throbber

`•• •
`.. . •
`
`Review
`
`M O L E C U L A R O N C O L O G Y 6 ( 2 0 1 2 ) 2 5 1 e2 5 9
`
`a v a i l a b l e a t w w w . s c i e n c e d i r e c t . c o m
`SciVerse ScienceDirect
`
`w w w . e l s e v i e r . c o m / l o c a t e / m o l o n c
`
`~lec~lar
`=~ ncoogy
`
`\
`
`-
`
`-
`
`Germline pharmacogenomics in oncology: Decoding the patient for
`targeting therapy
`
`Peter H. O’Donnella,b,c,d,*, Mark J. Rataina,b,c,d
`
`aSection of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue, MC 2115,
`Chicago, IL 60637, USA
`bCommittee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, 5841 S. Maryland Avenue, MC 2115,
`Chicago, IL 60637, USA
`cComprehensive Cancer Center, The University of Chicago, 5841 S. Maryland Avenue, MC 2115, Chicago, IL 60637, USA
`dCenter for Personalized Therapeutics, The University of Chicago, 5841 S. Maryland Avenue, MC 2115, Chicago, IL 60637, USA
`
`A R T I C L E
`
`I N F O
`
`A B S T R A C T
`
`Article history:
`Received 20 September 2011
`Received in revised form
`14 December 2011
`Accepted 13 January 2012
`Available online 21 January 2012
`
`Keywords:
`Pharmacogenomics
`Targeted therapy
`Oncology
`
`Pharmacogenomics is the study of genetic factors determining drug response or toxicity.
`The use of pharmacogenomics is especially desirable in oncology because the therapeutic
`index of oncology drugs is often narrow, the need for favorable drug response is often
`acute, and the consequences of drug toxicity can be life-threatening. In this review, we ex-
`amine the state of pharmacogenomics in oncology, focusing only on germline pharmaco-
`genomic variants. We consider several critical points when assessing the quality of
`pharmacogenomic findings and their relevance to clinical use, and discuss potential con-
`founding factors limiting interpretation and implementation. Several of the most exten-
`sively studied drugegene pairs (irinotecan and UGT1A1; tamoxifen and CYP2D6; 5-
`fluorouracil and DPYD) are inspected in depth as illustrations of both the state of advance-
`mentdand the current limitations ofdpresent knowledge. We argue that there will likely
`soon be a critical mass of important germline pharmacogenomic biomarkers in oncology
`which deserve clinical implementation to provide optimal, personalized oncologic care.
`We conclude with a vision of how routine clinical testing of such germline markers could
`one day change the paradigm for cancer care.
`ª 2012 Federation of European Biochemical Societies.
`Published by Elsevier B.V. All rights reserved.
`
`1.
`
`Introduction
`
`Pharmacogenomics is the study of genetic factors determin-
`ing response to, or toxicity from, drugs. While the field origi-
`nally centered on the relationship between drugs and single
`
`genes (pharmacogenetics), pharmacogenomics now encom-
`passes information from the entire genome including germ-
`line variation (single nucleotide polymorphisms [SNPs], gene
`copy number alterations) and acquired changes (tumor muta-
`tions) as they relate to drug response or toxicity (Wang et al.,
`
`* Corresponding author. Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue,
`MC 2115, Chicago, IL 60637, USA. Tel.: þ1 773 702 7564.
`E-mail address: podonnel@medicine.bsd.uchicago.edu (P.H. O’Donnell).
`1574-7891/$ e see front matter ª 2012 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.
`doi:10.1016/j.molonc.2012.01.005
`
`NOVARTIS EXHIBIT 2053
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 9
`
`

`

`252
`
`M O L E C U L A R O N C O L O G Y 6 ( 2 0 1 2 ) 2 5 1 e2 5 9
`
`2011; Watson and McLeod, 2011). In contrast to disease genet-
`ics, pharmacogenomics focuses specifically on predictive ge-
`netic markers of outcome from pharmacologic interventions.
