throbber
NEW TOOLS IN SYNTHESIS
`
`SYNLETT
`
`233
`
`Pro-Nucleotides - Recent Advances in the Design of Efficient Tools for the Delivery of
`Biologically Active Nucleoside Monophosphates
`C. Meier*
`Institut für Organische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
`Tel. +49-931-8885326; Fax +49-931-8884606; e-mail: meier@chemie.uni-wuerzburg.de
`Received 10 September 1997
`Dedicated to Professor Gernot Boche on the occasion of his 60th birthday
`
`This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.
`
`two nucleotide kinases)(8,9). Finally, the resistance of the human
`immunodeficiency
`virus
`to
`the
`clinically
`used
`antiviral
`dideoxynucleoside AZT 2 (Zidovudine, Retrovir®)(10) is on the one
`hand directly related to multiple point mutations within the HIV-1
`reverse transcriptase gene of the virus but may on the other hand be also
`due to lower susceptibility of resistant target cells to the drug related
`with a decreased activity or inability of the enzyme thymidine kinase to
`phosphorylate AZT 2
`to
`the dideoxynucleoside monophosphate
`AZTMP 9(11).
`
`Consequently, direct administration of the nucleotides d4TMP 4,
`ddAMP 8 and AZTMP 9 should bypass these limiting steps and hence
`has advantages for the biological activity. Unfortunately, because of the
`high polarity of the nucleoside monophosphates (nucleotides), these
`compounds are not able to easily penetrate cellular membranes or the
`blood-brain barrier. However, the phosphate moiety offers a suitable
`site to attach degradable lipophilic carrier residues. As a result, one
`effort to improve the therapeutic potential of nucleoside analogues is the
`delivery of the corresponding nucleotide from neutral, membrane-
`permeable prodrugs (Pro-Nucleotide Approach; Figure 2)(12).
`
`Abstract: A summary of the most recent advances to the design of pro-
`nucleotides will be presented. Approaches that have been designed to be
`activated by enzymes such as carboxyesterases [bis(POM)-, bis(POC)-,
`bis(SATE)-, bis(AB) phosphotriesters and the arylphosphoramidates] or
`by reductases [bis(SDTE) approach] will be discussed as well as the
`amino acids phosphoramidate diester concept with its still unknown
`delivery mechanism and the cycloSal approach that releases the
`nucleotides by an induced tandem reaction.
`Nucleoside analogues, e.g. 2',3'-dideoxy-2',3'-didehydrothymidine 1
`(d4T), 3'-azido-2',3'-dideoxythymidine 2
`(AZT) or 5-fluoro-2'-
`deoxyuridine 3 (5-FdU), are structurally different as compared to the
`corresponding natural DNA or RNA nucleosides with regard to
`modification of the glycon as well as the aglycon residue. Due to this
`modified structure, these compounds are widely used as antiviral or
`antitumor drugs in chemotherapy (Figure 1)(1). Since the discovery of
`AZT 2 as the first nucleoside drug for the treatment of AIDS,
`considerable efforts have been made to develop new nucleoside
`analogues that would be more active, less toxic inhibitors of the HIV-1
`reverse transcriptase (RT)(2). The general mode of action of nucleoside
`analogues is the inhibition of the HIV-1 RT by acting as competitive
`inhibitors or as DNA chain terminators. To act as DNA chain
`termination agents or RT inhibitors, intracellular conversion of the
`nucleoside analogues into their 5'-mono-, 5'-di- or 5'-triphosphates is a
`prerequisite after cell penetration(3). The enormous disparity in anti-
`HIV activity that is evident for a large number of dideoxynucleoside
`analogues belies their apparent structural similarity. Due to these
`structural differences as compared
`to natural nucleosides
`the
`metabolization to the corresponding dideoxynucleoside triphosphates is
`often inefficient and consequently the therapeutic efficacy is sometimes
`limited(4). For example,
`in
`the case of
`the anti-HIV active
`dideoxynucleoside analogue d4T 1 (Stavudine, Zerit®)(5) the first
`phosphorylation to d4T 5'-monophosphate 4 catalyzed by thymidine
`kinase (TK) is the rate-limiting step in human cells(6). More striking,
`however is 2',3'-dideoxyuridine triphosphate (ddUTP) which is one of
`the most powerful and selective inhibitors of HIV reverse transcriptase
`(Ki = 0.05 μM) while the parent nucleoside 2',3'-dideoxyuridine 5 (ddU)
`is virtually ineffective at blocking HIV infection in cultured cells.
