throbber
Petitioner Amerigen Pharmaceuticals Ltd.
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 1
`
`

`
`Petitioner Amerigen Pharmaceuticals Ltd.
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 2
`
`

`
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 3
`Petitioner Amerigen Pharmaceuticals Ltd.
`
`

`
`TARGETING PEPTIDES AND DRUGS TO THE CNS
`
`139
`
`0
`
`O
`
`'
`
`C’
`
`0
`
`NH—CH—é/
`\N
`\
`CH2
`
`N
`H
`
`R1
`
`'92
`NH
`Q’C\NH2
`
`(a) pyrogluiamyl-histidyl-prolinamid2
`(TRH)
`R1 = H. R2 :H
`
`(b) pymglutamyl-histidyl-3 methylprolinamide
`(Pyr-His-MepNI-I2, RX 74355)
`R] : H, R2 = CH3
`
`(c) pyroglutamyl—hisIidyl-3.3 dimethylprolinamide
`(Pyr-His-Dmp-NH2. RX T7368)
`R1: CH3, R2 2 CH3
`
`FIG. 3. Structure of TRH and related analogues RX 74355 and
`RX 77368. The substitution of the methyl groups into the proline
`residue increases the plasma half-lives and the central eflectiveness
`of the molecules.
`
`3. Enhancing or maintaining reactivity with existing BBB
`transport mechanisms.
`4. Retaining central nervous activity.
`5. Increasing the stability in brain extracellular fluid and
`reducing reactivity with efllux transport mechanisms in the
`CNS.
`
`A number of chemical modifications can be carried out to
`
`improve the CNS activity of drugs. For example the peptide
`thyrotropin
`releasing
`hormone
`(TRH—pyroglutamyl-
`histidyl-prolinamide) has a short plasma half life due to
`circulating peptidases. To extend the half-life, methyl
`groups may be added to the prolinamide residue to produce
`a 3-methylprolinamide (RX74355) and a 3,3-dimethy1proli-
`namide (RX77368) (Brewster et al 1981, 1983) (Fig. 3).
`These methyl substitutions increase the lipid solubility of
`the molecule and also the molecular volume. The substitu-
`
`tions also increase the first order half-lives in human plasma
`from 33 (TRH) to 210 (RX74355) and 1080 min (RX77368)
`
`(Table 2). The central nervous activity of the TRH
`analogues is also enhanced (Brewster et al 1981; Brewster
`1983), possibly as the result of increased brain penetration
`although an enhanced central potency should also be
`considered.
`
`Other simple modifications to peptides that may be made
`are to amidate the C-terminus and acetylate the N-terminus
`both of which confer an increased resistance to exopepti-
`dases.
`In addition acetylation of the N-terminus also
`increases lipid solubility significantly.
`An excellent example of increasing brain uptake by
`enhancing lipid solubility and reducing hydrogen bonding
`capacity is the chemical conversion of morphine to heroin
`(diacetyl-morphine). Substitution of the two hydroxyl
`groups of morphine by acetyl groups in heroin increases
`the brain uptake over twenty-fivefold (Oldendorf 1974). As
`a general rule for each pair of hydrogen bonds removed
`from a molecule there is a log order increase in BBB
`permeabilty. Once within the brain, heroin is rapidly con-
`verted to monoacetyl morphine and more slowly to mor-
`phine. Thus the rapid brain entry of heroin in comparison
`with morphine makes it a favoured drug of abuse and
`presumably enhances its addictive potential.
`In the case of many peptides
`the introduction of
`D-isomers of the naturally occurring amino acids in to the
`peptide sequence can greatly enhance the plasma half life of
`the peptide. Again the reactivity with plasma peptidases is
`greatly reduced. For example octreotide (D-Phe-Cys-Phe-
`D-Tyr-Lys-Thr-Cys-Thr-OH : sandostatin : SMS 201-995)
`is an octapeptide analogue of somatostatin Ala-Gly Cys-
`Lys-Asn-Phe-Phe-Trp-Lys-Thr-Phe-Thr-Ser-Cys-OH). The
`attenuated peptide with the D substitutions, D-Phe at
`position two of somatostatin and D-Trp at position five,
`extends the first order half-life of somatostatin from 2-3 min
`
`in human plasma to l 13 min (Lamberts 1987). The substitu-
`tion of D-Phe at position four also increases the growth
`hormone inhibiting potency of sandostatin compared with
`somatostatin by some 45 times. D-Amino acid substitutions
`also have marked efieets on the plasma half-lives of
`encephalin analogues. Examples are two analogues of
`leucine encephalin (Tyr-Gly-Gly-Phe-Leu), namely DADLE
`
`Table 2. First-order half-lives (min) of TRH, RX74355, and RX77368. There are
`significant species diflerenees in the rates at which plasma and a brain homogenate
`will break down TRH and its analogues.