`The use of pharmacogenomic markers is perhaps espe-
`cially desirable in the field of oncology, where the therapeutic
`index of drugs is often narrow, and the consequences of drug
`toxicity can be life-threatening. However, since adverse drug
`reactions are reported to be the fifth leading cause of death
`in the United States, the risks are not specific to oncology
`drugs (Davies et al., 2007). At the same time, it is likely that
`we have failed to capitalize on the increased benefit that could
`be achieved with some therapies if we knew which patients
`were most likely to respond, or which patients required alter-
`native dosing. If we could better predict which individuals are
`at the greatest risk of suffering chemotherapy-related toxic-
`ities while simultaneously identifying those most likely to
`benefit, then the overall care of cancer patients could be
`greatly improved.
`In this review, we will examine the state of pharmacoge-
`nomics in the field of oncology. We will specifically restrict
`our considerations to germline genetic discoveries related to
`oncologic therapeutics; a discussion of the growing number
`of “molecularly-targeted” drugs based upon tumor pharmaco-
`genomics is beyond the scope of this current manuscript. To
`date, most germline oncology pharmacogenomic information
`has simply been cataloged, or, in a few instances, has led to
`FDA drug label changes. Therefore, we will also consider the
`barriers and means by which oncology pharmacogenomic
`informationdwhich is increasing every daydcan become
`more commonly integrated into the routine care of cancer pa-
`tients. We will posit that use of such patient-specific informa-
`tion should soon become the standard of care, rather than the
`exception.
`
`Major current pharmacogenomic findings in
`2.
`oncology
`
`The number of germline oncology drugegene pharmacoge-
`nomic pairs having high levels of evidentiary support is rela-
`tively small compared to other drugs. Perhaps the strongest
`examples are those for which the strength and scope of the
`data has resulted in FDA-mandated label changes so that pre-
`scribing clinicians are aware of well-characterized, pertinent
`germline pharmacogenomic information when prescribing
`(United States Food and Drug Administration, 2011). These
`highest level drug-variant pairs, along with several other of
`the most extensively studied oncology drug-variant pairs,
`are summarized in Table 1.
`As can be seen from Table 1, most of the existing described
`relationships have focused on genetic predictors of oncology
`drug-related toxicity phenotypes, rather than disease out-
`come phenotypes, although accumulating data suggest that
`germline polymorphisms might also affect treatment out-
`comes (see references in Table 1; and selected others (Huang
`et al., 2011; Wu et al., 2010; Ziliak et al., 2011; Yang et al.,
`2009)). Of the drugegene pairs in Table 1, the pharmacoge-
`nomic relationships between irinotecan and UGT1A1 (for neu-
`tropenia risk)
`(Innocenti and Ratain,
`2006), and 6-
`TPMT
`mercaptopurine/thioguanine
`and
`(for
`severe
`
`myelosuppression) (Relling et al., 2011) have the most consis-
`tent, strong supporting evidence in favor of their routine use.
`For UGT1A1 as an example, several prospective studies have
`demonstrated that patients with the high-risk genotypes
`(UGT1A1*28 and UGT1A1*6) are significantly more likely to ex-
`perience neutropenia, with two of these studies corroborating
`the relationship with pharmacokinetic supportive data
`(Innocenti et al., 2004; Minami et al., 2007). In the largest
`such study of 250 metastatic colorectal cancer patients, the
`odds ratio of risk of cycle 1 grade 3 or 4 neutropenia was w9,
`although the relationship did not persist for subsequent cycles
`(Toffoli et al., 2006). A meta-analysis of published studies on
`UGT1A1-irinotecan (including 821 patients) also confirmed
`the association for patients homozygous for the UGT1A1*28 al-
`lele who are receiving higher doses of irinotecan (150 mg/m2)
`(Hoskins et al., 2007). Including other risk alleles within UGT1A
`in a haplotype-based analysis may increase the predictive
`value of pharmacogenomic testing, since several other vari-
`ants in these genes have now also been shown to alter enzy-
`matic activity and impact
`irinotecan-related outcomes
`(Cecchin et al., 2009).