`Biochemical and pharmacological studies in three different human T
`cell lines (CEM, ATH8 and Molt-4) showed that ddU 5 itself was not
`anabolized to the 5'-monophosphate, most apparently because it was a
`poor substrate for cellular nucleoside kinases because of
`the
`considerable substrate specificity of these enzymes(7). In contrast, in a
`few cases the limited efficacy is also due to a catabolic enzymatic
`reaction. For example, 2',3'-dideoxyadenosine 6 (ddA) is rapidly
`intracellularly deaminated to ddI 7 by adenosine deaminase (ADA)(8,9).
`As a consequence, ddI 7 has to be converted into its ultimate bioactive
`metabolite ddATP via ddAMP 8 by five enzymatic steps (5'-
`nucleotidase, adenylosuccinate synthase, adenylosuccinate lyase and
`
`1
`
`GIL2017
`I-MAK, INC. V GILEAD PHARMASSET LLC
`IPR2018-00119
`
`

`

`234
`
`C. Meier
`
`SYNLETT
`
`This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.
`
`group attached to the phosphorus are known to be toxic suicide
`inhibitors of acetylcholinesterase(15). The anti-acetylcholinesterase
`activity of phosphorus derivatives is an inverse function of the pKa of
`the leaving group on the phosphorus atom and parallels the rate of
`spontaneous hydrolysis by P-O bond cleavage(16). In order to
`circumvent the possible problem of anti-cholinesterase activity, neutral
`phosphate ester prodrugs should be designed to undergo heterolytic
`cleavage of the C-O bond rather than the P-O bond of the ester.
`Many strategies have been developed to achieve this goal. As a general
`motive, uncharged nucleotide triesters are used as membrane-permeable
`nucleotide precursors(12). The major differences of these approaches are
`the delivery mechanisms to the nucleotides. First attempts have been
`made with simple dialkyl phosphotriesters. These compounds generally
`belong to the class of bipartate prodrug systems. After a first, sometimes
`selective hydrolysis of the phosphotriester via a nucleophilic reaction at
`the phosphorus center, the resulting phosphodiester is often extremely
`stable against a further chemical hydrolysis due to the charge at the
`phosphate which prevents a second nucleophilic reaction(17). Even if the
`subsequent hydrolysis is possible, one should take the pseudorotation
`phenomenon into account that excludes a selective delivery of the
`nucleotide(18). As a consequence, almost all approaches based on
`chemical hydrolysis reported so far were unable to deliver the
`nucleotide selectively except the cycloSal approach that will be
`discussed later. For this reason, the newer pro-nucleotide approaches are
`based on the concept of a tripartate prodrug system(14) and are based on
`the general idea of a selective chemical or enzymatic reaction within the
`masking group which leads to a second, spontaneous successive
`reaction yielding the charged phosphate ester. These approaches utilize
`and exploit the differences in reducing potentials, enzyme activity, and
`pH value. The concepts working with enzymatic trigger processes
`(bis(POM)-, bis(POC)-, bis(DTE)-, bis(SATE)-, bis(AB)- and the
`arylphosphoramidate concept) as well as the cycloSal approach based
`on a pH-driven degradation have demonstrated
`the successful
`intracellular delivery of free nucleotides from lipophilic precursors.
