throbber

`
`ELSEVIER
`
`advanced
`drug delivery
`reviews
`Advanced Drug Delivery Reviews. 19 (1996) 287—310
`
`
`
`Review
`
`Prodrugs of phosphates, phosphonates, and phosphinates
`
`Jeffrey P. Krise, Valentino J. Stella*
`Department of Pharmaceutical Chemistry and the Center for Drug Delivery Research. The University of Kansas.
`2095 Constant Avenue. Lawrence. KS 6604?. USA
`
`Received 1 August 1995: accepted I October 1995
`
`
`
`Abstract
`
`The objective of this paper is to review the literature on the use of prodrugs to overcome the drug delivery
`obstacles associated with phosphate. phosphonate and phosphinate functional group-containing drugs. This is an
`important area of research because. although we have been successful at identifying numerous phosphate and
`phosphonate functional groupcontaining drugs as antiviral and anticancer agents, as well as for other uses. our
`ability to orally deliver these drugs and to target
`them to desired sites has led to limited success. Various
`acyloxyrnethyl— and aryl-ester prodrugs have shown promise. Alternative and imaginative approaches may be
`necessary before complete success is realized. It
`is our—hope that this review will stimulate further innovative
`prodrug research into overcoming the barriers to the delivery of these important drugs.
`
`Keywords: Nucleotide analog; Nucleotide prodrug; Bioavailability; Ester; Permeability
`
`
`Contents
`
`288
`2. Why prodrugs ofphosphates. phosphonales and phosphinates'?
`288
`3. How can prodrugs overcome these problems?
`
`290
`4. Specificcxamples“
`290
`4]. Nucleotide analog prCidi'Lig3"
`290
`4.1 I. Simple and substituted alkylaiidaryimester prodrugsof phosphates and phosphonates
`4.1.2. Acyloxyalkylcstcrs 295
`4.1.3. Phospholipid derivatives.
`29?
`
`4.1.4. (.‘yclic prodrugs..................
`298
`4.1.5. Nucleotide analog conjugate systems.
`299
`
`4.1.6. Carbohydratederivatives 301
`4.1.3". ‘SATE‘ andDTE’
`30]
`
`4.2. Miscellaneous prodrugapplications... 301
`
`4..2.l Fosiuopn'l: An ACE inhibitor...
`....
`301
`4.2.2. CBS 24562: A neutral cndopeptidase inhibitor
`302
`4.23 L690330: A inhibitor of Inosnol monophosphatase:
`303
`.
`......
`
`4.2.4. (Hydroxy—2—naphthalenylmethyl)phosphonic acid. an lRiK InthItor
`..............
`304
`
`304
`5. Conclusions.
`
`References 305
`
`*Corresponding author. Fax: + l 913 8425612.
`
`© 1996 Elsevier Science B.V. All rights reserved
`0169-409X!96i$32.00
`SSDI 0169-409X{95)00111—S
`
`1
`
`G|L2019
`I-MAK, INC. V GILEAD PHARMASSET LLC
`|PR201 8-00123
`
`1
`
`GIL2019
`I-MAK, INC. V GILEAD PHARMASSET LLC
`IPR2018-00123
`
`