`There has also been significant interest in the relationship
`between tamoxifen and CYP2D6 (discussed further below).
`While the preponderance of the published data support the
`utility of CYP2D6 testing for tamoxifen use, there has not
`been a recommended pharmacogenomic FDA label change
`for this drug, and recent data presented in abstract form
`have been contradictory (Goetz et al., 2009; Rae et al., 2010;
`Leyland-Jones et al., 2010). There is also a large body of grow-
`ing evidence for many more oncology drug polymorphisms
`and various phenotypes. The best-performed studies of
`emerging pharmacogenomic associations now routinely in-
`clude replication testing upfront, and these drug-variant pairs
`deserve further examination for how they might be consid-
`ered in clinical utility investigations.
`Despite the existence of well-performed studies and vali-
`dation in many cases, some have still questioned whether
`any present germline oncology findings are currently clini-
`cally actionable without further prospective follow-up trials
`being performed (Coate et al., 2010). Certainly even the best-
`studied drugegene relationships have recognized limitations
`in applicability which must be considered (Lee and McLeod,
`2011). We believe that the clinical utility of each finding
`must be interpreted not only in light of the composite evi-
`dence describing a given relationship but also in the context
`of the clinical scenario in which the relative benefit versus
`risk must be considered. If a pharmacogenomic test could po-
`tentially mitigate risk without compromising efficacy, then we
`believe its practical value is high. We will discuss this topic
`further below. It is important to first consider interpretation
`of published pharmacogenomic findings as a starting point.
`
`Limitations to pharmacogenomic data
`3.
`interpretability
`
`As evidenced by the examples shown above, the most con-
`vincing drug-variant
`relationships are those identified
`through well-performed studies in which prospective pheno-
`type collection is performed and in which the potential for
`
`NOVARTIS EXHIBIT 2053
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 9
`
`

`

`M O L E C U L A R O N C O L O G Y 6 ( 2 0 1 2 ) 2 5 1 e2 5 9
`
`253
`
`Table 1 e Summary of the most extensively studied germline pharmacogenomic relationships for oncology drugs.
`
`Drug
`
`Phenotype(s)
`evaluated
`
`Genes
`
`Variants
`
`FDA label includes
`pharmacogenomic
`prescribing
`considerations?
`
`Important considerations
`
`Key references
`
`Irinotecan
`
`Neutropenia
`
`UGT1A1
`
`*28; plus others
`likely important
`
`6-mercaptopurine
`/thioguanine
`
`Myelosuppression
`
`TPMT
`
`Tamoxifen
`
`Disease recurrence
`
`CYP2D6
`
`*1, *2, *3A, *3B, *3C,
`*4, plus others
`
`Loss-of-function
`alleles:
`*3 (rs35742686);
`*4 (rs3892097);
`*5 (gene deletion);
`*6 (rs5030655);
`*7 (rs5030867)
`
`Decreased
`function alleles:
`*10 (rs1065852); *41
`(rs28371725); *9
`(rs5030656)
`Plus potentially others
`
`5-fluorouracil
`/capecitabine
`
`Neutropenia, stomatitis,
`diarrhea
`
`DPYD
`
`DPYD*2A (IVS14
`þ 1 G > A),
`plus others
`
`YES, but genetic
`variants are not
`mentioned; only
`functional DPD
`deficiency is included
`as a consideration
`
`Rituximab/cetuximab
`/trastuzumab
`
`Disease progression,
`response
`
`FcgRIIa,
`FcgRIIIa
`
`FcgRIIa-131H/R;
`FcgRIIIa-158 V/F
`
`NO
`
`YES
`
`YES
`
`NO
`
`1) Variants may only be predictive for
`patients receiving higher
`drug doses;
`2) Unclear if other irinotecan
`toxicities (like diarrhea) are similarly
`governed;
`3) Optimal strategy for treating
`*28/*28 patients is not defined
`1) Complementary clinical laboratory
`tests are available to
`functionally assess TPMT activity
`1) Some studies have been unable
`to reproduce the relationships;
`
`Innocenti et al., 2004;
`Minami et al., 2007; Toffoli
`et al., 2006; Hoskins et al., 2007;
`Cecchin et al., 2009; Innocenti
`and Ratain, 2006
`
`Relling et al., 2011
`
`Schroth et al., 2007, 2009;
`Nowell et al., 2005; Kiyotani
`et al., 2010; Jin et al., 2005;
`Ferraldeschi and Newman,
`2010; Rae, 2011
`
`2) Many studies have not included
`all of the known, main alleles;
`
`3) Genotyping (consideration
`of gene duplication) may be
`technically difficult which
`could confound results
`1) Sensitivity of best-studied
`DPYD variant is only w30%
`and has not been consistently
`reproducible;
`2) Results with other DPYD