`
`TRH
`
`RX74355
`(Methylproline)
`
`RX77368
`(Dimethylproline)
`
`Rat
`Plasma
`Brain homogenate
`Dog
`Plasma
`Brain homogenate
`Man
`Plasma
`Brain homogenate
`Mouse
`Plasma
`Brain homogenate
`
`22
`9
`
`>l500
`11
`
`33
`18
`
`215
`12
`
`From Brewster (1983).
`
`I14
`54
`
`> 1500
`90
`
`210
`66
`
`> 1500
`28
`
`390
`190
`
`> 1 500
`174
`
`1080
`168
`
`> 1 500
`150
`
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 4
`Petitioner Mylan Pharmaceuticals Inc. — Exhibit 1031 — Page 4
`
`

`
`140
`
`DAVID J. BEGLEY
`
`Ala-GIy-Cys-Lys—Asn-Phe-PherTgLys-Thr—Phe—Thr~Ser-Cys—OH
`Somalostatin“
`
`D-Phe—Cys—Phe-D-TQLys-Thr-Cys-Thr—OH
`
`Ac—D-Phe—Cys—Tyr-D—T1;p-Lys-Val-Cys—Thr-NH;
`
`D»Phe»Cys—Tyr-D~T[p;Lys—Val-Cys-TrpNH2
`
`D-Phe—Cys-Tyr—D—Tgg-Lys-Val—Cys-Thr—Nl-lg
`
`RC12l
`
`0
`
`0.05
`
`0.1
`
`0.15
`
`0.2
`
`0.25
`
`Unidirectional cerebrovascular
`influx constant
`
`(/JL g" min“)
`
`FIG. 4. Unidirectional cerebrovascular permeability constants (Kin) of some somatostatin analogues. The brain uptakes
`are determined by intravenous bolus injection techniques. The structure of the analogues is given with that of
`somatostatin for comparison. Unfortunately the brain uptake of somatostatin cannot be determined by a comparable
`method as it is very unstable in plasma. Data from Begley et al (l992b) and Banks et al (1990).
`
`(D-Alaz, D-Leus-encephalin) and dalargin (Tyr-D-Ala-Gly-
`Phe-Leu-Arg) both of which have greatly extended half-lives
`in plasma compared with the parent peptide.
`A number of somatostatin analogues exist where the
`plasma half-life is extended and the lipid solubility is altered
`thus modifying two factors which influence brain uptake,
`plasma half life and biological potency of the molecules.
`Intravenous bolus injection studies have been carried out to
`measure the brain uptake of these peptides, (Banks et al
`1990; Begley 1992b, 1994). These somatostatin analogues
`are amphiphilic and their brain uptakes are non-saturable
`suggesting that
`their penetration into the brain will be
`passive. However changes in the molecular structure of the
`analogues produce significant changes in brain uptake. Fig. 4
`illustrates the brain uptakes of four octapeptide analogues
`of somatostatin, determined by an intravenous bolus injec-
`tion technique (Banks et al 1990; Begley 1992b, 1994) which
`have D-Phe and D-Trp substitutions in the molecule. Com-
`pared with RC 12]
`the substitution of a Trp residue at
`position 8 compared with RC 160,
`thus introducing an
`additional aromatic side chain into the molecule, approxi-
`mately doubles the cerebrovascular permeability constant.
`Acetylating the N-terminus of RC 121 as in RC 161, and
`
`therefore increasing the lipid solubility, increases the perme-
`ability constant almost sixfold. The peptide RC 160 has
`marked and prolonged analgesic actions after intravenous
`administration (Eschalier et al 1990).
`Using synthetic peptide sequences which have hydropho-
`bic properties may provide a mechanism for inserting a
`peptide into the cell membrane (Begley 1994). For example a
`number of naturally-occurring peptides have hydrophobic
`regions which demonstrate a natural aflfinity for cell mem-
`branes. Mellitin, a component of bee venom (Kaiser &
`Kezdy 1987),
`is a 26-amino acid peptide (Fig. 5). The N-
`terminal amino acids 1-20 contain two :1-helical regions
`1-10 and 13-20 which are hydrophobic and insert
`the
`molecule into the cell membrane, the lysine residues 21-26
`are highly cationic and are thought to open up pores thus
`permeabilizing the membrane (Kaiser & Kezdy 1987).