`It should be added, that it is not the intention of the author to give an
`entire overview of the pro-nucleotide field. The current review
`highlights some approaches that have been designed to deliver the
`nucleotide selectively by a special mechanism. With this selection, the
`author wants to point out that designing a delivery mechanism and
`bypassing a certain limiting metabolization the inherent biological
`potential of an already known nucleoside analogue could be used to a
`higher extent and consequently, the improvement in antiviral activity
`could be, in many cases, better than synthesizing new potential
`nucleoside analogues. Furthermore, this review is restricted to the
`delivery mechanism and so the synthesis of the compounds may be
`gleaned from the original literature.
`
`Chris Meier, born 1962 in Berlin, was trained in Chemistry at the
`University of Marburg. He passed his Diploma and Ph.D. thesis in the
`group of Prof. G. Boche in Organic Chemistry. Then he moved as post-
`doc fellow to the Pasteur-Institute in Paris where he started his work in
`nucleoside and oligonucleotide chemistry. In 1991 he joined the group
`of Prof. J.W. Engels at the University of Frankfurt/Main starting his
`Habilitation which he finished in July '96 and in May '97 he was
`appointed as an associate professor at the University of Würzburg.
`
`So, a lipophilic phosphotriester may penetrate into the target cell where
`first partial and at the end complete hydrolysis delivers the nucleotide.
`A suitable nucleotide prodrug has to fulfill two requirements: i) it has to
`be lipophilic enough for passive diffusion of the membrane and blood-
`brain barrier; ii) furthermore, it should be able to deliver the nucleoside
`hydrolytically or enzymatically leaving a non-toxic masking group(13).
`In principle, two different concepts for prodrug design are known:
`bipartate and tripartate prodrugs. In the former concept the drug is
`modified by a one-component masking group. In this form the drug is
`biologically inactive. After a simple cleavage of the mask, the active
`drug is liberated (Figure 3). In the latter concept, the drug is modified by
`a two-component masking group. Again, the drug is biologically
`inactive in this bound form. The mechanism of liberation involves a first
`chemical or enzymatic reaction under cleavage of part I of the masking
`moiety. The drug is still inactive but the effect of this first reaction is an
`activation of the remaining masking group II with the consequence of a
`spontaneous successive cleaving reaction releasing the now bioactive
`drug (Figure 3)(14).
`
`In the case of a nucleotide prodrug one should take into account that
`under physiological conditions two negatively charged phosphate
`oxygen's have to be masked in order to obtain a neutral, lipophilic
`phosphate ester. Consequently, not only one masking group is necessary
`but two. So, the efficient intracellular delivery of nucleotides from a
`prodrug requires the existence of a specific delivery mechanism or
`different rates of conversion of the prodrug to the drug intracellularly
`versus extracellularly. One comment with respect to toxic side events
`should be given. Neutral phosphorus derivatives with a good leaving
`
`Biographical Sketch
`
`2
`
`

`

`This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.
`
`March 1998
`
`Pro-Nucleotides - Recent Advances in the Design of Efficient Tools
`
`235
`
`Bis(POM)- and Bis(POC)-Nucleotides
`The first approach reported by D. Farquhar et al. towards a new class of
`tripartate pro-nucleotides are
`the bis(pivaloyloxymethyl)- [POM]
`phosphotriesters of general type 10 (shown exemplified as the ddU
`derivative)(19,20). This approach utilizes a carboxyesterase-catalyzed
`cleavage of the pivaloyl ester within the POM-masking group to yield
`reactive hydroxymethyl phosphotriester 11 which
`the highly
`subsequently eliminates spontaneously formaldehyde to give the
`mono(POM) phosphodiester 12. The carboxyesterase which is used for
`this activation process may be more prevalent inside the cells. To obtain
`the free nucleotide, this enzymatic activation has to be repeated via 13
`or, alternatively, a phosphodiesterase cleaves the phosphodiester 12
`directly to yield the nucleotide (Figure 4).
`
`been shown that the bis(POM) phosphotriesters 10 were chemically
`unstable and highly susceptible to serum-mediated hydrolysis, factors
`which limit their potential utility for intracellular drug delivery(22,24a).