`variants, or with variants in
`other genes (TYMS, MTHFR), have
`been inconsistent
`1) Some conflicting data; positive
`data mostly from small studies
`
`Yen and McLeod, 2007;
`van Kuilenburg, 2004
`
`Bibeau et al., 2009; Musolino
`et al., 2008; Kim et al., 2006;
`Weng and Levy, 2003; Carlotti
`et al., 2007
`
`NOVARTIS EXHIBIT 2053
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 9
`
`

`

`254
`
`M O L E C U L A R O N C O L O G Y 6 ( 2 0 1 2 ) 2 5 1 e2 5 9
`
`false discovery is minimized either (optimally) by inclusion of
`a replication set, or by conservative adjustment for multiple
`comparisons (Chanock et al., 2007; van den Oord, 2008). Stud-
`ies which are underpowered to adequately test less common
`variantsdvariants which in reality may be potentially impor-
`tant pharmacogenomic markersdcan have (falsely) negative
`results and can confuse the ability to understand conflicting
`data from several studies on a given drugegene pair. Inade-
`quate consideration of the potentially numerous different al-
`leles which may contribute to a given phenotype may also
`cause false negative results. This latter scenario may, in fact,
`be one of the causes of the recent conflicting data surrounding
`tamoxifen pharmacogenomics and CYP2D6 (Higgins and
`Stearns, 2011; Ferraldeschi and Newman, 2010; Rae, 2011).
`For that drugegene pair, multiple studies have demon-
`strated that patients with poor metabolizer genotypes are
`more likely to have worse outcomes. This is due to suboptimal
`conversion (primarily via CYP2D6) of tamoxifen into the more
`potent, active antiestrogenic metabolites, endoxifen and 4-
`hydroxytamoxifen (Higgins and Stearns, 2010), a relationship
`which is supported by pharmacokinetic data showing that pa-
`tients with these genotypes have lower levels of endoxifen
`(Borges et al., 2006). In one study, 206 tamoxifen-treated pa-
`tients receiving the drug in the adjuvant setting were com-
`pared based upon genotype groups (Schroth et al., 2007) for
`disease-related outcomes. Patients with poor metabolizer
`CYP2D6 genotypes were significantly more likely to experience
`recurrence of breast cancer, had shorter times to relapse, and
`worse event-free survival compared with patients having
`functional alleles (Schroth et al., 2007). Importantly, this study
`also examined genotypes for an identical control group of
`women not treated with adjuvant tamoxifen, and genotype
`had no bearing on disease-related outcomes. A 1325-patient
`international consortium study confirmed these findings
`(Schroth et al., 2009). A smaller prior study (also including
`a control group) had failed to demonstrate the association of
`three loss-of-function CYP2D6 genotypes (CYP2D6*3, *4, and
`*6) with reduced tamoxifen-related survival benefit, but im-
`portantly, this study did not test for any of the other now
`known loss-of-function and reduced-function alleles (Nowell
`et al., 2005). The recent data presented only in abstract form
`(Goetz et al., 2009) from the International Tamoxifen Pharma-
`cogenomics Consortium study on >2800 patients receiving ad-
`juvant tamoxifen did not show an association with survival
`outcomes, however, a number of patients was excluded
`from the analysis because of incomplete genotypic or clinical
`data, including lack of information about concomitant medi-
`cation use (Ferraldeschi and Newman, 2010). Two other, re-
`cent large prospective trials (both also only presented in
`abstract form thus far) which examined CYP2D6 genotypes
`with outcomes in patients receiving tamoxifen also failed to
`show associations (Rae et al., 2010; Leyland-Jones et al.,
`2010). The
`apparent
`importance of
`considering
`co-
`administered drugsdincluding simply whether the antineo-
`plastic drug being studied is being given as monotherapy or
`as part of a larger combination regimendhas been elegantly
`illustrated by Kiyotani et al. (2010). These authors showed
`that, for multiple studies (including theirs) where tamoxifen
`was given as part of a combination chemotherapy regimen,
`analyses were unable to demonstrate a positive relationship
`
`between CYP2D6 genotype and disease outcomes. However,
`in their study and in seven of eight other prior published stud-
`ies of patients receiving tamoxifen as monotherapy, the rela-
`tionship between CYP2D6 genotype and tamoxifen response
`was positive (Kiyotani et al., 2010).
