As
the Judge noted (in 103 of his judgment) Servier does not contend that Apotex’s
United Kingdom business in importing and selling perindopril erbumine was
unlawful.
[251] The examination of extra-territorial matters including foreign
judgments is accordingly required to ensure that double recovery for the same
loss does not occur but, absent illegality, it goes no further than that.
[252] It seems to me that if concurrent actions are permissible in more
than one jurisdiction and, indeed, are necessary to ensure complete recovery
across jurisdictions, the concerns about forum shopping, finality and
multiplicity of proceedings largely disappear. Indeed, concurrent infringement
actions in different jurisdictions claiming under similar patents for losses
that may overlap are not an uncommon occurrence. Because the laws and the
available legal options for recovery for patent infringement will vary from
place to place (including the chance of different outcomes), the strict
application of res judicata in all its forms and abuse of process by
re-litigation are to be generally avoided. What will always remain of concern
are the avoidance of either excess or inadequate recovery and the need to
respect foreign judgments to achieve an equitable result.
[253] In this case AstraZeneca will be deprived of a full recovery if
Scenario D is applied. Giving that effect to AstraZeneca’s claim would be to
afford extra-territorial reach to the 505 Patent. I am, therefore,
satisfied that Scenario A is the correct approach to the treatment of the
District Court award.
[254] For the foregoing reason, the Court makes the following declarations
in response to the issues presented by the parties for determination:
(a)
At no time during the infringing period did
Apotex have an available NIA;
(b)
Apotex is not entitled to any section 8
damages recovery;
(c)
AstraZeneca is entitled to recover an amount for
profits-on-profits at the prime rate compounded annually and without a
deduction for tax effects; and
(d)
AstraZeneca is entitled to fully recover its
claim to Apotex’s profits for United States sales under Scenario A.
[255] The issue of costs and any other matters requiring the Court’s
consideration in accordance with the streamlining agreement will be reserved
pending further submissions from the parties.