`A modification of the bis(POM)-approach and a way to overcome the
`limitations
`observed
`with
`these
`compounds
`are
`bis(isopropyloxycarbonyloxymethyl)-
`[bis(POC)]
`nucleotides
`of
`general type 14 (shown as the PMPA derivative) that have been
`published very recently by A. Fridland et al.(26) and L. Naesens et
`al.(27). This modification uses a carbonate diester within the masking
`group. The mechanism of action is again a carboxyesterase-catalyzed
`cleavage of the isopropyl ester to give isopropanol and in the shown
`example the intermediate phosphonate diester 15 that fragmentates via
`16 into carbon dioxide and formaldehyde and finally to the mono(POC)
`phosphonate ester 17. The mono(POC) ester 17 is subsequently
`degraded to yield PMPA or generally the nucleotide after a second
`carboxyesterase activation or by phosphodiesterase cleavage (Figure 5).
`
`This approach has been applied successfully to the delivery of 2',3'-
`bis(POM)-ddUMP(21).
`dideoxyuridine monophosphate
`from
`Furthermore, this concept has been used for the delivery of the anti-HIV
`drug AZTMP(22),
`the
`anti-herpes
`and
`anti-HIV
`drug
`phosphonomethoxyethyladenine (PMEA)(23) and for the antitumor
`active drug 5-fluoro-2'-deoxyuridine monophosphate (5-FdUMP)(24). In
`the case of the phosphonate analogue PMEA a highly improved
`bioavailability was obtained. Moreover, the bis(POM) derivatives of
`(R)-9-(2-phosphonomethoxypropyl)adenine
`(PMPA) and
`(R)-9-(2-
`phosphonomethoxypropyl)diaminopurine (PMPDAP) were found to be
`9- to 23-fold more active than the parental compounds(23). However, the
`cytotoxicity of the bis(POM) analogues was also increased by virtually
`similar degrees. Nevertheless, studies with radiolabelled bis(POM)-
`PMEA showed a considerable increase (100-fold) of the cellular uptake
`of PMEA by using the phosphonate diester(23). In an independent study,
`ten different PMEA prodrugs tested in vivo for oral bioavailability in
`cynomolgus monkeys. From the results of this study, bis(POM)-PMEA
`was selected for human clinical trails. In these clinical trails, the oral
`bioavailability of bis(POM)-PMEA from a single 500 mg dose was
`greater than 40% in fed subjects(25). The in vitro antitumor tests of the
`bis(POM)-5-FdUMP triester demonstrated high growth inhibition in
`cell lines that were resistant to the usually used drug 5-fluorouracil.
`Furthermore, this triester derivative showed in vivo bioactivity after
`intraperitonal application against P 388 leukemia in mice. Beside the
`antitumor activity, these results verified also the penetration of the pro-
`nucleotide through cellular membranes(24). A drawback of this approach
`is the requirement of a second identical activation reaction because the
`intermediate phosphodiester is a significantly poorer substrate for the
`activating carboxyesterases. Moreover, the delivery of one molecule of
`the nucleotides results in the liberation of two equivalents of potentially
`toxic formaldehyde and pivalinic acid (Figure 4). Additionally, it has
`
`In contrast to the bis(POM)-approach, the bis(POC) modification avoids
`the formation of two equivalents of pivalinic acid that accumulate in the
`cells and potentially cause toxicity. The bis(POC)-approach was applied
`to the anti-HIV drug PMPA in order to increase the low bioavailability
`of
`this phosphonate derivative (Figure 5). Bis(POC)-PMPA
`is
`chemically stable at low pH and has shown 30% bioavailability in dogs
`with minimal toxicity in repeat 5-day dosage of 60 mg/kg/day. The anti-
`HIV activity of bis(POC)-PMPA 14 in human peripheral blood
`lymphocytes and in dentritic T-cell coculture system was 35- and 16-
`fold, respectively, higher than that of PMPA. Bis(POC)-PMPA 14 was
`non-toxic at concentrations that completely suppressed viral replication.