`Signal peptides are similar hydrophobic regions in a pre-
`pro-peptide or protein which enable the entire molecule to
`be inserted through the unit membrane of the endoplasmic
`reticulum. The signal amino acid sequence is not conserved
`between pre-pro-proteins and thus the common feature is
`the hydrophobicity (lipohilicity) of the region (Engelman &
`Steitz 1981; Emr & Silhavey 1983; Begley 1994). The
`
`10
`15
`N-E ily-lle—Gly-Ala—Val-Leu-LyS-Val—Leu-ThriThr—Gly-ll Eu-Pro-Ala-Leu-
`+
`
`25
`20
`lle—Ser—Trp-Ile Lys—Arg-Lys-Arg—Gln—Gln NH2
`+
`+
`+
`+
`
`FIG. 5. Amino acid sequence of mellitin. The hydrophobic areas which insert into the cell membrane are boxed.
`Positively-charged side chains which disrupt the cell membrane are indicated with + .
`
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 5
`Petitioner Mylan Pharmaceuticals Inc. — Exhibit 1031 — Page 5
`
`

`
`TARGETING PEPTIDES AND DRUGS TO THE CNS
`
`141
`
`(Dalargin) /'
`
`Tyr—DAla-Gly-Phe-Leu~Arg
`
`Poly—butyl—cyanoacrylate nanoparticle
`
`ba -80
`‘C
`
`Polywr
`
`Fig. 6. Schematic diagram of a nanoparticle. The dalargin is
`absorbed onto the surface of the nanoparticle which is then
`coated with polysorbate-80.
`
`function of the signal sequence is thus to enable a large polar
`protein or peptide to traverse a unit membrane without
`damage to either the pre-pro-protein or the membrane.
`Indeed passive permeation of the BBB may not be limited
`to small molecules. Colloidal polymer particles (nanoparti-
`cles) may be formed from poly(buty1cyanoacrylate) with an
`average diameter of 230nm. Nanoparticles have been used
`to deliver the peptide dalargin to the CNS (Fig. 6) (Engel-
`man & Steitz 1981; Emr & Silhavey 1983; Kreuter et al
`1995). Dalargin is a hexapeptide analogue of encephalin
`which is stable in plasma but has little central analgesic
`action when injected intravenously. If dalargin is absorbed
`onto the surface ofnanoparticles and these particles are then
`coated with the detergent polysorbate-80 and the complex
`injected into mice, a pronounced analgesic effect is obtained
`reaching a maximum in 45 min (Fig. 7). Little elfect
`is
`produced with dalargin, nanoparticles or polysorbate-80
`alone. The effect
`is also dose-dependent with a greater
`absorption of dalargin onto the particles and is reversible
`by naloxone (Engelman & Steitz 1981; Emr & Silhavey 1983;
`Kreuter et al 1995). Presumably the detergent enables the
`particles to penetrate the BBB and the dalargin is released
`from the nanoparticles in effective amounts within the brain.
`In contrast, attempts to use liposomes to deliver drugs to the
`brain have been universally unsucessful (Pardridge 1991).
`
`(%)
`
`
`Maximumpossibleeffect
`
`O
`
`15
`
`30
`
`45
`
`60
`
`75
`
`90
`
`Time (min)
`
`FIG. 7. Analgesia in mice produced by dalargin-loaded nanoparti«
`cles. Analgesia is expressed as the percent of the maximally possible
`efl"ect after intravenous injection of the indicated dose. (11: 6.
`mean 3: s.d.)
`
`Exploiting existing transporters
`As mentioned earlier the presence of a blood—brain barrier
`to polar molecules dictates that a number of transporters
`must be present in the cerebral endothelium in order to
`supply the brain with an adequate supply of nutrients. The
`kinetic properties of some of these transporters are shown in
`Table 3.
`
`The glucose carrier GLUT-1 has a very large capacity to
`transport ,3-D-glucose across the BBB. The intestinal Na+/
`D-glucose transporter will also transport fi—glycosides but
`not as effectively as the preferred substrate glucose. Given
`that GLUT-1 and the intestinal transporter have a similar
`
`Table 3. Kinetic values for some transport systems present at the BBB.
`
`
`
` Representative substrate Transporter K,,, (MM) Vmx (nmol min“' gr‘) K) (ML g" min'')
`
`
`
`Glucose
`Hexose: Glut-l
`11000 :: 1400
`1420 :: 140
`Lactic acid
`Monocarboxylie acid
`1800 :: 600
`91 :: 35
`Phenylalanine (apparent)
`System-Ll
`30-0 :: 1-0
`14-0 :: 4-0
`Phenylalanine (real)
`System-Ll
`ll-0 :: 2-0
`25-0 :: 6-0
`Arginine (apparent)
`Basic amino acid
`40-0 i 24
`5-0 :: 3-O
`eucine encephalin
`Peptide specific
`39-0 :: 32
`160-0 :: 220*
`Arginine vasopressin
`Peptide specific
`2-08 :: 0-32
`5 -49 :: 0-74*
`Adenosine
`Nucleoside
`25-0 :: 3-0
`0-75 :: 0-08
`Adenine
`Purine base
`1 1-0 :: 3
`0-5 :: 0-O9
`
`44-0 :: 14-0
`8-0 3: 1-0
`
`0062 :: 0-086
`0-02] : 0044
`
`*pmol min" g".
`The Km value is the concentration of substrate at which the transporter is half-saturated and indicates the affinity of the substrate for the
`System. The Vmax is the maximal transporting capacity of the system. The Glut-l transporter has a high aflinity and a high capacity for
`glucose. As explained in the text many amino acids share a common transport system and will have ditferent affinities (Km) for the systems;
`bficause ofthis they will compete with each other for transport. Thus an amino acid will have a real Km and Vmax where their kinetic values are
`dfitermined in the absence ofeompeting amino acids and apparent values when competing amino acids are present. The Kd is a measure ofthe
`Passive diffusional component to the movement ofa solute and is significant in the case of the amino acids but is much smaller in the ease of
`the more polar peptides. Means i s.e.m. Compiled from various sources.
`
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 6
`Petitioner Mylan Pharmaceuticals Inc. — Exhibit 1031 — Page 6
`
`

`
`142
`
`DAVID J. BEGLEY
`
`1
`
`2
`
`3
`
`4
`
`5
`
`SH HS
`
`0
`ll
`7
`l_
`H2N-Tyr-DCys-Ser-Phe-DCys-Gly—C-NH2
`D-G|e—fl-(1->0)
`
`if
`rS—s7
`H2N-Tyr-DCys—Ser-Phe-DCys-Gly-C-NH2
`D-Glc-ll-(1—>O)
`
`s —— s
`
`0
`n
`_l
`F
`HZN-Tyr-LCys-Gly—Phe-LCys-Ser-C-NH2
`D-G|c—fi-(1-r0)
`
`E
`’—SH HST
`H2N-Tyr-DCys-Gly-Phe-DCys—Ser—GIy—C—NH2
`D-Glc-[5-(1—>O
`6 + p,
`
`if
`I"S“‘S‘l
`H,N-Tyr—DCys-Gly-Phe-DCys- er—GIy-C-NH2
`D-G|c—B~(1~>O
`5 + I‘
`
`6
`
`7
`
`8
`
`9
`
`10
`
`—
`
`2
`
`1?
`H81’
`H2N-Tyr-DPen-G|y-Phe-DPen—Ser-Gly-C-NH2
`D-Glc—fi-(1-O)——J
`r
`*rs—sw’
`H2N-Tyr-DPen-Gly-Phe-DPen-Ser—Gly-C-NH2
`D-G|c—p—(1~>O)
`
`H S
`
`S H
`
`O
`ll
`“l
`l_
`HZN-Tyr-Dcys-GlynPhe-DCys—Ser—Gly-C-NH2
`H
`6 + ll
`
`S —~ 3
`
`O
`I’
`‘l
`u
`H2N-Tyr-DCys-G|y-Phe-DCys-fer-Gly-C—NH2
`H
`5+1:
`
`HZN-Tyr-DPen—G|y-Phe—DPen—C—NH2
`DPDPE 6
`
`ineffective
`is
`FIG. 8. Some analogues of DPDPE (D-pen” encephalin). DPDPE is a 6-receptor agonist but
`intraperitoneally. Analogues 4 and 5 and 8 and 9 are 6- and H-receptor agonists. However only analogues 4 and 5
`produce analgesia determined by the tail flick test after intraperitoneal administration. From Polt et al (1994).
`
`structure it might be possible that peptide-fl-D-glycoside
`conjugates may be acceptable to the brain glucose transpor-
`ter (Polt 1994). Some glycopeptides administered intra-
`peritoneally as L-serinyl-,6-D-glyeoside analogues of Met5
`encephalin (Fig. 8) have been shown to be transported
`across the BBB and bind to u- and 6-opioid receptors in
`the brain. The glycopeptide encephalin analogues 4 and 5
`produce a marked and long-lasting analgesia after intraper-
`itoneal administration as determined by tail-flick and hot-
`plate assays in mice (Fig. 9). This analgesia is reversible with
`naloxone (Polt 1994).
`It
`is suggested that GLUT-1 is
`responsible for
`transporting the glyeopeptide into the
`CNS.
`Interestingly,
`these glyeopeptides have a reduced
`lipohilicity compared with the native encephalins but the
`conjugate has reactivity with both the glucose transporter
`
`Antinociception
`
`(%)
`
`0
`
`20
`
`40
`
`60
`Time (min)
`
`80
`
`100
`
`120
`
`Fro. 9. Analgesia produced by DPDPE glycopeptide analogue 5.