`This drugegene example is instructive for three reasons.
`First, for genes where genotyping may be difficult or complex,
`inaccurate or incomplete genotyping can be a significant bar-
`rier to pharmacogenomic interpretation. CYP2D6 is known to
`be frequently duplicated, which can confound interpretation
`of genotyping results if duplication is not well-characterized.
`Moreover, over 100 different alleles of CYP2D6 have been
`reported (Higgins and Stearns, 2010; Bradford, 2002), with at
`least five of these variants well-characterized as loss-of-func-
`tion alleles, and another three well-described as associated
`with decreased enzymatic function (Becquemont et al.,
`2011). None of the above studies comprehensively included
`all of the common functional variants. The lack of standard-
`ized inclusion of all of the various known functional variants
`in clinical studies may therefore be a source of inconsistency
`in reported response outcomes. Secondly, the presence of
`concomitant medications may be important when interrogat-
`ing pharmacogenomic relationships. For tamoxifen, co-
`administered inhibitors of CYP2D6 can functionally “cause”
`the poor metabolizer phenotype (Jin et al., 2005) and confound
`genetic influences. Or, as just mentioned, even the presence of
`drugs not known to be directly acting via the same pathway as
`the antineoplastic of interest (including other concomitant
`antineoplastics) may mask the “penetrance” of pharmacoge-
`nomic risk alleles. The reduced penetrance could be due to di-
`rect effects of the other drugs, plus potentially the reduced
`effect on the drug of interest, especially if there was dose re-
`duction. This issue has now been suggested to be important
`for both the tamoxifen (Kiyotani et al., 2010) and irinotecan ex-
`amples (Hoskins et al., 2007). Third, one of the common prob-
`lems confounding oncology pharmacogenomics is that
`evaluated studies often lack a control group (the relatively
`well-performed study referenced above on tamoxifendwhich
`diddis an exception). Especially when the phenotype of inter-
`est is progression-free survival or overall survival, without
`such a group, or without an intermediate phenotype which re-
`lates the ultimate outcome to drug response, it can be difficult
`to determine whether an associated variant is actually predic-
`tive of treatment effect (truly pharmacogenomic) rather than
`simply prognostic (i.e., a marker for disease severity). This
`consideration can be especially relevant when the gene(s) be-
`ing studied could theoretically be related to not only drug re-
`sponse, but also disease propensity or severity (like, for
`example, genes in DNA repair pathways). All three of these
`are important points to consider when assessing the quality
`of pharmacogenomic findings and their relevance to clinical
`use, and their confounding nature has tempered clinical
`implementation of some results.