`

`288
`
`JP. Kris-e. VJ. Stella l Advanced Drug Delivery Reviews l9 {I996} 287—310
`
`0
`RO-—i=|’—-OR'
`([)R'
`Phosphate
`
`0
`R——i=|‘—OR'
`(in
`t’hosphmmic
`
`fi
`R—tfuR
`0‘"
`Phosphinutc
`
`Drug:
`
`R = organic residue: R' = H or [-3
`
`Prodrug;
`
`R 2 organic residue: R' = promoeity
`
`I. General structure of phosphate. phosphonate. and
`Fig.
`phosphinate drugs. R represents an organic residue while R'
`represents H or anionic charge for the parent drug or the
`promoiety for the prodrug.
`
`1. Introduction
`
`to review the
`The intent of this paper is
`literature on the use of prodrugs to overcome
`drug delivery obstacles associated with phos-
`phate, phosphonate and phosphinate functional
`group containing drugs. The general structures of
`phosphate, phosphonate, and phosphinate-con—
`taining drugs are shown in Fig.
`1 where R
`represents an organic residue, and R’ represents
`either an anion charge or a hydrogen atom in the
`case of the parent compound. or R’ is a neutral
`ester in the case of the prodrug.
`Initial
`research on the problems associated
`with the delivery of phosphate functional group-
`containing drugs began in the early 19605 with
`phosphate—containing nucleoside analogs USed in
`cancer and viral chemotherapy. Effort continued
`to expand and has recently increased in interest
`with the advent of the variety of drug classes
`utilizing the ph0sphate, phosphonate or phosphi-
`nate functionalities, especially antiviral agents
`and various molecules designed to alter cell
`signaling processes.
`
`2. Why prodrugs of phosphates, phosphonates
`and phosphinates?
`
`Although the range of drugs containing either
`a phosphate, phosphonate or phosphinatc—con—
`taining functional has changed,
`the underlying
`problem with the delivery of such drugs has not.
`The shortcomings in the delivery of these drugs
`can be broken down into two basic problems:
`
`and phosphinate
`1. Phosphate, phosphonate
`groups impart an anionic charge (mono- or
`di—) at nearly all physiological pH values
`making them very polar. This high polarity
`can be the basis for many deficiencies in the
`efficacy of drug delivery. Specifically. highly
`ionized species do not readily undergo passive
`diffusion across cellular membranes.
`
`2. BecauSe of the increased polarity, these agents
`often exhibit a low volume of distribution and
`
`therefore tend to be subject to efficient renal
`clearance as well as possibly biliary excretion.
`In addition to renal clearance. phosphates.
`particularly those of primary alcohols and
`phenols, are known to be substrates for many
`phosphorylases present
`in the body which
`readily clip the phosphate group from the
`drug. The rapid dephosphorylation results in a
`short duration of action. Phosphonates and
`phosphinates have the advantage of being
`more chemically stable and showing essential—
`ly no enzymatic lability.
`
`3. How can prodrugs overcome these
`problems?
`
`In an attempt to overcome these shortcomings.
`the ionizable phosphate, phosphonate and phos—
`phinate groups have been neutralized via chemi—
`cal derivatization. This generally involves de—
`rivatization of the phosphorus—coupled oxygen(s)
`to form neutral ester(s). If the intention of the
`ester is for it to breakdown in the body to release
`the parent drug, then such a derivative would be
`a prodrug. The neutralization of the charge(s)
`has been proposed to serve a number of pur—
`poses:
`
`l. The first is to decrease the polarity by increas—
`ing the lipophilicity of the drug molecule thus
`allowing access to cells and tissues that might
`not be available to the non-modified specie
`and possibly altering the distributionlelimina-
`tion pattern of the parent drug.
`2. Another use of the neutral ester prodrugs is
`particularly important for phosphates which
`are substrates for nonspecific serum phos-
`phohydrolases, such as alkaline and acid phos—
`
`2
`
`

`

`JJ’. Krise. VJ. Stella t’ Advanced Drug Delivery Reviews 19 (£996) 287——3'IU
`
`289
`
`it has been pro-
`phatases. With these drugs,
`posed that the neutral ester serves to disguise
`the phosphate from the enzymes thereby
`altering the apparent elimination and half-life
`as illustrated in Scheme 1. A flaw in this
`
`thinking is that once deprotected. the intrinsic
`properties of the drug should not be dramati—
`cally altered. Nevertheless, entrapment of the
`parent drug in tissues in which it
`is not
`normally accessible could lead to apparent
`changes in its pharmacokineticipharmacody
`namic properties.
`
`Although alterations in apparent clearance
`rates may be important, the principal goals of
`most prodrug modification efforts on phosphate,
`phosphonate and phosphinate drugs is alteration
`of membrane permeability to improve oral (GI
`permeability), brain, tumor and cellular delivery
`(mainly to virally infected cells) of these agents.
`When these prodrugs are used for improving
`oral bioavailability, various issues dealing with
`G1 absorption of drugs must be considered. The
`ability to address these issues will ultimately
`determine the proper selection of the prodrug
`system and its
`likely success. The optimal
`scenario for enhanced systemic delivery of pro-
`drugs after oral dosing is as follows:
`
`1. The prodrug must display adequate chemical
`stability for formulation purposes as well as
`stability in the variable pH environment of the
`GI tract.
`
`2. The prodrug should have adequate solubility
`in the GI
`tract environment
`to allow for
`dissolution.
`
` 0 Phosphatase - II—-
`
`
`(DO We
`09
`CFUS
`
`CDC“
`mm
`
`+ He—a—m
`.
`0”
`
`Proclrug
`Neutralization
`
`0
`o
`I
`II
`Ph
`so
`—P-—
`_
`C30 . on 92?;- OOH m sea
`0-H
`0-1
`m
`NETAKJLITE
`
`3. Once dissolved, the prodrug should also dis-
`play enzymatic stability to lumenal contents as
`well as the enzymes found in the brush border
`membrane.
`
`4. The prodrug should have properties that allow
`for good permeability (generally associated
`with an adequate log P value).
`5. After permeation of the lumenal membrane,
`the prodrug could revert to the parent drug
`either in the enterocyte or once absorbed into
`systemic circulation. Post—enterocyte reversion
`is desired because conversion in the en-
`
`terocyte would also allow for back diffusion
`into the GI lumen, a problem which is not
`generally recognized.
`
`When the prodrug is formulated to increase
`cellular permeability into viral-infected cells,
`tumor cells or across barriers like the blood brain
`
`barrier, the desired characteristics might change.
`Replacing the desire for complete and rapid post
`absorption reversion.
`is a need for balance in
`lability. The most optimal scenario, however
`unrealistic, would be for the prodrug to have
`complete enzymatic and chemical stability during
`the absorption process and in blood but readily
`revert
`to the parent compound once it has
`permeated the targeted cell,
`thereby ‘trapping’
`the drug in the cell (Scheme 2).
`Considering both of these scenarios, prodrugs
`for
`improved oral delivery and prodrugs for
`improved cell
`targeted delivery.
`the rate of
`bioreversion is a very important process that
`
`SYSTEM”mm
`
`O
`(Do—see
`
`PM
`
`Newngizalmn
`
`l
`
`|
`mediating
`
`r"
`
`\
`
`\
`
`\
`
`TAHGET CEIJ.
`
`0
`'
`
`V -2{”0"}
`
`9
`CEO-e0“
`I
`0'“
`m RI
`
`ii
`Q—ewm
`on
`mo
`
`Scheme 1. Scheme showing the possible advantages of pro-
`drugs in altering the phosphatase cleavage of phosphate—
`containing drugs.
`
`Scheme 2. Proposed scheme illustrates the potential advan—
`tagescf prodrugs over the parent molecule for intracellular
`targeting.
`
`3
`
`

