throbber
Reprintedfrom Trends in Pharmacological Sciences - October 1990
`
`Vol. II, No. 10
`
`411
`
`
`
`Clinical significance of
`pharmacological modulation of
`homocysteine metabolism
`
`Helga Refsum and Per Magne Ueland
`
`The metabolic fate of homocysteine is linked to vitamin B”, reduced folates,
`vitamin B.-, and sulfur amino acids. Clinical and experimental data suggest that
`elevated plasma homocysteine is an independent risk factor for premature
`vascular disease. This is particularly significant because plasma homocysteine
`levels are altered in several diseases,
`including folate and vitamin B”
`deficiencies, and because many commonly used drugs have now been shown to
`interfere with homocysteine metabolism.
`In summarizing the data, Helga
`Refsum and Per Ueland highlight the clinical implications for these metabolic
`changes.
`
`ubiquitous enzyme methionine
`synthase (Fig. 1). This enzyme re-
`quires vitamin B12 |methy|(l)cobal-
`amin] as a cofactor and 5-metl1yl-
`tetrahydrofolate as methyl donor;
`thus5-methyltetrahydrofolateenters
`the pool of reduced folates, and
`homocysteine is remethylated to
`methionine‘.
`There
`are
`
`cobalamin-
`
`two
`
`dependent enzymes in vertebrates:
`methionine synthase which util-
`izes methy|cob(I)alamin and is
`cytosolic,
`and methylmalonyl-
`CoA mutase which is a mitochon-
`drial enzyme containing aden-
`osylcobalamin. A major fraction of
`intracellular cobalamin is associ-
`ated with these two enzymes?
`Homocysteine remethylation is
`also catalysed by an alternative
`enzyme,
`betaine—homocysteine
`methyltransferase, requiring be-
`taine as methyl donor. However,
`this enzyme is generally confined
`to the liver.
`
`The metabolism of homocys-
`teine along the trans-sulfuration
`pathway is catalysed by two vit-
`amin B6-dependent enzymes. The
`first step is the cystathione [3-
`synthase reaction, where homo-
`cysteine is condensed with serine
`to
`form cystathionine. Cysta-
`thionine is then cleaved to <x-l<eto-
`butyrate and cysteine, catalysed
`by cystathionine lyasel.
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`Homocysteine is a sulfur amino
`acid which is not itself incorpor-
`ated into proteins, but is import-
`ant as an intermediate in the
`metabolism of methionine and
`cysteine, and because its metab-
`olism is linked to the function of
`some vitamins.
`In the late 1960s, inborn errors
`homocysteine metabolism
`of
`(homocystinuria) were demon-
`strated in patients with mental
`retardation,
`skeletal abnormali-
`ties, lens dislocation and prema-
`ture vascular disease‘. Research
`
`into the physiological and patho-
`logical roles of homocysteine was
`subsequently promoted. Since the
`pioneering work of Wilcken and
`coworkers in 1976, accumulated
`epidemiological and experimental
`evidence has shown that homo-
`
`cysteine may provoke vascular
`lesions, and that moderate homo-
`cysteinemia is an independent
`risk factor for premature vascular
`diseasez.
`im-
`During the last few years,
`proved analytical techniques have
`allowed
`the
`investigation
`of
`plasma homocysteine in healthy
`subjects and in disease states
`other than homocystinuria. Folate
`and cobalamin (vitamin B12) de-
`ficiencies cause very high plasma
`
`Helga Re[sum is Research Fellow (NAVF) and
`Per Magm: Ueland is Professor of Clinical
`Pharmacology and Head of
`the Clinical
`Pharmacology Unit at Haukelanrl Llniversity
`Hospital, N-5021 Bergen, Norway.
`
`levels of homocysteine, and plasma
`homocysteine has been estab-
`lished as a sensitive and respon-
`sive
`indicator
`of
`intracellular
`folate and cobalamin function3.
`Some drugs influence homo-
`cysteine metabolism and plasma
`levels. This may have some im-
`portant implications. First, plasma
`homocysteine may reflect pharma-
`codynamic effects of some drugs,
`as most clearly demonstrated with
`methotrexate and nitrous oxide
`(see below). Secondly,
`the in-
`creased plasma levels induced by
`some agents may have impli-
`cations
`for
`their
`side-effects.
`Finally, drugs decreasing plasma
`homocysteine may reduce the risk
`of vascular disease imposed by
`homocysteinemia3.
`
`Homocysteine and vascular
`disease
`Patients with homocystinuria
`suffer
`from premature vascular
`disease,
`localized to the central
`and peripheral arteries and large
`veins. This is the major cause of
`the high mortality (20—75% before
`the age of 30)
`in these patients.
`Thromboembolism may occur at
`any age and has even been de-
`Homocysteine metabolism
`scribed in children‘. Both clinical
`Homocysteine holds a unique
`and
`experimental
`evidence
`position in metabolic regulation.