`Separately, racial/ethnic differences in genetic variation
`must be considered. The example of dihydropyrimidine dehy-
`drogenase (DPD) deficiency and 5-fluorouracil (5-FU) toxicity
`exemplifies this point. DPD catabolizes >80% of 5-FU into fluo-
`rinated b-alanine (Heggie et al., 1987). A causative link be-
`tween DPD deficiency and severe toxicity in response to 5-
`FU treatment has been repeatedly shown (Milano et al.,
`
`NOVARTIS EXHIBIT 2053
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 9
`
`

`

`M O L E C U L A R O N C O L O G Y 6 ( 2 0 1 2 ) 2 5 1 e2 5 9
`
`255
`
`1999; van Kuilenburg et al., 2000; Johnson et al., 1999). While
`clinical assays of enzymatic DPD activity are available, they
`are not always easy to obtain, and there has been a substantial
`effort to characterize causative genetic variants within the
`DPYD gene relating to the DPD deficient phenotype (Yen and
`McLeod, 2007; Van Kuilenburg et al., 1999). In fact, over 40
`SNPs and deletion mutations have been identified within
`DPYD, but most have been shown to have no functional conse-
`quences on enzymatic activity (Yen and McLeod, 2007). The
`the IVS14 þ 1 G > A variant
`best-studied of
`these SNPs,
`(DPYD*2A), has been found in up to 40e50% of people with par-
`tial or complete DPD deficiency (van Kuilenburg, 2004). Yet
`a recent summary of the data on DPYD*2A, including multiple
`studies of this variant alone or in combination with other
`common variants, showed a performance sensitivity (the per-
`centage of actual patients with severe toxicity who were cor-
`rectly identified by the allele) ranging between 6.3 and 83%,
`with a median sensitivity of 30% (Yen and McLeod, 2007).
`Even more importantly, despite the fact that the prevalence
`of functional enzymatic DPD deficiency is higher in African
`Americans (Mattison et al., 2006), the DPYD*2A variant is not
`even present in African Americans (van Kuilenburg, 2004),
`making such testing of limited generalizability and utility. Di-
`rect to consumer genetic testing services like 23andMe fail to
`convey these nuances: 23andMe Inc (2011) advertises genetic
`testing for 5-FU sensitivity, but their testing consists only of
`genotyping of the DPYD*2A variant, and it is not mentioned di-
`rectly on their website that there is likely to be no relevant in-
`formation about 5-FU susceptibility for certain ethnic groups
`like African Americans. This notwithstanding, the data on
`DPYD testing overall is insufficient to support routine pharma-
`cogenomic testing for 5-FU, in our opinion.
`Finally, even the results of well-performed studies which
`include replication may be of limited utility because of the op-
`posite problem: it might be unclear how to assimilate a larger
`number of different variantsdeach of which might have
`a small (but real) impact on the phenotype of interestdinto
`one coherent pharmacogenomic model, let alone a model
`which might also include clinical factors. The very novel find-
`ing of 102 different variants associated with treatment out-
`come in pediatric acute lymphoblastic leukemiadidentified
`through a very well-conducted analysis of two independent
`cohortsdmight beg that question (Yang et al., 2009). Even if
`a clinician could test for all these variants, how would he or
`she assimilate information about all the variants in composite
`when determining treatment options? These types of ques-
`tions are becoming more relevant as pharmacogenomic dis-
`coveries increase and as the field moves into tackling the
`issues not of discovery, but of clinical implementation.
`
`Clinical implementation of pharmacogenomics
`4.
`into oncology practice
`
`In 2001, when the first draft sequence of the human genome
`was released (Lander et al., 2001; Venter et al., 2001), there
`was significant public expectation that this information would
`be quickly utilized to individualize medical care (Ratain and
`Relling, 2001). However, work during the past decade has,
`within oncology, focused mostly on tumor-specific changes
`
`rather than germline variation as the keys to advancement
`in clinical care. The two disciplinesdtumor versus germline
`variationdare of course very different. The former explains
`variability in disease, which can usually be associated with
`differences in natural history and/or etiology, and occasion-
`ally in treatment response. On the other hand, germline vari-
`ation explains variability in the patient, which does affect
`both pharmacokinetics and pharmacodynamics, as well as
`potentially disease risk (even risk for specific mutations (Liu
`et al., 2011)). Some might argue that especially for the latter
`group of drug-related germline variants, the list of the most
`extensively studied within oncology (summarized in Table 1)
`and especially the list of those that has become routinely clin-
`ically tested remains relatively small.