`

`290
`
`.IJ’. Krffl’. VJ. Stella l Advanced Drug Delivery Reviews .1”) (”'96) 287—3.”)
`
`must be considered in detail when designing
`prodrug systems. For example, if bioreversion is
`very fast and non-specific, prodrug reversion may
`take place before the limiting barrier is over—
`come. On the other hand. if reversion is slow and
`
`inefficient at all sites. the prodrug may readily
`reach the site of action but never release enough
`parent drug to elicit a pharmacological response.
`With these factors in mind. choosing a suitable
`hioreversible protective group for phosphates.
`phosphonates and phosphinates presents a major
`challenge.
`
`4. Specific examples
`
`4.1. Nucleotide analog prodrugs
`
`Purine and pyrimidine nucleoside analogs have
`found great utility in treatment of neoplastic and
`viral diseases [1—8]. Please note, the word nu»
`cleoside will be equated with nucleoside analog
`for the remainder of this manuscript for simplici-
`ty purposes. These nucleosidic drugs mostly rely
`on viral or kinase—mediated (i.e.. thymidine ki-
`nase) activation step(s)
`to produce the phos-
`phorylated nucleotide. necessary to display bio-
`logical activity i.e.,
`the nucleosides are them~
`selves prodrugs. Unfortunately. dependency on
`kinase mediated synthesis can lead to the de-
`velopment of resistance [9—11]. The first step in
`the phosphorylation of a nucleoside to the mono—
`phosphate is known to be highly specific and
`often causes the development of resistance [12;
`
`15]. Therefore. it can be argued that an approach
`to circumventing the resistance development
`problem is
`to administer the monophosphate—
`containing nucleoside drug. This strategy has two
`flaws:
`
`l. The highly polar monophosphate has limited
`passive absorption properties and therefore,
`transcellular transport is very restricted [16—
`19]. This flaw is supported by the work of
`Leibman ct al. [18] who demonstrated the lack
`of passive as well as the absence of an active
`transport mechanism for ara-CMP (the mono
`phosphate derivative of ara C; Fig. 2).
`Rapid in vivo dephosphorylation of the mono—
`phosphate is observed with this class of drugs
`[20-122].
`
`go
`
`Could prodrugs be used to overcome these
`problems? The aim of this portion of the review
`is to focus on phosphorus—coupled oxygen pro~
`drug esters of nucleotides, namely the mono—
`phosphates and their phosphonate analogs. An
`effort has been made to group the various
`prodrug strategies according to the type of neu—
`tral ester chosen. The interested reader may wish
`to refer to other reviews on this subject that take
`a little different approach from those taken here
`[23-25].
`
`4.1.]. Simple and substituted alkyi and aryi ester
`pmdrags of phosphates and phosphonates
`Rosowsky et al.
`[26] have examined several
`mono-5’—(alkyl phosphate) esters of ara-C (Fig.
`
`N”;
`
`Nll_.
`
`IlU
`
`N/
`o‘J‘\
`
`N
`
`o
`
`no
`
`on
`
`ara-C
`
`|
`
`N/
`o‘j'\
`
`N
`
`|
`
`o
`
`in.
`
`Rape—o
`OR,
`
`0
`
`u
`
`on
`
`ara—(IMP {RJ = H. R2 = H)
`
`ill R1. R2 =C2H6» H
`{2} R]. R2 = n'Crle‘ H
`[3] RI- R2 2 n-C‘SHU. H
`[4] R.. R2 = n-Can. H
`(5] R.. R2 = n-CmHn‘ H
`[6} RI- R1 = Cszr Csz
`or) R]. R2 =n-C.H-;. D‘CiHst
`(R) R.. R2 : n-Cslln. I'I-Can
`(91 R.. R2 = n—C16H33.n'C|fiHJJ
`
`Fig. 2. Structures of ara-C. ara—(3MP. and araCMP alkyl prodrugs. Structures (1)—(4) are mono-alkyl prodrugs [26] while (5)—(9)
`are dialkyl prodrugs [30].
`
`4
`
`