`Its metabolism is linked to sulfur
`suggest
`that high homocysteine
`levels cause the vascular lesions
`amino acids, reduced folates and
`(see Ref. 1).
`vitamins B12 and B6.
`lts metab-
`Even moderate homocystein-
`olism is summarized in Fig. 1.
`emia may provoke venous throm-
`The only source of homocys-
`teine in vertebrates is the hydroly-
`bosis
`and premature
`vascular
`lesions in the cerebral, peripheral
`sis of S-adenosylhomocysteine, an
`and coronary arteries (L. Brati-
`inhibitor and product of S-aden-
`strom, Thesis, University of Lund,
`osylmethionine-dependent trans-
`1989). Such a relation was suggested
`methylation‘. The fate of
`intra-
`cellular homocysteine is either
`10 years ago from clinical studies
`based on measurement of acid-
`salvage to methionine through
`soluble mixed disulfides in plasma
`remethylation, or conversion to
`cysteine via the trans-sulfuration
`from a small number of patientsz.
`This has later been confirmed in
`pathway.
`In most
`tissues,
`the
`former reaction is catalysed by the
`several
`investigations, most of
`©1990, Elsovior Science Publishers Ltd. (UK)
`0165 - (-1-I7I‘l0l$u2.ti:J
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`

`
`412
`
`Protein
`
`Melhlonlne
`(extracellular)
`
`Serine
`
`/\AdoMet
`
`Methionine
`
`THF
`
`Dlmettlyl-
`glycine
`
`Betaine
`
`Bu @ “IMP
`
`d1‘MP
`
`DHF
`
`_
`
`Glycine
`
`5,10-CH2-THF
`
`AdoHcy
`W_ l-lomocystelne
`
`we
`
`Cystnthionine
`
`5 - CH3-THF 4—— 5-CHO-’I'Hl-‘
`
`./
`'3
`
`Follc acld
`
`..;l
`
`TiP$ — October 1990 (Vol. 717
`
`elevated plasma homocys-
`teine levelsz.
`
`Folate and cobalamin
`deficiency
`Cobalamin deficiency may
`increase plasma homocys-
`teine to levels observed in
`homocystinurics (high micro-
`molar range); there is a nega-
`tive
`correlation
`between
`serum cobalamin and total
`plasma homocysteine. Homo-
`cysteine levels may also be
`elevated
`in
`cobalamin-
`deficienl patients devoid of
`typical signs
`like vanemia,
`macrocytosis
`and
`reduced
`serum cobalamin. Levels are
`normalized following cobal-
`amin therapy3-'3.
`Similarly, folate deficiency
`is a common cause of el-
`evated plasma homocysteine
`levels, and a close negative
`correlation with serum folate
`has
`been
`demonstrated;
`again, folate therapy normal-
`izes levels. Moderate elevation
`
`Be
`
`Homocysteine
`(extracellular)
`Cysteine
`I.
`Fig.
`Homocysteine metabolism. AdoHcy, S~adenosyIhomocysteine; Adomot,
`S-adenosylmethionine; 5.10-CH,-THF. 5,10-methylene THF; 5-CH,-THF. 5-melhyI-
`tetlahydrololate; 5-CHO-THF, 5-Iormyltetrahydrololate (lalinate); CL, cystathionine
`Iyase: OS, cystathionine B-synthase: DHF, dlhydrololate; DH. dihydrololate reductase;
`MET, methionine; MS, methionine synthase (5-methyl-THF homocysteine methyItrans-
`lerase); SA. S-adenosylhomocysteina nydrolase: SH, serine nydroxymeIhyIIransIer-
`ase; THF, tetrahydrololate.
`
`which determined total plasma
`homocysteine3'°'1°. Furthermore,
`an increased incidence of hetero-
`zygous homocystinuria has been
`demonstrated in patients with
`early onset vascular disease7.
`Other clinical and experimental
`data support an association be-
`tween raised homocysteine levels
`and vascular lesions (reviewed in
`Ref.
`3). Plasma homocysteine
`levels show age- and sex-depen-
`dent variations resembling those
`described
`in
`arteriosclerotic
`disease. Men and postmenopausal
`women
`have
`higher
`plasma
`homocysteine levels during last-
`ing and alter methionine loading
`than
`young women.
`Plasma
`homocysteine is significantly in-
`creased in chronic renal failure,
`severe psoriasis, and in some
`patients with cancer. These are
`conditions associated with in-
`creased risk of vascular disease
`not adequately explained by risk
`factors like smoking, lipid abnor-
`malities, hypertension or other
`known predisposing conditions’.
`Moderate homocysteinemia should
`be considered as a possible cause
`of vascular disease in those cases
`(15-30°/o) when other risk factors
`cannot be identified. Down syn-
`drome, on the other hand, is an
`abnormality
`characterized
`by
`low plasma homocysteine levels,
`
`probably due to increased gene
`dosage for the enzyme cystathio-
`nine B-synthase
`residing
`on
`chromosome 21 (Ref. 11). In 1977
`Murdoch and co-workers suggested
`this state as an atheroma-free
`model because of the remarkable
`absence of arteriosclerotic lesions
`observed in five patients aged 44-
`66years12.
`direct
`also more
`is
`There
`that high
`levels
`of
`evidence
`homocysteine
`mediate
`the
`thrombogenesis and accelerated
`atherogenesis observed in homo-
`cystinuria, and that homocysteine
`may also be responsible for the
`vascular disease associated with
`moderate homocysteinemia. Homo-
`cysteine damages human endo-
`thelial cells in culture, possibly by
`producing hydrogen peroxide in
`an oxygen-dependent
`reaction.
`Moreover, endothelial cells from
`patients heterozygous for cysta-
`thionine
`[3-synthase gene de-
`ficiency may have an increased
`susceptibility to injury by homo-
`cysteine. Mechanisms linking mild
`homocysteinemia
`and vascular
`effects could also involve produc-
`tion of free radicals and oxidation
`of low~density lipoprotein. Con-
`flicting data exist on possible roles
`of platelet sequestration in the
`development
`of
`atherosclerotic
`lesions
`under
`conditions
`of
`
`of plasma homocysteine is
`also observed in subjects with low
`but normal serum folate levels,
`suggesting that
`increased homo-
`cysteine levels in these subjects
`may reflect an intracellular folate
`content
`insufficient
`for optimal
`folate-dependent remethylation of
`homocysteine3.
`Measurement of plasma homo-
`cysteine is therefore a promising
`laboratory
`test
`for
`evaluating
`cobalamin or
`folate deficiency
`states. lt may be particularly use-
`ful when used in conjunction with
`serum methylmalonic acid, which
`is a specific measure of disturb-
`ances of cobalamin metabolism“.
`
`Agents decreasing homocysteine
`concentrations
`
`co-
`serving as
`Compounds
`factors
`in homocysteine catab-
`olism or remethylation may en-
`hance homocysteine metabolism
`and thereby reduce plasma homo-
`cysteine levels in inherited en-
`zymic defects. Thus, vitamin B6
`reduces plasma homocysteine in
`homocystinurics with
`residual
`cystathionine fi—synthase activity,
`and vitamin B12 acts similarly in
`some mutations of
`cobalamin
`metabolism. Betaine and folic acid
`have been shown to efficiently
`reduce plasma homocysteine in
`patients with cystathionine l3-syn-
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`

`
`TiPS — October 1990 I Vol. 11]
`
`thase deficiency who are un-
`responsive to vitamin B6 (Ref. 1).
`
`Folic acid
`Folic acid (5 mg daily) efficiently
`decreases plasma homocysteine
`levels. It reduces elevated plasma
`homocysteine in renal transplant
`recipients and even in those with-
`out overt folate deficiency. Treat-
`ment of healthy subjects with folic
`acid for 14 days significantly re-
`duced
`plasma
`homocysteine,
`especially in persons with high
`pretreatment levels“.
`The marked effect of high doses
`of folic acid on the concentration
`of homocysteine in plasma is im-
`portant. Since moderate homo-
`cysteinemia may provoke vascular
`lesions,
`folic acid may prevent
`atherosclerotic disease in selected
`subjects. This
`intervention is
`particularly
`attractive
`because
`folic acid intake has essentially no
`side-effects“.
`Interestingly,
`the
`effect of folic acid suggests that
`the intracellular
`folate content
`is
`insufficient
`for an optimal
`remethylation of homocysteine.
`This may be a more common state
`than hitherto recognized, as
`it
`may not be detected by estab-
`lished
`laboratory
`procedures,
`including determination of folate
`in serum or erythrocytes’.
`Folic acid probably decreases
`plasma homocysteine levels by in-
`creasing the availability of intra-
`cellular 5-methyltetrahydrofolate,
`thereby enhancing homocysteine
`remethylation (Fig. 2). This re-
`action
`forms
`tetrahydrofolate,
`which enters the pools of reduced
`folates carrying one-carbon units
`via
`the
`serine hydroxymethyl
`transferase reaction (Figs 1 and 2).
`In this reaction, serine is con-
`sumed and glycine formed, as
`would be
`expected
`from the
`moderate reduction in plasma
`serine
`levels and increase
`in
`plasma glycine levels that follow
`folic acid administration".
`
`D-Penicillamine
`D-Penicillamine (D-[3,[3-dimethyl-
`cysteine) is currently used for the
`treatment of heavy metal poison-
`ing, rheumatoid arthritis, hepato-
`lenticular degeneration, cystinuria
`and scleroderma. It is metabolically
`stable, chelates heavy metals, pro-
`duces disulfides and forms a thi-
`azolidine ring with aldehydes and
`ketones. In plasma, penicillamine
`forms symmetrical penicillamine
`
`disulfides and mixed disulfides
`with cysteine, homocysteine and
`plasma proteins. The low mol-
`ecular weight disulfides undergo
`rapid renal excretion, which ex-
`plains the short plasma half-life
`and
`the
`therapeutic effect
`in
`cystinuria“.
`re-
`efficiently
`Penicillamine
`duces both free and protein-
`bound plasma homocysteine in
`homocystinurics“
`and
`total
`plasma homocysteine in patients
`with rheumatoid arthritis, a con-
`dition with normal homocysteine
`pretreatment
`levels". This
`re-
`duction in plasma levels may be
`associated with an intracellular
`homocysteine
`depletion
`suf-
`ficiently pronounced to decrease
`homocysteine remethylation and
`thereby induce methionine de-
`ficiency and secondary effects on
`folate metabolism. If such effects
`occur, penicillamine may act as an
`antifolate agent and may therefore
`interact adversely with metho-
`trexate used in low doses in the
`management of
`rheumatoid ar-
`thritis (see below)“.
`A further clinical implication of
`this work is that penicillamine
`may be a useful means to reduce
`plasma homocysteine.
`
`Adenosine (nucleoside) analogs
`The
`cleavage of S-adenosyl-
`homocysteine to adenosine and
`homocysteine, catalysed by S-
`adenosylhomocysteine hydrolase
`(Figs 1 and 2), is the only known
`source of homocysteine in ver-
`tebrates. Several nucleoside ana-
`logs block this reaction by serving
`either as an inactivator or
`in-
`
`hibitor of the enzyme. In addition,
`some analogs act as substrate and
`are thus converted to the corre-
`sponding S-adenosylhomocysteine
`analogue‘.
`Inhibition
`of
`S-
`adenosylhomocysteine hydrolase
`leads to massive accumulation of
`S-adenosylhomocysteine in iso-
`lated cells, whole animals, and in
`patients. This is important for the
`antiviral effects of
`this class of
`compound‘‘'‘‘’.
`immediate conse-
`The other
`quence of S-adenosylhomocys-
`teine hydrolase inhibition,
`re-
`duction
`of homocysteine
`for-
`mation (Fig. 2), has been studied
`only
`recently.
`Homocysteine
`depletion and inhibition of homo-
`cysteine export have been demon-
`strated in isolated cells exposed to
`nucleoside analogs’°'“. A reduc-
`
`413
`
`tion in plasma homocysteine was
`found in patients with acute
`leukemia treated with 2-deoxy-
`coforrnycin, which indirectly in-
`activates S-adenosylhomocysteine
`hydrolasezzl’.
`lnhibition
`of
`homocysteine formation plays an
`important role in the cytostatic
`action of some nucleoside analogs
`against some“-25 but not all2"'2°
`cell types, and probably mediates
`the differentiation of HL-60 cells
`induced by adenosine dialde-
`hyde”. The consequences of cellular
`homocysteine deficiency are two-
`fold. First, nucleoside analogs may
`induce severe methionine de-
`ficiency, since homocysteine sal-
`vage is
`a significant source of
`methionine in humans3°v3‘. Sec-
`ondly, lack of homocysteine may
`trap reduced folate as 5-methyl-
`tetrahydrofolate because homo-
`cysteine is the methyl acceptor in
`the methionine synthase reaction
`catalysing
`the
`conversion
`of
`5-methyltetrahydrofolate to tetra-
`hydrofolate. In this way, lack of
`tetrahydrofolate may ensue, and
`thereby inhibit folate-dependent
`purine and thymidylate synthesis.
`Both mechanisms
`have
`been
`demonstrated in cultured cells?‘-33.
`lnduced deficiencies of meth-
`ionine
`and
`folates might be
`avoided clinically by appropriate
`supplementation which would be
`expected to
`reduce
`associated
`side-effects. Moreover,
`adverse
`interactions of adenosine ana-
`logues with drugs that also inter-
`fere with folate or methionine
`metabolism such as nitrous oxide
`or methotrexate should be con-
`sidered. This has been suggested
`for the antiviral agent vidarabine
`(9-fl-D-arabinofuranosyladenine)
`and high-dose methotrexate with
`folinic acid rescue-"’°. Reduction in
`plasma homocysteine levels might
`however have beneficial vascular
`effects.
`
`Inhibitors of homocysteine
`remethylation and degradation
`Several important drugs interfere
`with homocysteine metabolism.
`
`Nitrous oxide
`The anesthetic agent nitrous
`oxide was used for a century be-
`fore it was discovered that long-
`term exposure caused megalo—
`blastic and aplastic bone marrow
`changes, anemia and myelopathy.
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`

`
`Folic acid
`
`Folic acid
`
`/
`5- CH3 -THF
`
`Hey
`
`Bu
`
`Met
`
`Nitrous oxide
`
`DHF\W—> THF
`5.10-CH,-THF /
`5- CH 3-THF
`/$3"
`
`Folic acid
`
`M3‘
`
`BIZ
`CH3oob(l)alamin
`c‘‘‘ \ \
`Hcy-SH
`(extracellular)
`
`Ado
`
`H9)’
`9‘.
`Cystathioninc
`$6
`Cysteine
`
`414
`
`TiPS — October 1990 lVol. 11]
`
`Methotrexate
`
`DI1:w?7w
`
`s.mcn,.-nu=
`i
`5. cu ,.m=
`
`HCY
`
`B 12
`
`Adenosine analogues
`
`AdoHcy
`
`Cystathionine
`)6
`Cysteine
`
`D-Penicillamine
`(P-SH)
`
`R-S-S-R 4
`
`5- CH3 -THFTHF'
`
`ctI,uos(tI)ul.msn Mu
`inactive B P
`
`excretion
`
`(extracellular)
`R-S-S-R \£
`Hey-S-S-R
`
`Fig. 2. Proposed or established mechan-
`isms for the effect of various agents on
`the metabolism and plasma level of
`homocystelne. The figure tn the center
`indicates normal metabolism and the six
`panels in the boxes show the effects of
`various drugs on different pathways.
`Sizes of type and arrows indicate the
`concentration of a particular metabolite
`and the flux through the pathway, re-
`spectively. Folic acid probably increases
`the cellular content of 5-melhyltetrahydrofolate (5-CH3-THF), which increases the homocysleine (Hey) remelhylation catalysed by
`methionine synthase (MS). n-Penicillamine (P-SH) forms a mixed disulfide with homocysteine (Hey-Sps-P): this disulfide has a high renal
`clearance. Adenosine analogs are inhibitors of S-adenosylhomocysteine hydrolase (SA). Nitrous oxide oxidizes methylcobtltalamin to
`melhylcob(ll)alamin and thereby irreversibly inhibits methionine synthase (MS). Methotrexale inhibits dihydrofolate reductase (DR),
`thereby inhibiting regeneration of letrahydrololate (THF) from dihydrofolate (DHF). Tetrahydrofofate supplies 5-methyltetrahydrofolate via
`5.10-melhylenetefrahydrofolale (5.10-CH,-Tl-IF). Methotrexate may therefore induce depletion ol 5-methyltetrahydrololate. and inhibit
`homocysteine remethylation catalysed by methionine synthase. Azauridine is a vitamin B6 antagonist, and may inhibit some vitamin B5-
`dependenl enzymes. including cystalhianine ti-synlhase (CS).
`Hey, homocysleine; Met, methionine; Ado. adenosine; P-SH, o-penicillamlne reduced form; Hey-S-S-R, homocysteine mixed disulfide;
`Hcy-S-S-P. a mixed disullide between homocysteine and o-penicillamina: P-S-S-R. o-penicillamine mixed disulfide.
`For other abbreviations, see Fig. 1.
`
`These side-effects are similar to
`the
`symptoms of vitamin 8,2
`deficiency-33.
`Nitrous oxideoxidizes the cob-
`alamine
`species methylcob(I)-
`alamine,
`inactivating specifically
`the enzyme methionine synthase,
`
`without affecting the adenosyl—
`cobalamine cofactor of methyl-
`malonyl-CoA mutase33.
`Inacti-
`vation of methionine synthase
`causes a cascade effect on folate
`metabolism including trapping of
`reduced folates such as 5-methyl-
`
`loss of folate in
`tetrahydrofolate,
`the urine, and reduction in tissue
`folate levels. There is
`a
`sub-
`sequent decrease of folate-depen-
`dent
`purine
`and
`thymidylate
`synthesis, as demonstrated by the
`deoxyuridine suppression (dU)
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`

`
`TiPS — October 1990 IVol. 11 I
`
`test, which reveals changes in
`human bone marrow within 5-6
`hours. In humans, 50% of meth-
`ionine synthase is inactivated in
`about two hours, and there is a
`reduction in plasma methionine
`after 8-24 hours of exposure.
`Megaloblastic
`bone
`marrow
`changes can be detected after 12-
`24 hours of exposure”.
`Data on the effect of nitrous
`oxide on homocysteine metab-
`olism are sparse, but this drug has
`been reported to increase the
`urinary excretion of homocysteine
`in sheep, and increase plasma
`homocysteine levels in fruit bats3.
`We have recently demonstrated
`that nitrous oxide
`induces
`a
`marked increase in plasma homo-
`cysteine within 90 minutes, with a
`concurrent
`increase in urinary
`homocysteine excretion. In patients
`receiving nitrous oxide for 3-6
`hours, plasma homocysteine re-
`mained above normal for at least 7
`days (Ermens, A. A. M. et al., un-
`published data). The homocysteine
`response evolved before other
`early signs of nitrous oxide-
`induced cobalamine inactivation”.
`Thus, significant cobalamine oxi-
`dation and methionine synthase
`inactivation
`occurs
`even after
`short-term exposure, which pre-
`viously has not been regarded as
`harmful. The resultant increase in
`plasma homocysteine observed in
`some patients may in itself be
`detrimental.
`Furthermore,
`the
`homocysteine response may re-
`flect loss of functional cobalamine
`and folate, and may enhance sen-
`sitivity towards antifolate drugs
`such as methotrexate-"‘
`(Ermens,
`A. A. M., PhD Thesis, University
`of Rotterdam, 1990). Since an in-
`crease in plasma homocysteine
`levels is both an early and sensi-
`tive measure of cobalamine oxi-
`dation,
`plasma
`homocysteine
`monitoring could be useful in the
`detection of such effects in the
`clinic.
`
`Methotrexate
`Methotrexate is an antifolate
`drug which has been used exten-
`sively in intermediate and high
`doses in the treatment of leukemia
`and some solid tumors. Low-dose
`methotrexate
`is
`used
`in
`the
`management of some non-malig-
`nant diseases such as rheumatoid
`arthritis and psoriasis.
`Methotrexate acts by inhibiting
`dihydrofolate reductase,
`thereby
`
`blocking the regeneration of tetra-
`hydrofolate
`from dihydrofolate
`(see Ref. 35). This leads to deple-
`tion of reduced folates, including
`5-methyltetrahydrofolate3°-37. Thus,
`methotrexate may also inhibit the
`folate-dependent remethylation of
`homocysteine catalysed by methio-
`nine synthase. This would explain
`the increased homocysteine export
`from cultured cells exposed to
`methotrexate, and the methotrex-
`ate-induced homocysteinemia and
`urinary homocysteine excretion in
`patients”.
`Low-dose methotrexate (25mg
`daily) given to psoriatics induced
`increased plasma homocysteine
`levels, which maximized after
`about two days, and normalized
`within one week”. This shows
`that plasma homocysteine is a
`sensitive measure of the antifolate
`effect.
`(1—13.6g)
`lntermediate doses
`given
`to
`patients with
`solid
`tumors, induced a rapid increase
`in plasma homocysteine within
`hours, which was reversed on
`administration of folinic acid 24
`hours after start of infusion. This
`response was observed following
`several methotrexate doses in a
`single patient‘°. High doses of
`methotrexate (8—33.6 m—2) given to
`children gave a similar response,
`i.e. a rapid increase a few hours
`after start of administration and
`a
`decline
`following
`‘rescue’
`therapy“. This is analogous to the
`results obtained with cultured
`cells”.
`in
`Basal homocysteine levels
`patients with acute lymphoblastic
`leukemia were often above normal
`before treatment, and declined
`markedly
`following
`treatment
`with cytotoxic agents including
`methotrexate (Refsum, H. et al.,
`unpublished). This may be due to
`eradication of proliferating cells
`exporting large amounts of homo-
`cysteine.
`The
`high-dose methotrexate
`regimen also induced a transient
`but marked reduction in plasma
`methionine“ which may contrib-
`ute to the killing of tumor cells or
`toxicity of methotrexate. Plasma
`homocysteine
`response
`and
`methionine depletion may corre-
`late with the therapeutic as well as
`the side-effects of methotrexate,
`including liver toxicity“ and an
`increased incidence of thrombo-
`
`embolism“; plasma homocysteine
`measurements could provide a
`
`415
`
`to serum metho-
`useful adjunct
`trexate
`determination
`in
`the
`management
`of methotrexate
`therapy.
`
`Vitamin B5 antagonists
`Azauridine is an antimetabolite
`interfering with de novo synthesis
`of uridine-5’-monophosphate.
`It
`was initially used for the treat-
`ment of refractory psoriasis, but
`was withdrawn by the FDA in
`1976 because its use was associ-
`ated with an increased incidence
`
`of vascular episodes“. This may
`be due to effects on homocysteine
`metabolism. Azauridine causes
`homocysteinemia, abnormal homo-
`cysteine excretion and a signifi-
`cant increase in serum methionine
`
`levels in patients. Studies in rab-
`bits suggest that it functions as a
`pyridoxal 5’-phosphate antagonist
`and causes homocysteinemia by
`inhibiting vitamin B6-dependent
`cystathionine
`synthesis“. This
`suggests
`that
`supplementing
`vitamin B6 would prevent the in-
`hibition of homocysteine catab-
`olism; determination of plasma
`homocysteine
`may
`identify
`patients at risk of vascular epi-
`sodes.
`Several other drugs also inter-
`fere with the function of vitamin
`B6:
`isoniazid,
`cycloserine, hy-
`dralazine, penicillamine, phenel-
`zine and procarbazine“. Pertur-
`bation of homocysteine metab-
`olism in patients has been demon-
`strated with isoniazid“. In one
`out of six patients given 300mg
`isoniazid daily for one month,
`urinary homocysteine excretion
`was fivefold higher than normal.
`Inhibition
`of
`cystathionine
`metabolism in these patients is
`supported by increased excretion
`of
`this compound after meth-
`ionine loading“.
`
`Otheragents
`Premenopausal women have
`lower plasma homocysteine than
`men
`and
`postmenopausal
`women“, and plasma levels are
`low during pregnancy. However
`there is no conclusive evidence
`that homocysteine metabolism
`and plasma homocysteine levels
`are under the influence of estro-
`gens. Preliminary data in women
`given contraceptive steroids or the
`antiestrogen tamoxifen suggest a
`polymorphic response.
`In some
`women, altered estrogen status
`may cause a decrease, and in
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`

`
`416
`
`others, an increase in plasma
`homocysteine levels. Because of
`the widespread use of contracep-
`tives, and the suggested use of
`tamoxifen as prophylactic inter-
`vention in healthy women at high
`risk of developing breast cancer.
`the effect on plasma homocysteine
`from altered estrogen status is a
`question of major concern to
`public health’.
`drugs,
`Various
`antiepileptic
`particularly phenytoin but also
`phenobarbital, primiclone, carba-
`mazepine, and valproic acid, may
`induce folate deficiency”. The
`activity of methylenetetrahydro-
`folate
`reductase,
`the
`enzyme
`producing
`5-methy|tetrahydro-
`folate,
`is altered in mouse liver
`following exposure to these drugs.
`Preliminary data
`suggest
`that
`phenytoin and possibly carbama-
`zepine may
`increase
`plasma
`homocysteine, but its relation to
`overt
`folate deficiency has not
`been established3.
`Several other drugs are known
`to interfere with folate metab-
`olism or function. These include
`some phenothiazines and tricyclic
`antidepressants, oral contracep-
`tives, possibly some tuberculo-
`static drugs, and antifolate drugs
`such as trimethoprim“. Thiols or
`disulfide forming drugs, such as
`cysteamine and N-acetylcysteine
`are actual candidates as modifiers
`of plasma homocysteine levels,
`due to a possibly unique dis-
`position of the mixed disulfides
`with homocysteine, as demon-
`strated with penicillamine
`in
`humans“.
`
`El
`
`E1
`
`U
`
`Various pharmacological agents
`have been shown to enhance
`homocysteine remethylation and
`urinary
`excretion,
`or
`inhibit
`homocysteine
`production,
`re-
`methylation or catabolism thereby
`affecting plasma homocysteine
`levels. Modulation of homocys-
`teine metabolism and plasma
`concentrations may be an import-
`ant component of drug action.
`
`Agents that reduce plasma homo-
`cysteine (e.g. folic acid, penicill-
`amine) may prevent
`vascular
`disease, while agents increasing
`plasma homocysteine (e.g. nitrous
`oxide, methotrexate, azauridine)
`may provoke vascular episodes.
`Also for some drugs (e.g. nitrous
`oxide and methotrexate), elevation
`of plasma homocysteine is an
`early and sensitive measure of
`drug action and plasma homocys-
`teine has been shown to be use-
`ful in the diagnosis and follow-up
`of some diseases,
`in particular
`homocystinuria, folale deficiency
`and cobalamine deficiency.
`References
`1 Mudd, S. H., Levy, H. L. and Skovby, F.
`(1989) in The Metabolic Basis for Inherited
`Diseases (Scriver, C. R. at al., eds), pp.
`693-734, McGraw-Hill
`2 Wilcken, D. E. L. and Dudman, N. P. B.
`(1989) Hnetnostasis 19 (suppl. 1), 14-23
`3 Ueland, P. M. and Relsum. H.
`(1989)
`I. Lab. Clin. Med. 114, 473-501
`4 Ueland, I’. M. (1982) Pharmacnl. Rev. 34,
`223-253
`5 Kolhouse, ]. F. and Allen, R. H. (1977)
`Proc. Natl Acad. Sci. USA 74, 921-925
`6 Brattstrom,
`L.,
`lsraelsson, B.
`and
`Hultberg, B.
`(1989) Haernostasis
`19
`(suppl. 1), 35-44
`7 Boers, G. H. J. (1989) Haemoslasis 19
`(suppt. 1), 29-34
`8 Kang, S-S.,Wong, P. W. l<.,Cool<,H. Y.,
`Norusis, M. and Messer, J. V.
`(1986)
`I. Clin. invest. 77, 1482-1486
`9 Mallnow, M. R. et nl. (1988) Circ. Res. 79,
`1180-1188
`10 Araki, A. et al. (1989) Atherosclerosis 79,
`139-146
`11 Chadefaux, B. et al. (1988) Lancet ii, 741
`12 Murdoch, J. C., Rodger,J. C., Rao, 5. S.,
`Fletcher, C. D. and Dunnigan, M. G.
`(1977) Br. Med. I. 2, 226-228
`13 Allen, R. H., Stabler, 5. l’., Savage, D. G.
`and Lindenbaum,
`J.
`(1990) Am.
`I.