`Implementation into routine practice has been hindered by
`lack of knowledge about such information (on the part of both
`patients and physicians), uncertainty about how to order such
`tests, and reimbursement, and timeliness of results. We be-
`lieve that we are now at a point where SNP genotyping has be-
`come so widely available and inexpensive that this should no
`longer be the barrier. Indeed, whole genome sequencing is
`itself likely to quickly surmount these same barriers in the
`very near future. And it is also likely that in the very near fu-
`ture, we will have a critical mass of information regarding
`germline pharmacogenomic biomarkers in oncology which
`deserve clinical implementation to provide optimal (personal-
`ized) oncologic care. Before discussing the ways to bring this
`goal to fruition, it is worthwhile to examine the question of
`whether prospective, randomized data need to be demon-
`strated for a drug-variant pair before clinical implementation
`can be considered.
`
`5.
`
`Necessity of prospective validation?
`
`Pharmacogenomic findings even from a well-performed sin-
`gle study require validation in a separate patient population
`to confirm that such results are reproducible (Chanock et al.,
`2007). Successful reproducibility in a separate cohort provides
`considerable confidence that the original findings were not
`false positives and were not misleading due to some unique
`phenotypic or measurement characteristics of the original
`population. Outside of the oncology realm, however, even
`two of the most prominent drugs with repeatedly reproduc-
`ible pharmacogenomic informationdwarfarin and clopidog-
`reldhave not yet seen widespread clinical implementation
`of genomic prescribing. It has been felt that prospective, ran-
`domized studies for each of these drugs (the ongoing Clarifica-
`tion of Optimal Anticoagulation through Genetics [COAG] trial
`for warfarin (French et al., 2010); and the proposed and funded
`Pharmacogenomics of Anti-Platelet
`Intervention [PAPI-2]
`study for clopidogrel (United States Department of Health
`and Human Services, 2011)) are needed to demonstrate the
`clear utility of the pharmacogenomic information. Skeptics
`of pharmacogenomics will argue that this type of prospective
`randomized validation (ideally double-blind) might be neces-
`sary for any pharmacogenomic discovery before it is clinically
`implemented, including those for oncology drugs. In contrast,
`we, like others (Altman, 2011; Frueh, 2009), argue that this will
`not only be practically infeasible given the burgeoning
`
`NOVARTIS EXHIBIT 2053
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 9
`
`

`

`256
`
`M O L E C U L A R O N C O L O G Y 6 ( 2 0 1 2 ) 2 5 1 e2 5 9
`
`number of pharmacogenomic discoveries that continue to be
`reported, but also potentially unnecessary given the diminish-
`ing costs of genotyping, the idea that in many cases a pharma-
`cogenomically-informed prescribing decision will at least be
`non-inferior to the alternatives (Altman, 2011) (and potentially
`highly advantageous to the individual patient being consid-
`ered), and the idea that comparative effectiveness research
`will
`likely continue to validate pharmacogenomically-
`informed prescribing in real practice without needing to
`wait for the randomized controlled trial (Epstein et al., 2010).
`We propose instead that using a broad pre-emptive genotyp-
`ing model, described further below, which eliminates mar-
`ginal
`costs,
`that
`the
`plausible
`benefit
`of
`using
`pharmacogenomics for any given patient should only be
`weighed against the plausible risk.
`
`6.