`

`LP. Krfse, VJ. Stella 1' Advanced Drug Delivery Reviews l9 {I996} 287—310
`
`291
`
`2, 1—5) in an effort to deliver ara—CMP to cancer
`cells. The cytotoxicity of the proposed prodrugs
`toward cultured L1210 leukemia
`and B16
`
`melanoma cells appeared to obey an inverse
`structure—activity relationship with respect
`to
`alkyl chain length. The n-butyl and n—hexyl
`prodrugs were approximately half as active as the
`ethyl prodrug. The structure—activity relationship
`seemed to plateau as chain length became longer
`as exemplified by n-octyl and n-Clfi H33 esters
`having nearly the same lDSO values. Similarly,
`Mullah and coworkers [27] produced a 5'-O
`methyl and 5’—0 phenyl esters of 2’,3’-didehydro-
`2',3'—dideoxyadenosine and 2',3’-didehydro-2',3’~
`dideoxycytosine which displayed similar in vitro
`results to the parent nucleosidcs. When incuw
`bated in serum containing medium, the phenyl
`prodrug produced parent nucleoside along with
`nucleoside monophoSphate; the methyl prodrug
`was not evaluated in this manner. In an in vivo
`
`experiment, assessing activity against adenocar-
`cinoma 755 in mice, Montgomery et al. [28] has
`evaluated the mono- and diethyl. butyl, and
`phenyl
`esters
`of 6-mercaptopurine
`ribonu—
`cleotide. The monoester prodrugs were of the
`same order of effectiveness as (Hnercaptopurinc
`ribonucleotide, whilst the diesters were markedly
`less effective.
`
`In general, the mono-alkylfaryl ester analogs
`of phosphates failed to act as efficient prodrugs
`for the delivery of nucleoside—monophosphate
`analogs intracellularly. The poor activity of the
`monoalkyllaryl esters can be attributed to:
`
`1. High degree of polarity. Due to presence of
`the mono—anionic
`charge,
`limited passive
`transport across cells can be expected.
`2. The relative ease of in vivo conversion of the
`
`monoalkyl esters in vivo back to the parent
`nucleoside before the cell barrier has been
`
`overcome. This could be a possible explana-
`tion for the monoalkyl prodrugs and parent
`nucleoside sharing similar activities.
`
`McGuigan et al. [29] recently synthesized and
`evaluated, in vitro, 3 series of alkyl prodrugs of a
`hydrogen-phosphonate derivative of AZT (Fig.
`3) in an attempt to increase its antiviral activity.
`They showed that the short chain (Cl—C7) alkyl
`
`NH2
`
`n / "‘9
`
`l
`
`J\
`
`o
`
`N
`
`R= Me
`E :E-ll-lcpl
`R 2 "am“
`
`(III)
`RO-Ir—O
`H
`
`0
`
`"3
`
`AZ'I‘ Hephosphonate analog (R = H)
`
`Fig. 3. Structures of an AZT H-phosphonate analog and its
`prodrugs [29].
`
`hydrogen phosphonates to be more active than
`the long-chain (C18) ester prodrugs in HIV-1—
`infected C8166 cells. The short chain phospho-
`nate prodrugs were 5—10 times more potent than
`the parent phosphonate; however, all prodrugs
`were Poorly active in an infected JM cell
`line,
`which is thought
`to lack the kinase mediated
`phosphorylation to produce the active nucleotide
`analog. This demonstrated lack of activity in the
`J M cell line suggested that these prodrugs were
`acting as depot forms of the nucleoside rather
`than the nucleotide.
`
`To cause a further reduction in polarity, inves-
`tigators have also synthesized diesters of phos-
`phatesiphosphonates and evaluated their effec-
`tiveness as prodrugs in in vitro and in in vivo
`tests. The results were not consistent with the
`
`observation with the monoesters. For example,
`Colin et al.
`[30]
`shOWed a clear
`relationship
`between inhibition of thymidine incorporation by
`mammalian epithelial cells and lipophilicity (in-
`creased alkyl chain length) of diester prodrugs of
`ara-CMP utilizing a similar series of protecting
`groups to those studied by Rosowsky et al. (Fig.
`2, 6—9). The in vitro activity was lowest for the
`ethyl ester and highest
`for
`the hexy] ester.
`Similar work performed on ara-AMP gave a
`correlation between an in vitro inhibition of
`
`DNA synthesis and lipophilicity; however, no
`anti—viral activity at concentrations up to 100
`,ugfml was detected against a range of viruses
`[31,32]. AZTMP alkyl esters were also employed
`to improve membrane permeability [33,34].
`In
`vitro.
`the tricsters showed a complete lack of
`
`5
`
`

`

`292
`
`LP. Kris-e. VJ. Stella a“ Advanced Drug Delivery Reviews 1'9 (119%) 287—3“)
`
`inhibition of HIV, which was attributed to the
`
`high stability of the simple alkyl esters with little
`or no conversion to the active 5'—phosphate.
`
`</N
`0
`0
`R 0 III?
`N
`1 _
`R25 \/ \/\0/
`
`NH;
`
`\ N
`N)
`
`9-[2-[Phosphonomedmxykthoxyladeninc (R.. R2 = H}
`
`Fig. 4. Structure of 9-[2-(phosPhonomethonyethoxyladeninc
`(R1. R2 = H). Refer to Table l for representations of R1 and
`R2 [35].
`
`Ta blc l
`
`Serafinowska and co-workers [35] have made a
`series of dialkyl (among others) prodrugs of 9-[2-
`(phosphonomethoxy)ethoxy}adenine
`(Fig.
`4,
`Table 'l)
`in an effort
`to improve the poor
`bioavailability, 7% in rats [36], < 1% in monkeys
`[37].
`After oral administration to mice, the inves-
`
`tigators monitored blood concentrations of the
`diester, monoester, and free acid (1—5, Table 1).
`In all cases,
`the diester was well absorbed:
`
`however, subsequent conversion to the monoes—
`ter and free acid was more dependent upon the
`nature of the alkyl ester. Short chain diesters,
`being chemically (and probably enzymatically)
`
`Concentration of 9—[2—{phosphonomethoxy)ethoxy]adenine 1 (see Fig. 4) and its mono— and dialkyl esters in the blood following
`oral administration of esters to mice (Reprinted with permission from [35] American Chemical Society.)
`Number
`R!
`R2
`Total AUC.‘ 15,180 min (nM)
`
`l
`Monoestcr
`Diester
`Biowailabilit}I
`
`of l (%}“
`
`l
`2
`3
`4
`5
`
`H
`Me
`Et
`i—Pr
`n—Bu
`
`H
`Me
`Et
`i—Pr
`n‘Bu
`
`—
`U
`4
`5
`[J
`
`,
`9
`35
`2‘)
`69
`
`—
`39
`9
`10
`2
`
`2
`0
`8
`It}
`t]
`
`3t]
`0
`U
`[S
`NleJJCO_.CH2
`Me‘CCOZCH,
`6
`o
`o
`u
`n
`Me
`Me‘CCOZCH:
`?
`{l
`t)
`40
`0
`Et
`MefiCOfiH2
`8
`I)
`0
`8
`[1
`i—Pr
`lVle_1(.“.CtC)_,Cl-l2
`9
`[l
`n
`I l
`[l
`n—Bu
`Me‘(jt’leCH2
`It]
`[J
`0
`I}
`I)
`l'vle3C(Cl-l_,CI)CO,C'H2
`Me,(I(CI-I2CI)CO:CH3
`1]
`24
`0
`0
`12
`MeJCHC02CH(Me)
`Mc2CHCOZCH(Me}
`12
`74
`t]
`0
`31'
`MeJCCOICH(Me)
`Mc_.CC02CH(Me)
`13
`4
`t]
`18
`2
`(“hHfiCHJD
`ChflfiCHPO
`I4
`2
`9
`3
`I
`4—BrCuH‘CHEO
`Lurchiiqcup
`:5
`8
`3|
`12
`4
`4—C1C6H4CH30
`4-CIC,,H,CH2(J
`If:
`1
`I)
`0
`(1.5
`4-Me2CHC03Cfil-I4CHEO
`4—Me2CHC0?CfiH¢CH20
`l?
`s
`o
`o
`4
`apacocfifldcm
`4—AcOCfiH4CH3
`la
`8
`I)
`0
`4
`Br(CH3)20
`Br(CH_,_)30
`19
`6
`IS
`[3
`3
`CIECHCHzC}
`C12CHCH,O
`20
`o
`[3
`22
`o
`EtO{CH,}:O
`Et0(CH?)3O
`21
`26
`{J
`l
`13
`C,,H,O
`ChHSO
`22
`50
`U
`0
`25
`CnHFO
`CfiHSO
`23
`6
`0
`o
`3
`4-BrC6HdU
`4—BrChH40
`24
`22
`0
`t]
`l
`Z-McCfiH‘O
`2—MeCth0
`25
`It}
`0
`16
`5
`2—Ac0CbH‘O
`2—AcOCbH40
`26
`8
`0
`5
`4
`4-AcOCfiH40
`4-Ac0C,_I-[,O
`2'?
`0
`[l
`I
`0
`4»ACOCH_,C6H‘O
`4-AcOCl-IECbH40
`28
`m
`0
`l4
`5
`4AMcOCfiHJO
`cmeocth
`29
`
`30 I} 4-MeJCC02C6HJO 4-Me‘CCOJChH40 0 U 0
`
`
`
`
`
`"The bioavailability of 1 after oral administration of prodrugs was calculated from the equation: % bioavailability = (AUC l)!{iv
`AUG 1) X 100. where iv AUG 1 = 50 pM I1.
`
`I
`
`6
`
`

`

`J'.P. Krise. VJ. Stella ! Advanced Drug Delivery Reviews 19(1'996) 287—310
`
`293
`
`stable, were predominantly detected unchanged
`in the serum after oral administration. As the
`
`alkyl diester size increased the prodrugs tended
`to break down more efficiently to the monocster;
`however, the monoester tended to build up in
`the blood and failed to be converted to the
`
`[38] also
`parent phosphonate. Starrett et al.
`explored the effectiveness of dialkyl prodrugs to
`improve
`the
`bioavailability
`of
`9—[2—(phos-
`phonomcthoxy)ethyl]adenine (PMEA) and ob—
`tained similar results.
`
`Investigators also described the utility of the
`aryl functionality in prodrug design, both with
`unsubstituted and substituted aryl phosphate
`esters. When these were designed to increase
`cellular permeability, the ability of the prodrug
`to deliver
`the phosphate-containing drug in-
`tracellularly was minimal [39—43] probably due
`to removal of one or both of the esters prior to
`reaching the target cell. However, when the aryl
`functionality was used to increase GI tract per-
`meability several authors reported dramatic in—
`creases in bioavailability [35.38,44.45] (also refer
`to section 4.3.2 of this review). Table 1 contains
`oral bioavailability data for a number of aryl
`prodrugs of 9-[2—(phosphonomethoxy)ethoxy]-
`adenine (22-30). The most promising aryl pro-
`drug was the phenyl prodrug 23 (23 is a hydro—
`chloride salt of 22) which gave 50% oral bio—
`availability of the parent phosphonate.
`In an effort
`to produce more chemically!
`metabolically labile prodrugs, Mitchell et al. [46]
`synthesized the dibenzyl ester of methoxycar-
`bonylphosphonate. The hydrolysis
`in pH 7.4
`buffers (36.4°C) was rapid and resulted in a
`complex product profile. The major complication
`with this potential prodrug was that P—C cleavage
`occurred, yielding the dibenzyl phosphitc. In an
`attempt to shift the hydrolysis more toward P-O,
`or 0—bcnzyl group bond cleavage possibly yield-
`ing parent monophosphate-containing drug after
`hydrolysis,
`para-substituents
`on
`the
`benzyl
`groups were employed [4?]. As expected, para—
`electron withdrawing substituents yielded esters
`that hydrolyzed to the monobenzyl ester but
`little or no parent drug was formed. Only hydro—
`Iytic data was provided with these studies. A
`potential draw back in the clinical usefulness of
`benzyl esters with porn electron donating sub-
`
`stituents is the potential formation of quinone
`methide intermediates which are known to be
`toxic.
`
`By what mechanism do these alkyl esters of
`monophosphates exert their potential biological
`effect? The prodrugs could either act as depot
`for the nucleoside in which the P—O-nucleoside
`
`link would be broken to provide the nucleoside
`(pathway 1, Scheme 3). or act as an intracellular
`source of
`the nucleoside monophosphate by
`cleavage of
`the P-O-alkyl bond (pathway 2.
`Scheme 3).
`The first possibility (pathway 1), although
`potentially useful [48]. would fail to overcome
`the resistance seen with many of these drugs i.e.,
`it still delivers the nucleoside rather than its
`
`monophosphate.
`[31]
`To assess this issue, McGuigan ct al.
`prepared compounds analogous to conventional
`dialkyl phosphate prodrugs through the use of
`methylene bridges from the phosphorus to the
`alkyl chain (phosphinate linkage). The phosphi-
`nate (P-C)
`linkage is well known to be bio-
`logically stable [49]. The activity of di-n-butyl
`phosphinate ((C4H9)2PO—nucleoside) and dim-
`propyl
`phosphate
`((C3H,O)2PO—nucleoside)
`were compared (assumes similar Van der Waals
`radii for methyl and oxygen). Similar biological
`activities of di-n-butyl phosphinate and dim—pro-
`pyl phosphate could be expected if the mecha—
`nism of action was that both acted as depot
`prodrugs of the parent nucleoside. However, the
`phosphate was nearly twice as effective as the
`phosphinate. The fact that the phosphinate dis-
`played activity (albeil
`low) provides some evi-
`dence that
`the prodrugs may act,
`in part, as
`depots for the nuclebside. On the other hand.
`since the phosphates were consistently more
`active than their phosphinate counterparts, it was
`possible to infer that an increase in the phos-
`phate-to-alkyl link lability would produce more
`efficient prodrugs.
`In an effort
`to increase the [ability of the
`phosphate-promoiety bond (pathway 2, Scheme
`3), several halo alkyl prodrug esters have been
`made and evaluated [50,51]. The halo substituv
`tion was thought
`to favor
`the formation of
`nucleoside monophosphate and increase biologi-
`cal activity. Bis(2,2,2~trihaloethyl) phosphate de-
`
`7
`
`