`Hematol. 34, 90-98
`14 Anon. (1989) Nutr. Rev. 47, 247-249
`15 Joyce, D. A. (1989) Pharmacol. Ther. 42,
`405-427
`16 Kang, 5-S., Wong, 1’. W. K. and Curley,
`K. (1982) Perliatr. Res. 16. 370-372
`17 Kang, 5-S., Wong, P. W. K., Glickman,
`P. B., MacLeod, C. M. and Jaffe, l. A.
`(1986) I. Clin. Pharmacol. 26, 712-715
`18 Wilke, W. S. and Mackenzie, A. H.
`(1986) Drugs 32, 103-113
`19 De Clercq, E. (1987) Biochem. Pharmacol.
`36, 2567-2575
`20 Svardal, A. M.. Djurhuus, R. and
`Ueland, P. M. (1986) Mol. Pharmacol. 30,
`154-158
`21 Svardal, A. M., Djurhuus, R., Relsum,
`H. and Ueland, P. M. (1986) Cancer Res.
`
`I. 242.
`
`TiPS - October 1990 [VOL 11]
`46. 5095-5100
`22 Kredich, N. M. et al. (1981) Clin. Res. 29,
`541A
`23 Hershfield, M. 5. (1984) Cancer Treat.
`Syrup. 2, 29-32
`24 Kim, l-K., Aksamit, R. R. and Cantoni,
`C.
`L.
`(1932)
`I. Biol. Chem.
`257,
`14726-14729
`25 Wolfson, C., Chisholm, J., Tashjian,
`A. H. J., Fish, 5. and Abeles. R. H. (1986)
`I. Biol. Chem. 261, 4492-4498
`26 Djurhuus, R., Svardal, A. M. and
`Ueland, P. M. (1989) Clncer Res. 49,
`324-330
`27 De Clercq, E., Cools, M. and Ba|1arini,J.
`(1989)
`Biachem.
`Pharmacol.
`38,
`1771-1778
`28 Cools, M., Hasobe, M., De Clercq, E.
`and Borchardt, R. T.
`(1990) Biocitem.
`Pharmacol. 39, 195-202
`29 Pilz, R. 3., Van den Berghe, C. and Boss,
`G. R. (1987) Blood 70, 1161-1164
`30 Cantoni, G. L., Aksamit, R. R. and Kim,
`I-K. (1982) N. Engl. I. Med. 307, 1079
`31 Boss, C. R. and Pilz, R. B. (1984) I. Clin.
`Invest. 74, 1262-1268
`32 Boss, G. R.
`(1987) Biachem.
`425-431
`33 Nurm,J. F. (1987) Br. I. Amreslh. 59, 3-13
`34 Ermens, A. A. M., Schoester, M.,
`Spijkers, L. J. M., Lindemans, J. and
`Abels,
`J.
`(1989) Cancer Res.
`49.
`6337-6341
`35 Matherly, L H., Seither, R. L. and
`Goldman. l. D. (1987) Pharmncal. Ther.
`35. 27-56
`36 Allegra, C. J., Fine, R. L., Drake, J. C.
`and Chabner, B. A. (1986) I. Biol. Chem.
`261, 6478-6485
`37 Baram, J., Allegra, C. J., Fine, R. L. and
`Chabner, B. A. (1987) J. Clin. Invest. 79,
`692-697
`38 Ueland, P. M., Relsum, H., Male, R. and
`Lillehaug, J. R. (1986) I. Natl Cancer inst.
`77, 283-289
`39 Refsum, H., Helland, S. and Ueland,
`P. M. (1989) Clin. Phnrmacol. Tiler. 46.
`510-520
`and
`P. M.
`40 Relsum, H., Ueland,
`Kvinnsland, S.
`(1986) Cancer Res. 46,
`5385-5391
`41 Broxson, E. H., Stork, L. C., Allen, R. H.,
`Stabler, S. P. and Kolhouse, J. F. (1989)
`Cancer Res. 49, 5858-5862
`and
`l.
`42 Barak, A. T., Tuma, D.
`Beckenhauer, H. C. (1984) I. Am. Coll.
`Nuir. 3, 93-96
`43 Brattstriim, L., Ueland, P. M. and
`Refsum, H. (1988) N. Engl. I. Med. 319,
`443-444
`44 Drell, W. and Welcli, A. D.
`(1989)
`Pharmacol. Ther. 41, 195-206
`45 Krishnaswamy, K. (1974) lnt. J. Vitam.
`Nulr. Res. 44, 457-465
`46 Boers, G. H. J. (1988) in Genetic Suscepti-
`bility to Environmental Factors 1 Chal-
`lenge for Public Intervention (Smith, U.,
`Eriksson,S. and Lindgirde, F., eds), pp.
`35-42, Almqvist & Wiksell International
`47 Lambie, D. C. and Johnson, R. H. (1985)
`Drugs 30, 145-155
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318
`
`Lilly Ex. 2068
`Sandoz v. Lilly IPR2016-00318

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