`Intrapcritoneal administration of this pe tide produces dose-related
`and long-lasting antinociception in a 55 C tail-flick test. From Poll
`et al (1994).
`
`and with opioid receptors within the brain (Polt 1994). The
`brain transport appears to be specific for ,3-0-linked glyco-
`sides as oz-linked and N-linked and O-acyl linked glucose
`conjugates (Fig. 8) do not appear to cross the BBB and have
`no analgesic effects. Also only glycosides linked via the
`serine residue showed any CNS activity. In this connection
`it is interesting to note that morphine-6-glucuronide formed
`naturally in the liver is 10-50 times more potent in producing
`analgesia than morphine itself and that morphine may act as
`a prodrug with the 6-glucuronide producing a significant
`part of the analgesic effect. There is thus a good possibility
`that glycosylation might be applicable to the delivery of a
`range of drugs across the BBB.
`System-L transporting neutral amino acids into the CNS
`is another obvious target for drug analogues or complexes
`which might have a reactivity with this system. To date, the
`most successful exploitation of system-L is the delivery
`of L-dopa to the brain for the treatment of Parkinsonism.
`Dopamine given orally is ineffective as it is not a substrate
`for system-L and thus does not enter the brain readily. It is
`also subject to rapid metabolism at the level of the BBB by
`MAO and COMT. Fortunately L-dopa is a substrate for
`system-L and the enzyme L-amino acid decarboxylase
`(AAD or dopa decarboyxlase) within the BBB will convert
`L-dopa to dopamine during passage through the barrier.
`The dopamine can then be taken up by cells in the substantia
`nigra to replace their missing neurotransmitter. Although
`some further dopamine must be lost as the result of MAO-B
`and COMT activity in the CNS suflicient appears to reach
`the nerve cells.
`
`The antineoplasic alkylating agent melphalan also has a
`reactivity with system-L (Grieg 1992). It
`is chemically a
`nitrogen mustard derivative of phenylalanine (Fig. 10).
`However its reactivity with system-L is only some 20% of
`that of phenylalanine and endogenous amino acids will
`compete for transport. The steric requirements for system-
`L are that the substrate must possess an O-8IT1lI1O group and
`
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 7
`Petitioner Mylan Pharmaceuticals Inc. — Exhibit 1031 — Page 7
`
`

`
`TARGETING PEPTIDES AND DRUGS TO THE CNS
`
`143
`
`Cfia
`
`>—:CH2{<
`
`Leucifle
`
`CH3
`
`H3N‘
`
`Coo‘
`
`H3N+
`
`H2
`
`Phenylalanine 000.
`
`C
`
`H
`
`H‘-,N*
`
`H2~—<
`
`L-DOPA
`
`COO
`
`\ H3N+
`C
`
`H
`
`,
`
`C
`9/ \C coo
`BCH : 2-amino-bicyclo[2,2,1]-heptnne-2-carboxylic acid
`Definitive specific substrate for the L-system of Christensen
`
`H3N*
`
`coo‘
`
`H3N*
`
`coo’
`
`Baclofen
`
`Gabapentin
`
`Cl
`
`CH2
`
`C|——CH2
`
`CH2
`
`CH2
`
`>~@-2+
`
`H3N"
`
`COO‘
`
`Melphalan
`
`CI-——CH,
`
`CH
`
`CI—C H2?CH2
`
`’>N
`
`H,N*
`
`COO‘
`
`D, L NAM : (D,L-Zaamino-7-bisl(2-ch|oroethyl)amino]-
`l,2,3,4-tetrahydro-2-napthoic acid)
`
`FIG. 10. Substrates known to be transported by system-L at the
`blood—brain barrier. All of the structures have in common an amino
`and a carboxylic acid group attached to a single carbon atom, with
`the exception of baclofen and gabapentin. In the case of these two
`drugs it is thought that the amino and carboxylic acid groups have
`suflicient flexibility to make the molecules sterically acceptable to
`the transport mechanism. BCH is the defining substrate for system-
`L and only reacts with that amino acid transporter.
`
`a carboxylic acid group attached to the same carbon atom
`plus a hydrophobic side group. However there are some
`known exceptions (Fig. 10). The size of the hydrophobic side
`group does not appear to be a hindrance to transport and
`the amino acids phenylalanine and tyrosine have a high
`aflinity for the transporter. The importance of the relative
`positions of the a-amino and carboxylic acids groups is
`emphasized by the fact that when the small peptide glycyl-
`ieucine is formed reactivity with system-L is lost. Other
`Substrates with significant afiinity for system-L have been
`designed (Grieg 1992). The nitrogen mustard DL-2-amino-
`7-bis((2—chloroethyl)amino)-1,2,3,4-tetrahydro-2-naphthoic
`acid (DL-NAM) is a good example. It possesses an addi-
`tional naphthoic side chain compared with melphalan
`(Fig. 10), which renders it more hydrophobic and confers
`0n the molecule 20 times the affinity for system-L than
`Phenylalanine and 100 times the affinity of Melphalan and
`L'd0pa for the transporter. Additional advantages of DL-
`NAM are that it has 1.5 times the alkylating activity of
`melphalan and it is only 20% bound to plasma proteins
`°°mpared with 80% for mephalan giving it
`a high
`
`therapeutic index (Grieg 1992). Its high aflinity for system-
`L means that it competes effectively with endogenous amino
`acids to achieve an eflicient brain uptake even though the
`Vmax is lower.
`The antiepileptic drug gabapentin (neurontin) also enters
`the CNS via system-L even though its does not fully satisfy
`the strict steric requirements suggested above. Its uptake
`into the CNS is inhibited by BCH the model substrate for
`system-L (Fig. 10). Gabapentin is not a substrate for the
`glutamate transporter and is thus unusual in that it is a 7-
`amino acid that is acceptable to system-L. It is suggested
`that the amino and carboxylic acid groups are sufliciently
`flexible to render it sterically acceptable to system-L. The
`binding site for gabapentin in nervous tissue is distinct from
`system-L as leucine is more potent in displacing it than is
`BCH. It acts as an inhibitor of branched-chain amino
`transferase and both it and leucine act to stimulate gluta-
`mate dehydrogenase in the CNS.
`A similar situation pertains for the muscle relaxant baclo-
`fen (4-amino-3-(p-chlorophenyl)-butyric acid (Fig. 10) (Van
`Bree et al 1988). Baclofen acts at the spinal and supraspinal
`levels and is transported into the CNS by system-L.
`A number of specific uptake mechanisms for peptides
`exist at the BBB (Begley 1994). Those for leucine encephalin
`and arginine vasopressin have been kinetically described as
`high aflinity low capacity systems (Zlokovic et al 1989, 1990)
`as shown in Table 3. The uptake mechanism for leucine
`encephalin is remarkably specific showing little afiinity for
`fragments of the molecule with almost the entire pentapep-
`tide being sterically required. Fig. 11 shows the results of
`inhibition studies carried out in an in—situ brain perfusion
`technique with the addition of a number of inhibitors and
`peptide fragments. Under the conditions of perfusion the
`
`Kml;IL9"min") UCLU
`
`3Q!
`._l
`
`+SmMTG
`
`+2mMBacitracin
`
`+0.5mMBestatin
`
`+SmMTyrosine
`
`+2mMLeuEnc
`
`+SmMTGG
`
`+SmMTGGP
`
`+SmMGGPL
`
`
`
`
`
`+0.044ICI174846
`
`
`
`+0.053mMDAGO
`
`1 1. Specificity of leucine encephalin uptake at the blood—brain
`1:16.
`barrier. Inhibition studies to determine the specificity of the uptake
`mechanism for leucine encephalin at the blood—brain barrier of the
`guinea-pig hippocampus determined by an in—situ vascular brain
`perfusion method. Vertical axis unidirectional cerebrovascular con-
`stant Kin pL g“ min'1:ks.e.m. Horizontal axis Kin for leucine
`encephalin tracer alone (control); plus 2mM bacitracin, 0.5mM
`bestatin. 5 mM L-tyrosine, 2mM leucine encephalin, 5 mM tyrosyl-
`glycine, 5mM tyrosyl-glycyl-glycine, 5mM_ t_yrosyl-glycyl-glycyl-
`phenylalanine, 0.044 mM ICI 174846 (a 6 Oplold ligand), 0.053 mM
`DAGO (a ,u opioid |igand.). Only the addition of 5mM leucine
`encephalin produces a significant
`inhibition of the Km value
`(P < 0-01) and 5 mM tyrosyl-glycyl-glycyl-phenylalanine (P < 005).
`Data from Zlokovic et al (1987, 1989). Means :: s.e.m.
`
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 8
`Petitioner Mylan Pharmaceuticals Inc. — Exhibit 1031 — Page 8
`
`

`
`144
`
`DAVID J. BEGLEY
`
`(3HTyr)-encephalin molecule is stable as the addition of
`2.0 mM bacitracin or 02 mM bestatin to the perfusate has no
`effect on the cerebrovascular permeability constant, Kin.
`Significant reduction of the Km value, indicating competi-
`tive inhibition, was only produced by intact leucine ence-
`phalin and des-leucine encephalin. DAGO and ICI 174846,
`ligands for the p- and 6-opioid receptors, respectively, had
`no effect in the K-,,, indicating that the carrier is distinct from
`the receptor producing the central nervous effects of these
`opioid peptides (Zlokovic et al 1989).
`Transporters for macromolecules also exist at the BBB
`which might be utilised as vectors to transport material into
`the CNS. Strategies utilizing the transferrin receptor in this
`way have been attempted (Friden et al 1991, 1993; Pardridge
`et al 1991). A monoclonal antibody,
`to the transferrin
`receptor OX26 has been utilised as this avoids competition
`between any administered transferrin and endogenous
`transferrin in the experimental animal. A variety of mole-
`cules can then be attached to the monoclonal antibody. In
`experimental animals this approach has been used to deliver
`horseradish peroxidase, nerve growth factor (NGF), meth-
`otrexate and vasoactive intestinal polypeptide (VIP), across
`the blood-brain barrier.
`In some respects these studies
`remain controversial as there is evidence to suggest that in
`the intra-endothelial transport of iron by transferrin at the
`BBB, the iron is decoupled from the transferrin within the
`cells and the transferrin molecule itself may not necessarily
`be transcytosed. (Taylor & Morgan 1990; Ueda et al 1993).
`However, the NGF-OX26-transferrin complex may act as a
`vector to allow large proteins such an NGF to enter a
`protected compartment within the endothelial cells and the
`whole complex may not necessarily have to be exocytosed at
`the abluminal membrane.
`
`Efflux mechanisms
`
`As mentioned earlier the efflux pump P-glycoprotein is
`expressed constitutively at the luminal surface of the cere-
`bral endothelial cells forming the BBB. A number of lipid
`soluble cytotoxic agents, such as Vincristine and vinblastine
`and other substrates for P-glycoprotein, do not exhibit the
`brain uptakes that would be predicted by their lipid solu-
`bility (Begley 1992) and are outliers on the plot shown in
`Fig. 2. One strategy for enhancing the uptake of substrates
`
`for P-glycoprotein is to employ non-competitive or compe_
`titive inhibitors of the eflfux pump. These agents will reverse
`multidrug resistance in peripheral tissues that have acquired
`it and thus have potentially a wide application in cancer
`chemotherapy. Table 4 shows the effect of a number of
`substrates and inhibitors of Pgp activity on the accumula-
`tion of [3H]colchicine in-vitro into a cultured monolayer of
`an immortalized clone of rat brain endothelial cells (Begley
`et al 1994). Similar results have also been obtained in-vivo
`by administering therapeutic doses of Vincristine to guinea-
`pigs (Begley & Evans 1992). In this study co-administration
`of Vincristine and [3H]colchicine increases BBB uptake of
`colchicine compared with control animals receiving tracer
`colchicine alone, suggesting an inhibition of Pgp activity in
`the cerebral endothelium.
`
`A spectrum of efflux pumps exists within the CNS and
`contributes to the homeostasis of the brain extracellular
`
`fluid. These pumps may be located in the cerebral endothe-
`lium and also at the choroid plexus. For example penicillin,
`p-aminohippuric acid and AZT (azidothymidine), are
`pumped out of cerebrospinal fluid (CSF) at the choroid
`plexus by an organic acid pump which is probenecid-sensi-
`tive (Spector & Lorenzo 1974). Probenecid can therefore be
`used to increase the CSF levels of these drugs. Amino acids
`are cleared rapidly from CSF by saturable transport
`mechanisms (Davson et al 1982) as are a number of peptides
`(Begley & Chain 1988, 1992; Banks & Kastin I992). Amino
`acids with neurotransmitter function have very low free
`concentrations in CSF. Thus the design of peptide- or
`amino acid-based drugs with a reduced affinity for these
`eflfux mechanisms or the inhibition of these efflux mechan-
`isms will lead to an increase in CNS levels. There are also free
`
`peptidases in CSF (Begley & Chain 1988, 1992; Begley 1994)
`which can hydrolyse peptides, for example leucine encepha-
`lin and angiotensin II. The substitution of D-isomers into the
`peptide confers resistance to the action of these enzymes in
`the same manner as with plasma peptidases.
`
`Opening and Permeabilizing the BBB
`
`A number of techniques exist which enable the permeability
`of the BBB to be modified. Most of these are non-selective
`
`and open the barrier to a range of solutes of varying
`molecular weight.
`
`Table 4. Inhibition of Pgp activity in RBE4 cells.