`
`Pre-emptive genotyping
`
`It is our vision that the implementation of pharmacogenomics
`in the clinic will require a transformation from the current
`paradigm of genotyping as a laboratory test, to a future para-
`digm of genotyping conducted at a single time, and perpetu-
`ally available as part of the “genetic examination”. Whether
`this will involve whole genome sequencing or multiplexed
`genotyping is irrelevant, as the key concept is that the genetic
`examination will be performed in anticipation of future med-
`ical needs, as opposed to the current paradigm where geno-
`typing is performed when clinically indicated (like all other
`current laboratory tests). Thus, we envision that future physi-
`cians will utilize a “genomic prescribing system” at every pre-
`scribing encounter (Ratain, 2007),
`including if and when
`a patient might be diagnosed with cancer. Others have argued
`for a similar approach (Relling et al., 2010). In this model, pa-
`tients could be examined with a single blood sample, used
`for genotyping all polymorphisms of potential pharmacoge-
`nomic significance. If the same panel is used for all patients,
`and if patients are genotyped in large batches, this would
`also reduce the costs of genotyping, creating feasibility for
`the approach.
`This approach would address one of the key barriers to
`pharmacogenomic implementationdthat physicians are re-
`luctant to wait for pharmacogenomic results before prescrib-
`ing. This might be especially true for oncologists and for
`their patients diagnosed with cancer, since there is often
`(much warranted) urgency on the part of oncologists, and pa-
`tients, to begin treatment quickly.
`Successful implementation of this type of model (Ratain,
`2007) will require the training of physicians (and related
`health care providers) with expertise in pharmacogenomics,
`genetic testing, and informatics. It will also require the devel-
`opment of medical records systems which can accommodate
`large-scale, patient-specific genotypic information and deliver
`such information in a usable, succinct format to busy
`clinicians.
`We at the University of Chicago (www.clinicaltrials.gov
`study identifier NCT01280825) and others (e.g. Ohio State-
`Coriell Personalized Medicine Study (Coriell, 2011); Vander-
`bilt’s VESPA project
`(Snyder, 2009; St.
`Jude Children’s
`Research Hospital, 2011)) currently have pharmacogenomic
`
`implementation efforts underway which are beginning to
`studydand realizedthe goal of routine incorporation of phar-
`macogenomics in patient care,
`including oncology care.
`Implementation of the best-evidence variants should be the
`starting point for such studies. Equally important will be si-
`multaneous pharmacogenomic discovery research and ongo-
`ing confirmatory studies of contradictory drugegene pairs, in
`order to properly define the scope of which variants to include
`and to increase the number of drugegene pairs having ade-
`quate clinical evidence to warrant implementation.
`
`Special considerations in implementing oncology
`7.
`pharmacogenomics
`
`Knowledge about pharmacogenomic susceptibility (either re-
`sponse or toxicity prediction) may be most obviously usable
`to inform treatment choices in disease settings where several
`equivalent therapies exist. This would allow the physician to
`potentially choose one therapy over another if specific toxicity
`risks were high for one of the given drugs, or, alternatively, to
`select a therapy if the expected likelihood of response was
`higher. In settings where more than one treatment choice
`may not exist, information about a drug may allow the physi-
`cian to weigh toxicity risks of that treatment versus potential
`benefits, and such questions are of course even more relevant
`when treatments are being used in palliative settings. The
`third, different, scenario where implementation questions
`can arise is that of potential patient-specific dose modifica-
`tions based upon pharmacogenomic knowledge of toxicity
`risk. In other words, if a patient is known to be at high (genetic)
`risk of toxicity from a drug at standard treatment doses, could
`the clinician still use that drug, but at a lower dose? Specific
`dose-reduction pharmacogenomic prescribing recommenda-
`tions are, to our knowledge, not available for any oncology
`drugs except for the above-mentioned TPMT substrates
`(Relling et al., 2011). For irinotecan, a genotype-driven dose-
`finding study using pharmacogenomics showed that the rec-
`ommended 180 mg/m2 dose for irinotecan in the FOLFIRI reg-
`imen is considerably lower than what can be actually
`tolerated if patients with high-risk UGT1A1 genotypes are ex-
`cluded (Toffoli et al., 2010).
`A final potential concern surrounding implementation is
`that, especially when considering toxicity pharmacogenom-
`ics, one must consider the additional question of whether
`the same variants governing toxicity susceptibility might
`also govern antitumor response. If that were known to be
`true, the question arises whether dose reduction (to avoid tox-
`icity) would still be appropriate. In the absence of prospect

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket