throbber
Journal of
`Medicinal
`Chemistry
`
`© Copyright 2004 by the American Chemical Society
`
`Volume 47, Number 10
`
`May 6, 2004
`
`Perspective
`
`Lessons Learned from Marketed and Investigational Prodrugs
`
`Peter Ettmayer.*~l Gordon L. Amidon} Bernd Clement,§ and Bernard Testaii
`Novartis Institute for BioMedicai Research. Brunnerstrasse 5.9, A-I235 Vienna. Austria. College of Pharmacy,
`The Univeisity ofiviichigan, Ann Arbor, Michigan 48109-1065, Piiarmaceuticai Institute, Univeisity ofKie1,
`19-241 18 Kiel. Germany. and Department ofPharmacy. University Hospital’ Centre (CHUV). CI-1-1011 Lausanne. Switzerland
`
`Received August 7. 2003
`
`1. Introduction
`
`to overcome
`Prodrugs are an established concept
`barriers to a drug’s usefulness. In Germany. about 6.9%
`of all marketed medicines can be classified as prodrugs,
`an estimate based on a conservative prodrug definition
`that does not include soft drugs and limited prodrugs.
`The limited prodrugs. which comprise an estimated 4%
`of the medicines marketed in Germany, are defined as
`active agents whose metabolite(s) also contribute(s) to
`the observed therapeutic activity. Approximately 49%
`of all marketed prodrugs are activated by hydrolysis,
`and 23% are bioprecursors (i.e., lacking a promoiety)
`activated by a biosynthetic reaction.‘ Noteworthy block-
`buster prodrugs are, for example, omeprazole, simvas-
`tatin, lovastatin, enalapril. and aciclovir (Figure 1).
`Regulatory guidelines pay limited attention to issues
`specific to prodrugs?’ Thus, active drugs developed from
`prodrugs are considered as active metabolites. In me-
`dicinal chemistry, a prodrug strategy is practically never
`considered in the early phases of drug design but only
`when classical analoguing programs fail to provide the
`required drug profile. What.
`then. makes prodrug
`development so special and interesting? Prodrugs pro-
`vide a rationale and opportunities to reach target
`physicochemical, pharmacokinetic. and pharmaco-
`dynamic properties. They can be designed to overcome
`pharmaceutical, pharmacokinetic, or pharmacodynamic
`
`* To whom correspondence should be addressed. Phone: +43-1-
`86634-378.
`Fax:
`+43-1-86634-383.
`E-mail:
`peter.ettmayer@
`pha.rma.novartis.com.
`‘ Novartis Institute for BioMedical Research.
`‘The University of Michigan.
`3‘ University of Kiel.
`" University Hospital Centre.
`
`
`
`iovastatin
`
`enaiapril
`
`Figure 1. Some blockbuster prodrugs.
`
`barriers such as insufficient chemical stability, poor
`solubility, unacceptable taste or odor. irritation or pain,
`insufficient oral absorption,
`inadequate blood—brain
`barrier permeability. marked presystemic metabolism.
`and toxicity.3 A developing field of high importance is
`that of rationally designed prodrugs for tissue or cell
`targeting. However. it is worth recalling that many
`successful prodrugs in current use are in fact accidental
`prodrugs, namely, agents that were not designed as
`prodrugs and were recognized as such only late in
`development or even postmarketing. This article intends
`to provide some arguments and guidelines for the early
`
`© 2004 American Chemical Society
`lO.l02lfjm03038l2 CCC: $27.50
`Published on Web 04/13/2004
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2050 - 0001
`
`

`
`2394 Journal‘ of Medicinal Chemistry, .2004, V01. 47. No. 10
`
`Perspective
`
`recourse to, and successful application of, a prodrug
`strategy in drug discovery.
`
`2. Definitions and Classification of Prodrugs
`
`The term prodrug was first introduced in 1958 by
`Adrien Albert‘ to describe compounds that undergo
`biotransformation prior to eliciting their pharmacologi-
`cal eifects. According to his definition (which we consider
`the best available), prodrugs are “therapeutic agents
`which are inactive per se but are transformed into one
`or more active metabolites." A later definition by Bund-
`gaard5 states that "By attachment of a pro-moiety to
`the active moiety, a prodrug is formed which is designed
`to overcome the barrier that hinders the optimal use of
`the active principle." Such a definition. however,
`is
`restricted to prodrugs carrying a promoiety. This defini-
`tion is somewhat narrow because it excludes biopre-
`cursors, modular prodrugs (i.e., comprising a trigger, a
`linker, and the active agent), and metabolites produced
`by conjugation reactions.
`Albert's broader definition posits prodrugs as the
`opposite of soft drugs. The soft drugs (see section 3.3)
`are active analogues of a lead compound that are
`deactivated in a predictable and controllable way after
`having achieved their therapeutic effects But since both
`prodrugs and soft drugs find applications in local tissue
`targeting, they are sometimes confused in the literature.
`Prodrugs can be classified according to two major
`criteria, namely, (a) chemical classes (carrier—linked pro-
`drugs, bioprecursors (i.e., prodrugs lacking a promoiety),
`site—specific chemical delivery systems, macromolecular
`prodrugs, and drug-antibody conjugates) and (b) mech-
`anism of activation (enzymatic versus nonenzymatic,
`activation by oxidation. reduction or hydrolysis. cata-
`bolic versus anabolic reactions).
`The enzymatic versus nonenzymatic activation is of
`particular interest because both routes have advantages
`and disadvantages. While prodrug activation through
`bioconversion as a time— and tissue—controlled process
`has a clear benefit, inter- and intraspecies variability.
`genetic polymorphism, and the potential for drug—drug
`interactions (see section 3.1.4) pose significant chal-
`lenges for such a prodrug Strategy. When purely chemi-
`cal prodrug activation is chosen (e.g., spontaneous
`chemical cleavage at physiologic pH), interspecies vari-
`ability, genetic polymorphisms, and drug—drug interac~
`tion problems are no concern. However, there might be
`chemical stability issues (insufficient shelf life) and the
`site of prodrug activation is undefined. Interestingly,
`there are only a few reports on prodrugs designed to
`rely exclusively on a nonenzymatic activation principle7
`maybe because it is very difficulty to ascertain the
`absence of enzymatic participation.
`The complexity of this broad multidisciplinary field
`makes it difficult to cover in depth all complementary
`viewpoints. Because some topics have already been
`covered by reviews and monographs,3‘1° this report will
`focus mainly on the inherent therapeutic benefit pro-
`drugs can offer in terms of overcoming pharmaceutical,
`pharmacokinetic, or pharmacodynamic barriers and
`increased patient convenience.
`
`3. Major Objectives of Prodrug Design
`The drugs mentioned in the sections below are meant
`to illustrate various opportunities medicinal chemists
`
`
`
`celecoxib prodrugs
`
`parecoxib sodium
`
`Figure 2. Examples of promoieties for improved aqueous
`solubility.
`
`may find in a prodrug strategy. However, this presenta-
`tion should not be understood as covering all prodrugs
`in therapeutic use or in clinical development.
`3.1. Improved Bioavailability. There are a variety
`of possibilities for a prodrug to improve bioavailability
`following oral dosing. We examine in turn aqueous
`solubility, passive intestinal absorption, targeted active
`absorption, and metabolic switching as well—established
`factors limiting bioavailability.
`3.1.1. Improved Aqueous Solubility. Inadequate
`aqueous solubility is an important factor limiting
`parenteral, percutaneous, and oral bioavailability. In
`such cases, a prodrug strategy may bring great phar-
`maceutical and pharmacokinetic benefit. Charged pro-
`moieties (e.g., esters such as phosphates, hemisucci—
`nates. aminoacyl conjugates. dimethylamino acetates)
`and neutral promoieties (e.g., poly(ethylene glyco1)s,
`PEG) can be used. The latter promoieties, however,
`require PEG of high molecular weight to avoid a rapid
`clearance characteristic of low MW PEG conjugates.
`Improved aqueous solubility for better iv administration
`has been demonstrated for PEG-paclitaxel" (Figure 2).
`but we note here that 2’-PEG esters, but not 7’-PEG
`esters, are hydrolyzed enzymatically. Representative
`water solubilities of PEG-paclitaxel prodrugs are as
`follows:
`2’—PEIG5°°°, 666 mg mL‘1: PEG3°°°°, 200 mg
`mI_.‘1; PEG‘°°°°, 125 mg mL“; compared to 25 ,ug mL"
`for paclitaxel itself.”
`Fosphenytoin is another example showing how paren-
`teral delivery problems associated with a sparingly
`water—soluble drug can be overcome by a prodrug.