`

`294
`
`LP. Krist’. VJ. Swim it Advanced Drug Delivery Reviews I9 ([996) 287—31’0
`
`./”“‘~.
`
`I
`
`1H:
`
`i
`\N/
`o\l
`"\.__________.9-
`
`no
`
`+
`
`it
`RO-P—OH
`I
`OR
`
`
`
`{gang}
`
`0
`\N/
`ll
`Ho—f—o—l/Oq
`a\.....
`5_J
`OH
`\_
`
`t
`
`[120le
`
`Scheme 3. illustration of the possible decomposition pathways for a generic nucleotide prodrug. Pathway | represents cleavage of
`the P-O—nucleosidc link and results in nuclcosidc release. Pathway 2 represents Cleavage of the P—O—alkyl linkage and results in
`accumulation of nucleotide inside the cell.
`
`2’,3’-dideoxycytidine
`of AZT and
`rivatives
`(Fig. 5) have been synthesized and
`(ddCD)
`evaluated [52].
`The in vitro anti—HIV activities of these com—
`
`pounds were evaluated and it was found that
`none of the prodrugs were as active as the parent
`nucleosides. The trichloroethyl moiety was found
`to be more efficacious than the trifluoroethyl
`moiety for ddCDMP: however, the two halogen
`prodrugs were
`nearly equally effective
`as
`AZTMP. When this promoeity was subsequently
`applied to ara-AMP and ara-CMP [53],
`in vitro
`testing showed the trichloro-containing prodrugs
`were consistently more active than the trifluoro—
`
`containing prodrugs. For the ara-CMP prodrug,
`the activity was found to be greater than that of
`the parent nucleoside. These observations were
`not consistent with the expected lability of the
`prodrug esters, as one would expect the trifluoro
`analogs to be the most labile. The observed trend
`in activity, however. was found to correlate with
`lip0philicity as exemplified by the larger OCIaI'IOU
`water partition coefficient for the tfichloro—con—
`taining prodrug. Subsequently, experiments were
`conducted to discover the potential utility of
`other related halo—alkyl prodtugs [54]. Mono—,
`di-. and trichloro substituted alkyl prodrugs of
`AZTMP (Fig. 5) were synthesized and com—
`
`0
`
`n
`RU-Fr'o
`0“
`
`NH2
`
`Me
`
`N/ I
`0J\
`
`N
`
`0
`
`H
`
`N3
`
`AZTMP (R = H}
`
`R= Hit?!l;
`R -—. CI3CCH;
`
`R : Elect-[CHE
`
`R = CICHZCH;
`
`OOJU
`
`N
`
`ll
`RO-FI’—O
`OR
`
`NH2
`
`N/
`
`R : cheer]2
`
`a rjccn1
`
`0
`
`ddCDMP(R = H]
`
`Fig. 5. Structures of AZ'I‘ mon0phosphate and ddCD monophosphate along with their corresponding haloalkyl prodrugs {52.54}.
`
`8
`
`

`

`LP. Krise. VJ. Stella 1" Advanced Drug Delivery Reviews [9 ([996) 287—310
`
`295
`
`pared. The expected lability of these prodrugs
`would be in the order
`trichloro>dichloro>
`
`monochloro. The observed activity. however, was
`in
`the
`order
`of
`triehloro>monoehloro>
`
`dichloro. When the haloalkyl prodrugs were used
`to increase bioavailability, only marginal
`in—
`creases were observed (see Table 1) [35,38].
`The results on alkyl, haloalkyl, benzyl and aryl
`prodrugs tend to imply that ability of
`these
`prodrugs to biorevert from the diester to mono--
`ester prodrug to the parent phosphate or phos-
`phonate does not solely rely on their chemical
`lability. For example, prodrugs with good leaving
`groups were not always the most efficient pro-
`drugs. What mechanism(s) are involved in the in
`vivo reversion of these prodrugs? The conversion
`of the diester prodrug to the monoester prodrug
`is not well understood and the in vivo rate of
`
`conversion of the dies‘rer prodrug to the monoes-
`ter prodrug is too rapid to be explained solely by
`chemical hydrolysis. On the other hand, no one,
`to our knowledge. has identified a true phosphot—
`riesterase in the mammalian system. One could
`speculate either non-specific enzymes or micro
`environmental pH changes are responsible for
`the conversion. Conversion of the monoester
`
`prodrug to the parent phosphatetphoSphonate
`drug can be explained by the presence of phos—
`phodiesterases (and phosphonodiesterases) [55—
`5?].
`In addition, phosphonate monoesters and
`phosphate diesters with systematically varied
`leaving groups were tested as substrates for 5’—
`nucleotide phosphodiesterase [58]. Although ali—
`phatic and benzyl esters were poor substrates,
`the aryl ester derivatives were shown to be good
`substrates for their diesterases. It was suggested
`that the geometry of the aryl group may be more
`important than the inductive nature of the aro—
`matic group.
`
`4.1.2. Acyloxyalkyt esters
`One approach to overcome the hydrolytic
`resistance observed with many of the prodrugs
`previously reviewed would be to utilize an en-
`zymatically labile promoiety; acyloxymethyl pro—
`drugs could serve as neutral lipophilic prodrugs.
`In theory, these prodrugs could transverse cell
`membranes by passive diffusion and revert, in-
`tracellularly.
`to the parent
`ionic phosphate or
`phosphonale after cleavage of the acyl group by
`esterases and rapid elimination of formaldehyde
`spacer group (Scheme 4).
`
`O
`ll /\ II
`2 a—c—o
`o P—O
`
`{/J MN“:
`
`\N/
`o\|
`1/
`\......-x
`
`Ester-rise
`——~—
`
`/""‘-. K
`
`0
`\N/
`li
`ll
`o
`R—vc—-—~0/\0-P—0
`. 1/ a
`<
`‘-._.....J
`
`OH
`
`{1120
`Fast
`
`/"\_:
`
`...___ +¢—~—
`
`li
`R—C—O
`
`\N/
`ii
`o
`O-P--0
`a. 1/ \.|
`L4
`
`{Ml/"Ra‘s
`
`ii
`\N/
`“04.4)
`0\J
`5.. 1/
`.
`no;
`
`Scheme 4. General mechanism for the bioreversion of acyloxymethyl phosphate prodrugs. The acyl group is cleaved through
`esterase activity to yield the hydroxymethyl analog. This analog quickly decomposes to formaldehyde and the monoester prodrug.
`The second group is subsequently cleaved by the same mechanism but possibly a different enzyme.
`
`9
`
`

`

`296
`
`JJ’. Krise. VJ. Stella it Advanced Drug Delivery Reviews [9 {F996} 287—3”)
`
`Investigators have utilized this strategy and
`evaluated the acyloxymethyl ester prodrugs for a
`variety of antiviral agents [59—61,l39] and CAMP
`[60]. Starrett et al.
`[62] have synthesized the
`bis(pivaloyloxymethyl) prodrug of the antiviral
`agent PMEA to increase antiviral activity. Star-
`rett’s studies showed the prodrug to have sub—
`stantially increased anti—herpes
`simplex virus
`activity in vitro compared with PMEA. Similarly.
`Sastry et
`al.
`[63] have produced the bis-
`(pivaloyloxymethyl) prodrug of 2’,3'-dideoxy-
`uridine 5'-mon0ph05phate (ddUMP). Addition-
`ally, metabolism studies in two human T cell
`lines lead to the formation of 5’-mono-. di~. and
`
`triphosphates of ddU after exposure to bis—
`(pivaloyloxymethyl) ddUMP.
`In contrast.
`these
`phosphorylated metabolites were not observed in
`cells treated with ddU or ddUMP alone,
`sug—
`gesting that this prodrug was able to intracellu-
`larIy release ddUMP. Freed et al.
`[64] utilized
`this same prodrug strategy for 5—fluorodcoxy—
`uridine monophosphate (5dUMP) and obtained
`results consistent with intracellular delivery of
`the monophosphate. Bis(pivaloyloxymethyl) pro—
`drugs of 9-(2—phosphonylmethoxycthyl)adenine
`( PMEA)‘
`9—( 2—phosphonylmethoxypropyl )ad—
`eninc (PMPA). and 9-(2—phosphonylmethoxy-
`propyl)diaminopurine
`{PMPDAP} were
`also
`synthesized and compared with the unmodified
`analogs. The bis(pivaloyloxymethyl) derivatives
`were biologically more active and showed en—
`hanced antiviral activities in vitro [65], most
`probably due to increased permeability of the
`prodrugs compared to the parent compound. In
`order to gain an understanding of the metabo~
`lism
`of
`this
`prodrug
`system,
`the
`[3H]bis(pivaloyloxymethyl) PMEA was
`incu-
`bated in an in vitro cell culture system. It was
`observed that
`the
`["H[bis(pivaloyloxymethy|}
`PMEA was rapidly hydrolyzed to the parent
`compound (PMEA) within the cells and was
`further metabolized to the mono- and sub-
`
`sequently to the diphosphate analog. It was also
`found that the diester prodrug releases a substan-
`tial quantity of the rnono(pivaloyloxymethyl)
`PMEA extracellularly. which could be a limita—
`tion
`of
`these
`prodrugs
`since
`the mono-
`(pivaloyloxymethyl)
`specie would not
`likely
`penetrate biological membranes.
`
`In addition to increasing antiviral activity,
`investigators have also evaluated acyloxyalkyl
`ester
`prodrugs of PMEA and
`9-[2-(phos-
`phonomethoxy)ethoxy] adenine in order to in—
`crease their poor oral bioavailabilities [35.38].
`Table 1, compounds 6—13, showed dramatically
`increased oral bioavailability for some acyloxy
`alkyl prodrugs. The bis(pivaloyloxymethyl) pro—
`drug (6) had reached an oral bioavailability of
`30%: however, methyl substitution at the alpha
`carbon of 6. led to an oral bioavailability of 74%
`for 13. Shaw et al. [66] tested alkyl. aryl, alkox-
`yalkyl. and alkyl amino acid phosphoramidate
`esters. Based upon an in vitro stability and
`transport
`screen.
`the
`bis(pivaloyloxymethyl)
`PMEA was selected as a potential oral prodrug
`for further in vivo animal studies. The bioavail-
`
`ability of bis(pivaloyloxymethyIJPMEA utilizing
`three different
`formulations was subsequently
`evaluated in monkeys [67]. The three formula-
`tions (hydroxypropyl—fi—cyclodextrin. PEG, and
`aqueous suspension) were used to explore any
`dissolution limitations with

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