`
`Inhibitor
`Vincristine
`AZT
`Chlorpromazine
`Verapamil
`
`Concn (pm)
`50
`50
`50
`100
`
`[3H] Colchicine uptake (mL (pg protein)'*')
`
`Control
`136411-19
`141610-82
`12911093
`12-45] l~l9
`
`Experimental
`533918-62
`339313-24
`25751 1'88
`17461862
`
`% control
`39l**
`232**
`200**
`140*
`
`Cells were either pre-treated with inhibitor in HBSS for 30 min (experimental), or left for
`30min in HBSS alone (control). The incubation medium was then changed for one
`containing [3H]colchicine (18 nM, 1.38 mCi) plus inhibitor (experimental) or the same
`chemical and radioactive concentrations of [31-I]colchicine alone (control) and incubated for
`a further 30 min. An increase of [31-I]colchicine accumulation into the cells is interpreted as
`an inhibition of the efflux activity of Pgp. Mean ::s.e.m. Experimental values were
`1
`.
`sig9r(1)i4fi)cantly different from control values by unpaired Student’s t—teSL From Begley et al
`
`Petitioner Mylan Pharmaceuticals Inc. - Exhibit 1031 - Page 9
`Petitioner Mylan Pharmaceuticals Inc. — Exhibit 1031 — Page 9
`
`

`
`TARGETING PEPTIDES AND DRUGS TO THE CNS
`
`145
`
`The most widely used technique is that of osmotic open-
`ing where a short
`infusion of a hyperosmotic solution,
`usually mannitol,
`is
`introduced into a carotid artery.
`Large molecules can be given access to the CNS by this
`technique and its therefore thought that the osmotic agent is
`affecting the integrity of the tight junctions between the
`cerebral endothelial cells. One explanation that has been
`offered is that the osmotic agent is shrinking the endothelial
`cells and physically opening paracellular pathways. Osmotic
`blood-brain barrier disruption coupled with the infusion of
`chemotherapeutic drugs has been used to treat a number of
`central nervous tumours with marked improvements in
`survival
`time and little apparent ill effect from the BBB
`disruption (Gumerlock & Neuwalt 1992). Osmotic BBB
`disruption can be used to deliver therapeutic substances
`that would otherwise be excluded from the CNS because of
`their size, including the possibility of introducing geneti-
`cally-engineered retroviruses in order to replace defective
`genes.
`The peptide bradykinin and some of its analogues
`increase BBB permeability by apparently permeabilizing
`the tight junctions via B2 receptors (Unterberg et al 1984);
`this general phenomenon is referred to as receptor—mediated
`permeabilization (RMP). Similar permeabilization can be
`achieved with leukotrienes, histamine and 5-hydroxytrypta-
`mine. In in-vitro cultures of monolayers of bovine cerebral
`endothelial cells leucine encephalin, a-adrenergics, arachi—
`donic acid, bradykinin, aluminium, phorbolmyristate esters,
`and or-thrombin all increase permeability of the monolayer,
`whilst angiotensin II, saralasin (an angiotensin analogue), B-
`adrenergics, lowered temperature, and 2-deoxyglucose all
`reduced the permeability of the monolayer (Grieg 1992).
`Some of these factors used to permeabilize the barrier are
`more selective than others. As previously mentioned, the
`BBB exists for the specific purpose of protecting the brain
`from potential toxins and providing a separate and stable
`environment within which the neurones can perform their
`integrative functions. Thus any non-selective opening of the
`barrier will transitorily disturb the brain fluid environment
`and cause possible long term damage.
`
`Conclusions
`
`Clearly then, there is considerable scope for optimizing drug
`delivery to the brain in order to treat a whole range of
`Central nervous diseases from neoplasm to degenerative
`disorders and psychiatric disturbance.
`There still remains a great need for a systematic study of
`molecular structure and properties of drugs in relation to
`their BBB penetration. For substances that enter passively,
`the aim is to extend the plasma half life,
`increase the
`Penetration of the blood brain barrier, retain and enhance
`central activity and reduce reactivity with central nervous
`mechanisms degrading or removing the drug from the CNS.
`In practice it is unlikely that all of these ideals will be
`Satisfied for a single drug, but optimizing some of them
`will probably produce gains in most cases.
`Specifically designing drugs which mimic the substrates
`f°T uptake mechanisms located in the BBB, appears to hold
`great promise, particularly if a range of chemical groups can
`be designed which can be attached to a variety of
`
`compounds rendering them substrates with a high aflinity
`for particular carriers. The two transporters GLUT-1 and
`system-L have the highest capacity and perhaps offer the
`best chance for delivering substances to the brain in quan-
`tity. However it may not be necessary to deliver large
`quantities of some substances to the brain which possess
`very potent central activities and which may influence, for
`example, behavioural activity. In these cases lower-capacity
`transporters or an enhanced passive diffusion might suflice.
`Opening and permeabilizing the barrier are generally non-
`selective and are useful
`in allowing an acute entry of
`substances into the brain where a once-only opening is
`required in order to introduce a genetic vector or potent
`drug. Non-selectively lifting the protective BBB repeatedly
`may carry some risks but these risks might be clinically

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