`Studies of the metabolism of [3H]-bis(POC)-PMPA showed that is was
`readily taken into the human cells, hydrolyzed to PMPA, and
`phosphorylated to the mono- and diphosphate derivatives. These results
`show that bis(POC)-PMPA 14 is a membrane-permeable form of PMPA
`and shows promise as a drug for the treatment of HIV infections(26).
`
`Bis(SDTE)- and Bis(SATE)-Nucleotides
`Two further approaches that are also based on enzymatic activation
`have been reported by J.-L. Imbach and G. Gosselin(28). They designed
`the bis(S-[2-hydroxyethylsulfidyl]-2-thioethyl)- [bis(SDTE)] 18 and the
`bis(S-acyl-2-thioethyl)- [bis(SATE)] nucleotides 19. The former
`concept was constructed to take advantage of the greater reducing
`potential within the cells to liberate the nucleotide into the cytosol. After
`
`3
`
`

`

`236
`
`C. Meier
`
`SYNLETT
`
`This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.
`
`stability of the bis(SATE) phosphotriesters could be adjusted by varying
`the thio ester residue: the lowest stability was obtained for thio esters of
`acetic acid whereas with increasing alkyl residue of the carboxylic acid
`the stability was increased. The optimized compounds are bis(SATE)
`compounds bearing two S-pivaloyl-2-thioethyl side chains as shown in
`Figure 6 because the highly lipophilic and sterically demanding t-butyl
`residue protects the masking group from too rapid cleavage as compared
`to the S-acetyl or S-i-propyl counterparts(28d,30a,31). Again, J.-L. Imbach
`et al. demonstrated an increased stability of the bis(SATE) derivatives
`of AZTMP against degradation in culture medium as compared to
`cellular extracts(34). The bis(SATE) approach has been successfully
`applied to the thymidine kinase-bypass (TK-bypass) of d4T 1(35)
`(shown in Figure 6) and AZT 2(34), the adenosine deaminase-bypass
`(ADA-bypass) of the anti-HIV active nucleoside ddA 6(36) as well as for
`an improvement of the bioavailability of the anti-herpes and anti-HIV
`drug PMEA. Furthermore, the corresponding bis(SATE)-ddAMP
`triesters exhibited a higher stability than the dideoxynucleoside ddA 6
`against acid-catalyzed depurination(36a). It should be mentioned that the
`bis(SATE)-AZTMP derivatives demonstrated a 10-fold decrease in
`activity from the wild-type CEM/O cell line to the mutant TK- CEM cell
`line(34). This result is puzzling but has also been observed by others. On
`the other hand, J. Cinatl, Jr. et al.(11a) have shown that the bis(SATE)-
`AZTMP derivatives retained the biological activity of AZT 2 in AZT
`resistant Molt4/8 cells. Especially bis(S-pivaloyl-2-thioethyl)-AZTMP
`showed comparable cytotoxic and antiviral activity in sensitive and
`resistant cells. With these experiments, J. Cinatl et al. demonstrated that
`Molt4/8rAZT cells exert resistance to the anti-HIV activity of the drug
`mainly owing to the lack of AZT phosphorylation and that resistance
`may be bypassed by using AZT monophosphate prodrugs. However, as
`for the bis(POM) phosphotriesters 10 reported by D. Farquhar, the
`intermediate mono(SATE) phosphonate esters 21 and 23, respectively,
`are significantly lower substrates for the activating carboxyesterases due
`to the negative charge at the phosphonate residue in the vicinity of the
`enzyme cleavage site. Before hydrolysis takes place, the bis(SDTE) as
`well as the bis(SATE) phosphotriesters may serve as lipophilic
`precursors for a passive membrane permeation.
`J.-L. Imbach et al. presented the comparison of the bis(SATE)-approach
`to the isomeric S,S'-bis(O-acyl-2-oxyethyl) phosphorodithiolates(37).
`Surprisingly, these derivatives proved as active as the original
`bis(SATE) triesters with the nucleosides d4T 1 and ddA 6, although the
`delivery mechanism could not be the same. Further work is in progress
`to study the cleavage mechanism. Recently, the same group extended
`their prodrug design to bis(S-glycopyranosyl-2-thioethyl) [bis(SGTE)]
`nucleoside phosphotriesters(38). These compounds were designed to
`deliver the nucleotide by activation of glycosidases. However, no
`antiviral data have yet been published.
`
`Bis(AB)-Nucleotides
`One possibility to avoid the close vicinity of the negative charge of the
`intermediate phosphodiester and the enzyme cleavage site has been
`developed independently by S. Freeman et al.(39) and A. Glazier(40).
`The common motive of their approaches is to separate the phosphate
`group and the carboxyesterase cleaving site by a rigid spacer.
`Consequently,
`they are using bis(4'-acyloxybenzyl)-
`[bis(AB)]
`nucleotides 24 and 25, respectively (shown as their AZTMP derivatives)
`instead of the O-acyloxymethyl or the S-acyl-2-thioethyl residue,
`respectively. As Freeman calculated a separation of about 4 Å, which
`she states is enough to avoid electrostatic disturbance of the anionic
`phosphate and the carboxyester residue(41). The degradation mechanism
`of the bis(AB) nucleotides of type 24 is the following: the enzyme
`cleaves the ester moiety in the 4'-position of the aromatic ring to give
`
`the reductase cleavage of the disulfide bond in 18 with formation of
`thioethanol and thioethyl phosphotriester 20, this compound eliminates
`spontaneously episulfide to yield the intermediate phosphodiester 21.
`As in the case of the bis(POM)- (10) and the bis(POC) phosphotriester
`14, the bis(SDTE) derivative 18 requires a second identical enzyme-
`catalyzed activation step (Figure 6). The initial idea of this concept was,
`that the "soft" thiol-nucleophile in 20 will attack the "soft" electrophilic
`α-carbon atom rather than the "hard" phosphorus atom in the
`elimination reaction avoiding possible pseudorotation processes(18)
`because all reactions take place within the masking group without
`involving the phosphate ester moiety. This concept belongs also to the
`class of tripartate prodrugs. It could be shown that the bis(SDTE)
`triester of the nucleotides ddUMP and AZTMP 9 were cleaved 30-fold
`faster by reductases in cell extracts than in culture medium(28c).
`Furthermore, the bis(SDTE) approach has been applied to the
`nucleoside 5-FdU 3(29) and to the phosphonate nucleotide PMEA(30).
`Whereas an improvement in antiviral activity most probably due to an
`enhanced bioavailability for the bis(SDTE)-PMEA derivative 18
`compared with PMEA was found, the bis(SDTE)-5-FdUMP failed to
`show better antitumor potency as compared to the free nucleoside(29).
`The major limitation of this approach is the restricted chemically
`stability and high susceptibility to serum-mediated hydrolysis (Figure
`6).
`
`In contrast to the bis(SDTE) approach, the bis(SATE) approach is
`similar to the bis(POM) concept with respect to the activation process.
`Again, carboxyesterases are used to trigger the nucleotide delivery.
`Here, the enzyme cleaves a thioester in 19 to yield a carboxylic acid (as
`in the bis(POM) approach pivalinic acid) as well as the above
`mentioned thioethyl phosphonate diester 22 which undergoes a
`fragmentation to episulfide and the phosphonate monoester 23 (Figure
`6). Thus, the spontaneous, second step is identical in the bis(SDTE) and
`the bis(SATE) concepts. Nevertheless, again two independent activation
`steps are required to yield the free nucleotide. With these two
`approaches, two equivalents of episulfide, which has been shown to be
`chronically and acutely toxic in mice and rats, are released besides the
`nucleotide(31). The toxicity and moreover also mutagenicity has also
`been observed in vitro(32). J.-L. Imbach and J.-P. Sommadoussi have
`shown that the introduction of the SDTE and the SATE group do not
`induce additional cytotoxicity of the phosphotriesters as compared to
`the parent nucleosides (e.g. AZT) in human myeloid colony-forming
`cells (human bone marrow cells)(33). It should be mentioned that the
`
`4
`
`

`

`March 1998
`
`Pro-Nucleotides - Recent Advances in the Design of Efficient Tools
`
`237
`
`This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.