`Fosphenytoin” (Figure 2) is a hydrophilic phosphate
`prodrug of the anticonvulsant phenytoin and is hydro-
`lyzed rapidly by phosphatases. Another example can be
`found in the parental administration of COX-2 inhibi-
`tors of the diarylheterocyclic class (e.g., celecoxib” and
`valdecoxib'5). Intravenous treatment of acute pain and
`inflammation is hampered by their modest aqueous
`solubility (<50 yg mL‘1). A solution to the problem was
`afforded by the sodium salt of the acetylated sulfon-
`amide prodrug (parecoxib sodium”) (Figure 2), which
`exhibits a significantly improved aqueous solubility
`(~15 mg mL"). The acyl residue is rapidly cleaved in
`hepatic and intestinal preparations and in vivo to
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2050 - 0002
`
`

`
`Perspectivs
`
`Journal ofMed1'cina1 Chemistry, 2004, Vol‘. 4 7, No. 10 2395
`
`NH2
`
`H
`
`9 OH no
`OH
`
`H0
`
`HO—fPqLII3—OH
`
`NH2
`pamidronate
`
`OH
`
`NH?
`
`OH
`
`1
`l
`Ii’-OH
`HO-fl‘
`0 OHO
`alendronate
`
`.‘ N
`
`S H
`
`_ iHg N
`
`0
`
`O
`
`bacampicillin
`
`
`
`OH
`
`O
`
`HO
`
`O
`
`HO HN
`.
`OA\HNYNH
`NH,
`Zal'lal'l'llVll'
`
`benazepril
`
`fosinopril 0
`
`Figure 3. Examples of ester prodrugs showing improved
`passive intestinal absorption.
`
`liberate the active drug. Parecoxib sodium is currently
`in the preregistration phase for parental administration.
`Improved oral bioavailability and dose linearity in
`pharmacokinetics have also been reported for acylated
`celecoxib prodrugs” whose sodium salt showed im-
`proved oral bioavailability with dose linearity over a
`wider dose range compared to celecoxib itself.
`One of the potential problems in this approach is that
`solubilizing groups may sometimes generate toxic ef-
`fects. Thus, the phosphate prodrug fosphenytoin was
`designed to enhance the aqueous solubility of phenytoin
`for intravenous administration, but the high phosphate
`concentrations at the site of injection produced some
`hypocalcemjc effects leading to mild pruritus and par-
`esthesia. 17 Interestingly, such side effects were moder-
`ate compared to those of the standard injectable sodium
`phenytoin.
`3.1.2. Improved Passive Intestinal Absorption.
`Providing enhanced lipophilicity for increased passive
`intestinal absorption is the most frequent rationale
`when adopting a prodrug strategy (approximately 49%
`of all marked prodrugs are activated by hydrolysisl).
`Various esters of carboxylic acids (a frequently encoun-
`tered group of carrier-linked prodrugs) are cleaved by
`hydrolysis (enzymatic and/or chemical) to liberate the
`active carboxylic acid. There are many examples such
`as ACE inhibitors” benazepril and fosinopril (Figure
`3). statins such as simvastatin and lovastatin (Figure
`1). and some antibiotics such as pivampicillin and
`bacampicillin (Figure 3). While this article was in
`preparation. Kevin Beaumont et al.3 comprehensively
`reviewed the design of ester prodrugs to enhance oral
`absorption. We refer the reader to this in«depth analysis
`of ester prodrugs and the related challenges for the
`discovery scientists.
`A nice example of the competitive advantage provided
`by a prodrug strategy is found in the class of neuram-
`
`adefovir dipivoxil
`
`Figure 4. Examples of drugs incorporating phosphonate
`moieties and prodrugs thereof.
`
`AZT
`6
`O:g_ O
`1
`-~/ENH
`Cl)
`0
`
`AZT
`
`carboxylesterase
`—-' ‘
`
`AZT
`I
`3:3
`Q'l,°‘0
`NH
`..
`HO/£0
`
`SN?
`—p-
`
`‘(R-0
`05?
`HN
`
`O
`
`H 0
`2
`'
`
`,0-AZT
`°~‘p
`NI ‘0
`H
`
`h
`h
`o~AzT
`P “P °‘
`0,?-
`dieslerase
`‘OH
`*" -
`on
`
`HO
`
`0
`
`Figure 5. Proposed conversion of AZT-phosphoramidate to
`AZT-monophosphate.
`
`inidase inhibitors used against type A and type B
`influenza viruses. The prototypal drug is zanamivir
`(Figure 3), which shows that a very high polarity is
`incompatible with intestinal absorption and has to be
`administered via inhaler. Another neuramidase inhibi-
`tor is RO-64-0802, which like zanamivir is not absorbed
`orally because of its high hydrophilicity. However, RO+
`64-0802 is not marketed as such but as the ethyl ester
`known as oseltamivir (Figure 3). This prodrug is well
`absorbed orally, and its rapid in vivo enzymatic hy-
`drolysis provides high and sustained plasma levels of
`the active parent drug.”
`A comparable situation is found in the field of his-
`phosphonates, where drugs with improved oral bio-
`availability are badly needed.” Indeed, pamidronate,
`alendronate (Figure 4), and analogues have a poor oral
`bioavailability (<1%).“ While the methyl and ethyl
`phosphate esters are chemically and enzymatically too
`stable,
`the pivaloyloxymethyl and .S'—acetylthioethyl—
`phosphonate esters are converted enzymatically to the
`free phosphonate.” Similarly. adefovir dipivoxil. a his-
`(pivaloyloxymethyl) ester of the antiviral adenine nucleo-
`tide analogue adefovir,” was recently launched in the
`U.S. for the treatment of hepatitis B virus infection
`(Figure 4). Other conceptually successful phosphate
`prodrugs are the aryl phosphoramidates, which deliver
`the nucleoside monophosphates to cells. This has al-
`lowed the efficacy of AZT to be increased by several
`orders of magnitude (Figure 5).“ Interestingly,
`this
`promising prodrug concept has not yet been applied to
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2050 - 0003
`
`

`
`2396 Journal‘ of Medicinal Chemistry, .2004, V01. 47. No. 10
`
`Perspective
`
`
`
`O
`
`0
`
`.
`.
`O ssbraflban
`
`ximelagalran
`
`Figure B. Amidoxime prodrugs on the market or in develop-
`ment.
`
`
`
`desmopressin pivaloyl ester
`
`Figure 7. Examples of prodrugs of peptidomimetics.
`
`phosphotyrosines. It should be mentioned in this context
`that one should be cautious with prodrugs of highly
`charged drugs, since their intracellular release might
`lead to entrapment and thus to toxicity.25
`Besides carrier—linked prodrugs,
`there exist other
`approaches to reach optimal lipophilicity. Amidoximes
`can be used as bioprecursors for amidines (Figure 6).
`Amidoximes are less basic and thus unprotonated under
`physiological conditions, thereby enhancing intestinal
`absorption.” Reductases in the kidneys, liver, brain,
`lungs, and gastrointestinal tract are responsible for the
`rapid conversion of the inactive amidoximes to amidines.
`A GP IIb/IIIa-receptor—antagonist (sibrafiban) and a
`thrombin inhibitor ximelagatran (the "double” prodrug
`of melagatran) are based on the amidoxime strategy and
`are in clinical development. Ximelagatran is expected
`to become the first orally acting, direct thrombin inhibi-
`tor in medical use.“
`
`Increasing evidence is being published on prodrugs
`of peptides, a class of pharmacologically important
`compounds that almost always suffer from mucosa]
`permeation problems and rapid enzymatic degrada-
`tion.3-33 The same is true for some peptidomimetics.
`Thus, the pivaloyl ester of desmopressin (Figure 7), a
`synthetic analogue of vasopressin, showed improved
`transport across Caco—2 monolayers.” The perbutan—
`oylated prodrug glycovir showed improved bioavailabil-
`ity in rat, dog, and monkey compared to the parent
`compound SC-48334. Transient cyclization of linear
`peptides using an esterase-sensitive linker yielded pep-
`tide prodrugs with improved Caco—2 permeability and
`increased stability toward peptidases. The 0-acetyl
`
`H
`
`NH2
`E
`
`H NH,
`”°
`
`0
`
`‘—\N
`(\
`N
`
`N\ NH2
`| Y
`NH
`
`0
`
`‘W
`N
`<\
`N
`
`I
`
`NYNH2
`NH
`
`valaciclovir
`
`O
`
`valganciclovir
`
`O
`
`
`
`H
`
`“
`
`i
`HN
`/
`O5)
`
`zidovudine
`
`H
`0
`H
`
`0
`H2N;\¢J
`
`I
`
`zaicitabine
`
`Figure 8. Example of (pro)drugS utilizing oligopeptide and
`nucleoside transporters.
`
`prodrug of the E-IIV—protease inhibitor saquinavir showed
`improved oral bioavailability (Figure 7).“
`Designing carrier—linked prodrugs, one should keep
`in mind to balance the increased lipophilicity necessary
`for transcellular absorption with sufficient aqueous
`solubility. otherwise oral bioavailability will become
`dissolution limited. Such cases have been reported for
`the bifunctional prodrugs of cephalosporines“ and for
`the pivaloyloxymethyl ester of ceftizoxime.” In addition
`the toxicity potential of the promoiety should be evalu-
`ated or promoieties used that are already accepted by
`registration authorities and are known to be nontoxic.
`Recently the commonly used pivalic acid (trimethyl-
`acetic acid) was associated with some toxicity.33 Also,
`the formaldehyde released upon hydrolysis of methyl-
`ene—bridged double ester prodrugs can be a toxicological
`concern. The toxicologically more acceptable acetalde-
`hyde, on the other hand, introduces a chiral center that
`increases complexity, since the two enantiomers might
`have distinct activation profiles.
`3.1.3. Improved Transporter-Mediated Intestinal
`Absorption. Utilizing carrier-mediated transport is
`another successful prodrug strategy to actively enhance
`intestinal absorption.“ For example, oligopeptide trans-
`porters are responsible for the active uptake of ,6-lactam
`antibiotics, the ACE inhibitor enalapril35 (Figure 1),
`valaciclovir, and valganciclovir, to name a few (Figure
`8). Valaciclovir, the valine ester of aciclovir, showed an
`oral bioavailability 5-fold higher than that of aciclovir
`itself.“ The increased oral absorption is due to transport
`by the intestinal dipeptide transporter PEPTl.37 Nu-
`cleoside transporters are involved in the uptake of
`nucleoside analogues such as zidovudine. zalcitabine
`(Figure 8), cladribine, ara—C, ara-A, and fludarabine.
`Bile acid transporters are used for the uptake of
`thyroxine, chlorambucil, and crilvastatin. Further classes
`of transporters include vitamin transporters (whose
`substrates include methotrexate, nicotinic acid,
`thi-
`amine, and vitamin B12), organic cation transporters
`(e.g., choline, spermine, (+)-tubocurarine, and dopam-
`ine), organic anion transporters (e.g., methotrexate,
`cefodizime, ceftriaxone. and pravastatin), and glucose
`transporters (e.g., glucopyranosides and galactopyran-
`osides). Successful targeting of active transport depends
`on the specificity and capacity of the transporters.
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2050 - 0004
`
`

`
`Perspective
`
`Journal ofMeo'1'ci'na1 Chemistry, 2004, Vol‘. 4 7, No. 10 2397
`
` HHNN
`
`Ievormeloxifene
`
`docarpamina
`
`Figure 9. Example of prodrugs of metabolically labile drugs.
`
`Furthermore. one has to be aware of potential drug-
`drug interactions due to saturationfinhibition of intes-
`tinal transporters.
`3.1.4. Protection against Fast Metabolism (Slow-
`Release Prodrugs). The modification of metabolic
`pathways (metabolic switching) to alter the biotrans-
`formation pattern and protect against fast metabolic
`breakdown is another validated concept in prodrug
`design. While drugs with decreased metabolic stability
`(soft drugs) will be considered in section 3.3. we focus
`here on prodrugs offering increased metabolic or chemi-
`cal stability and hence intensified and prolonged activ-
`ity. In other words. the prodrug here is a protected form
`of the drug. Cases where the prodrug protects the drug
`against fast metabolism are presented first. At the end
`of the section, we discuss prodrugs that release a highly
`reactive and unstable active agent.
`Metabolically labile but important pharmacophoric
`elements can be masked or capped to avoid rapid
`metabolism. In bambuterol (Figure 9). a prodrug of
`terbutaline, the phenolic groups are masked as carbam-
`ates.“ This designed,
`long-acting bronchodilator is
`hydrolyzed by nonspecific cholinesterases. Its competi-
`tive advantage is demonstrated by once-daily dosage
`compared to 3 times daily for terbutaline.
`A wealth of other examples can be briefly mentioned.
`Thus, docarpamine (Figure 9) is a cardiotonic, orally
`bioavailable double prodrug of dopamine whose bis-
`ethylcarbonate groups are cleaved in the intestine.
`followed by amide hydrolysis and conjugation in the
`liver.“ Yet another example is levormeloxifene (Figure
`9). an 0-methylated prodrug of a selective estrogen
`receptor modulator (SERM).4” Oxidative demethylation
`in vivo leads to enhanced oral bioavailability of the
`parent drug. Estrogen deficiency is also the target of
`SCH-57050, a bis-pivaloyl derivative of a 4-hydroxy—
`phenylchromenfl-ol compound. Despite such success
`stories, one should not forget that in oxidative activation
`catalyzed by the cytochrome P450 (CYP) enzymes,
`special attention should be paid to potential drugadrug
`interactions. Concomitantly administered drugs utiliz-
`ing the same CYPs for metabolism can produce drug-
`drug interactions potentially causing unacceptable sys-
`temic exposure and hence toxicity.
`
`
`
`mitomycin
`
`O
`
`Nm
`
`c;o0cH3
`
`QOOCH3
`
`5
`
`CI
`
`5
`
`Cl
`
`c|0pidogre|
`
`2-Oxo-clopidogrel
`
`l
`
`QOOH
`
`woe
`
`CI
`
`5
`
`l
`
`QOOCH3
`
`HS
`
`CI
`
`Figure 10. Prodrugs activated to a highly unstable active
`agent. The antiaggregating agent clopidogrel undergoes ex-
`tensive hydrolysis in humans (ca. 85% of a dose) to the inactive
`acid. A smaller proportion of the dose is activated by cyto-
`chrome P450 3A to 2-oxoclopidogrel, which irreversibly an-
`tagonizes platelet ADP receptors via a covalent S-S bridge.“
`
`Groups of particular interest are prodrugs activated
`to a highly unstable active agent. Thus. a number of
`antitumor drugs are in fact reduced within tumor cells
`to a reactive metabolite. This is exemplified by mito-
`mycin, which is reduced by NADPH-cytochrome c re-
`ductase to provide the active semiquinone, which would
`not be stable enough to be effective in the cerebrospinal
`fluid (Figure 10).“ While NADPH-cytochrome creduc-
`tase is not specific for tumor cells, its activity is favored
`in such cells because of their higher reductive capacity.
`A comparable situation exists for the polyol nitrates
`marketed as coronary vasodilators (e.g., glycerol tri-
`nitrate. isosorbide dinitrate, isosorbide mononitrate),
`which can be seen as clinical release forms of nitric
`oxide.
`
`A recent and quite revealing example is that of
`clopidogrel. whose mechanism of activation and action
`has been uncovered only recently‘? (Figure 10). Clo-
`pidogrel is an antiaggregating medicine that under-
`goes extensive hydrolysis in humans (ca. 85% of a dose)
`to the inactive acid. A smaller proportion of the dose is
`activated by cytochromes P450 3A to 2-oxoclopidogrel,
`which spontaneously and rapidly hydrolyzes to a highly
`unstable thiol.43 The latter is the active agent. whose
`mechanism of action involves irreversible binding to
`platelet ADP receptors via a covalent S-S bridge.
`3.2. Tissue-Selective Delivery. Selective delivery
`to the target cells or tissues is known to the public as
`the "magic bullet” metaphor.“ While some drugs have
`a built—in capacity to accumulate in target tissues or
`cells. such a favorable pharmacokinetic feature is usu-
`ally the objective of a prodrug strategy. And indeed,
`rationally designed tissue targeting is probably the most
`exciting objective of a prodrug strategy. This can be
`achieved by (a) passive enrichment in the target tissue,
`(b) targeting specific transporters. (c) targeting tissue-
`or cell-specific enzymes. and (d) targeting surface an-
`tigens. These strategies are presented and exemplified
`in turn below.
`
`Patent Owner. UCB Pharma GmbH — Exhibit 2050 - 0005
`
`

`
`2398 Journal‘ of Medicinal Chemistry, .2004, V01. 47. No. 10
`
`Perspective
`
`d
`
`(Iii
`H
`N
`
`s
`
`.5
`
`H*
`N
`.4
`r 4?
`
`omeprazole
`
`/0
`
`I
`0
`
`n —
`@~’.+~w \
`'
`
`O
`
`H §
`O
`
`
`HO OH
`
`NH
`
`levodopa
`
`dopamine
`
`Figure 11. Example of prodrugs targeting the brain.
`
`3.2.1. Passive Enrichment in the Target Tissue.
`Cytostatic and cytotoxic agents conjugated to PEG with
`MW > 50 000 (e.g., PEG—paclitaxel, Figure 2) are
`reported to exhibit an improved therapeutic efficacy due
`to their longer half-lives and selective accumulation in
`tumor cells. This passive tumor targeting is called the
`"enhanced permeability and retention” (“EPR") effect.
`and its mechanism is not fully understood.“ Clinical
`phase II trials are ongoing using a PEG—Ala-campto-
`thecin conjugate (prothecanw) for the treatment of
`stomach tumors. The low drug load achievable by such
`macromolecular conjugates requires more potently cy-
`totoxic agents than traditional anticancer drugs.
`An interesting prodrug strategy for brain targeting
`is that of site~specif1c chemical delivery systems (CDS).’”
`After brain penetration, a lipophilic prodrug is converted
`there into a more hydrophilic molecule that remains
`"locked in". The same conversion taking place in the rest
`of the body results in increased peripheral elimination.
`The most frequently reported CDS is the 1,4—dihydro-
`trigonelline~trigonelline targetor system attached to
`the drug via an esterase-sensitive bond (Figure 11).“
`1.4-Dihydrotrigonelline is rapidly oxidized by oxidoreduc-
`tases, resulting in the formation of the inactive quater-
`nary ammonium metabolite in the periphery and the
`brain. When formed in the periphery, this polar me-
`tabolite is rapidly excreted. In contrast, its formation
`in the brain leads to entrapment followed by enzymatic
`hydrolysis. This liberates the drug in situ, whereas the
`polar quaternary pyridinium ion moiety is rapidly
`eliminated from the brain, probably by a transporter.
`Dexamethasone—CDS and estradiol—CDS entered clinical
`
`development (phase I/II), but trials were stopped in
`2001.
`
`Prodrug activation in osteoclasts and osteoblasts can
`be utilized for bone targeting. The osteotropic drug
`delivery system (ODDS) relies on the bone-homing
`properties of bisphosphonates for the targeted delivery
`of drugs to the bone and bone marrow.“ A few examples
`of ODDS delivery systems of estrogen and diclofenac are
`in exploratory stages.
`3.2.2. Targeting Specific Transporters. The num-
`ber of well-documented examples of prodrugs targeting
`tissue-specific transporters is relatively limited. Levodopa
`affords an apt example of enrichment resulting from
`active import of the prodrug into the brain followed by
`tissue entrapment of the active metabolite dopamine
`
`‘o@J»~~:« we :1
`
`...
`
`N_
`
`S
`
`I
`O 4 r__
`
`NH
`
`sulfenamidez Active specices
`
`sulfenic acid
`
`S‘oH
`
`Figure 12. Activation of omeprazole.
`
`
`
`Amtolrneiin guacil: prodrug of lolmelin
`
`Figure 13. Examples of prodrugs for colon targeting or to
`avoid gastrointestinal side effects.
`
`(Figure 11).49 Levodopa is a substrate for the neutral
`amino acid transporters present at the blood—brain
`barrier (BBB). After brain entry, levodopa is decarbox—
`ylated to dopamine, which can act locally, being no
`longer a substrate for the neutral amino acid trans-
`porter. To increase the systemic half—life of levodopa, it
`has become customary to coadminister peripherally
`acting inhibitors of DOPA decarboxylase.
`Prodrugs that target the liver utilize transporters
`predominately expressed in this organ. There are pub-
`lications on bile acid conjugates of various cytostatic
`compounds, but no clinical reports appear to have been
`published.5°
`3.2.3. Targeting Tissue- or Cell—Specific En-
`zymes. The acidic environment of the stomach is
`utilized by the proton pump inhibitor omeprazole (Fig-
`ure 12).51~52 This potent antiulcer agent is a prodrug of
`a sulfenamide that exerts its effects by covalently
`modifying cysteine residues on the luminal side of the
`proton pump. i.e., the H+fK+—ATPase of the parietal cell
`in the oxyntic mucosa of the stomach. The prodrug
`omeprazole is only activated in the acidic enviroment
`of the oxyntic mucosa of the stomach where the drug
`exercises its antisecretory effect.
`The reductive environment or the bacterial flora can
`
`be utilized for colon targeting. Balsalzide, a prodrug of
`5-aminosalicylic acid (5-ASA) and an analogue of sulfa»
`salazine. is specifically converted to 5-aminosalicilic acid
`by azo-reducing bacteria present in the colon (Figure
`l3).53 The prodrug remains intact in the gastrointestinal
`tract until it reaches the colon. where it releases 5-ASA
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2050 - 0006
`
`

`
`Perspective
`
`capacitabine H0-/_/_
`
`Journal ofMed1'cina1 Chemistry, 2004, V01. 4 7, No. 10 2399
`
`\
`
`N—§
`F
`O
`N
`/
`N
`w 0
`
`OH
`
`'
`
`carboxyiesterasa
`——u
`Liver
`
`HO
`
`0
`
`F
`cyiidin-
`f/'S,NH2 d68mi|"859
`
`I
`N
`N
`71’
`liver 5.
`
`tumor
`
`OH O
`
`be
`
`O
`
`thymidine
`phosphoryiase
`
`
`,.—
`O
`‘i_7’”7r”“
`,
`,
`tumor
`HO
`OH O
`Figure 14. Activation of capecitabine.
`
`HN
`
`F
`
`/
`

`
`NH
`
`237/
`
`-
`
`to exert a local anti—inflammatory effect directly on the
`inflamed lining of the intestinal wall.
`Alternatively to colon targeting, many prodrug projects
`are directed toward avoiding gastrointestinal side ef-
`fects. A nice example of prodrug design is amtolmetin
`guacil, a lipophilic prodrug of the anti—inflammatory
`drug tolmetin created to reduce gastrointestinal side
`effects. In addition to beneficial lipophilicity,
`its hy-
`drolysis in the gastrointestinal (GI) tract releases gua-
`jacol, which in turn activates inducible gastric nitric
`oxide synthase,54 thereby reducing GI irritation (Figure
`13). It is interesting in this context to mention that the
`extensively investigated esters of nonselective COX
`inhibitors did not show the desired GI protection, since
`GI side effects are a consequence of COX-1 inhibition
`and not of local irritation.
`
`Specific targeting of virus-infected cell is elegantly
`realized by the antiviral prodrug aciclovir (Figure 1) and
`all other selective antimetabolites (a subgroup of bio-
`precursor prodrugs) used as antiviral agents. The activ-
`ity of such antimetabolites implies their intracellular
`phosphorylation by kinases to form a nucleotide ana-
`logue that inhibits DNA synthesis. The older nonselec-
`tive antiviral drugs were phosphorylated by native and
`by virus-induced thymidine kinases. In contrast, the
`selective antimetabolites are phosphorylated only by
`kinases coded by the viral genome and expressed in
`infected cells. As a result. aciclovir and all other
`selective antimetabolites are active only in infected
`cells.55
`
`The selective delivery of cytotoxic drugs to tumor cells,
`without concomitant damage to normal tissues. is a
`major challenge in cancer chemotherapy. Prodrugs for
`active tumor targeting are therefore of high interest.
`Tumor specificity can be achieved by targeting trans-
`porter systems, enzymes having a higher activity in
`tumor cells, as discussed here. or surface markers (see
`section 3.2.4) or by gene-directed enzyme prodrug
`strategy (see section 3.2.5).
`Capecitabine, an orally available triple prodrug of
`5—fluorouracil (5—FU), offers a good example for a pro-
`drug activated by tumor-specific enzymes (Figure 14).
`Following oral absorption, capecitabine undergoes three
`activation steps resulting in high tumor concentra-
`tions of the active drug, namely, (a) carboxylesterase
`mediated hydrolysis in the liver. (b) cytidine deaminase
`mediated deamination in the liver and tumor cells, and
`(c) specific liberation of 5—FU by thymidine phosphoryl-
`ase in tumor cells.“
`
` mAb-NH T
`
`Lysine attachment
`
`Figure 15. N-Acetyl-y-calicheamicin linked via a hydrolyti-
`cally labile hydrazone linker to a lysin N -atom of recombinant
`humanized anti-CD33 antibody.
`
`3.2.4. Targeting Surface Antigens. Targeting sur-
`face markers appears as a selective and promising strat-
`egy in developing tumor-selective prodrugs. In 2000, the
`FDA approved the use of CMA—676 {gemtuzumab ozo-
`gamicin, mylotarg) for the treatment of acute myelotic
`leukemia (AML).57 CMA-676 is an antibody-targeted
`chemotherapy agent consisting of a recombinant hu-
`manized anti-CD33 antibody linked via the lysin N-
`atom to N—acetyl—y-calicheamicin, a potent cytotoxic
`agent {Figure 15). The linker incorporates a hydrazone
`moiety, a site for hydrolytic release.” previously shown
`to be required for activity.“ Since this antibody recog-
`nizes AML blast cells but not hematopoietic stem cells,
`it allows selective delivery of the cytotoxic agent to
`leukemia cells. It was demonstrated that after CMA—
`
`676 infusion, the CD33 sites are quickly saturated and
`internalization of the conjugate occurs. However, elimi-
`nation of leukemia correlates with the low capacity of
`these cells to extrude the drug conjugate. This finding
`is in good agreement with the fact that CMA-676 was
`ineffective in multidrug-resistant (MDR) sublines ex-
`pressing P-glycoprotein. However, by combination of
`CMA-676 and MDR modifiers. an increased effect in
`MDR AML may be possible. While there are numerous
`preclinical reports on such carrier—linked prodrugs,
`successful clinical developments are slow to come.
`3.2.5. Enzyme—Prodrug Cancer Therapy. Selec-
`tive activation of prodrugs in tumor tissues can also be
`achieved by exogenous enzymes in a two—step approach.
`Prodrug-activating enzyme gene or functional protein
`is delivered selectively to tumor tissues, followed by
`systemic administration of a nontoxic prodrug that is
`activated by the exogenous enzyme. The net gain is a
`high local concentration of an active anticancer drug in
`tumors. Delivery of the enzyme gene is accomplished
`via viral vectors (VDEPT, virus—directed enzyme pro-
`drug therapy) and

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