`
`benzyl)methylphosphonate. As expected, the final products after
`enzymatic deesterification were methylphosphonic acid, acetic acid and
`trans-4'-hydroxycinnamic acid 34. Without addition of esterase no
`detectable decomposition was noted. Secondly, Glazier demonstrated
`convincingly, that after two hours of incubation with porcine liver
`esterase a nucleoside phosphotriester bearing the anti-herpes drug
`acyclovir monophosphate (ACVMP) has nearly completely been
`converted to methanol, acetic acid, trans-4'-hydroxycinnamic acid 34 as
`well as ACVMP as sole reaction products(45). This concept has been
`applied to the delivery of AZTMP(40) and ACVMP(45). However, a
`limitation of this approach may be the very short degradation half-lives
`of the compounds (t1/2 = 16 min). Furthermore, the bis(AB) derivatives
`reported by A. Glazier et al. were highly lipophilic (log Pa value = 1.07
`to 4.18; depending on the carboxyacid ester residue) making systemic
`applications problematic(45). Nevertheless,
`the bis(AB)-ACVMP
`derivatives showed promising in vivo activity against Herpes virus type-
`1 infections without toxic side effects using concentrations up to 100
`mg/kg body weight(45). Furthermore, no mutagenicity was found for the
`prodrugs in the Ames test with or without microsomal activation(45).
`However, the compounds showed only a limited improvement in
`antiviral activity in vitro(45). As acyclovir, the prodrugs showed no
`toxicity in vitro (CD50 >100 μM). Moreover, bis(AB) esters of PMEA
`were prepared and were found to be more potent in vitro than PMEA
`itself(40).
`
`Nucleoside Arylphosphoramidates
`triggered
`Another novel class of a membrane-soluble enzyme
`nucleotide delivery concepts are the phosphoramidate derivatives of
`type 35 recently reported by C. McGuigan et al.(46). The basic structure
`of these compounds (shown with the anti-HIV drug d4T 1) is depicted
`in Figure 8. In contrast to the previously mentioned phosphotriester
`approaches, in this case the structural motive involves a phosphate
`moiety that is linked to the nucleoside, a phenyl group and, through a
`phosphoramidate linkage, also to the methyl ester of α-amino acid ester.
`This concept has been applied to the wide variety of nucleotides active
`against several viruses: d4TMP(47), AZTMP(48), 3'-thiacytidine
`monophosphate (3TCMP)(49), ddAMP(50) and 2',3'-dideoxy-2',3'-
`didehydroadenosine monophosphate (d4AMP)(50,51).
`In in vitro antiviral tests, the d4T containing phosphoramidate was
`found to be slightly active as the parent nucleoside in wild-type CEM/O
`and MT-4 cells. More importantly and in contrast to d4T 1, the d4TMP
`phosphoramidate completely retained the biological activity in mutant
`thymidine kinase-deficient CEM/TK- cells(47) and suppresses HIV-1
`infection in natural peripheral blood lymphocytes and freshly isolated
`monocyte/macrophages. This result proves that d4TMP 4 was delivered
`intracellularly making the observed activity of the compound almost
`entirely independent of thymidine kinase and consequently these
`compounds serve as efficient tools for the TK-bypass. The d4T-
`phosphoramidate was found to be equally active against HIV-1 and
`HIV-2 replication, and has also proved inhibitory to other retroviruses
`including
`simian
`immunodeficiency virus
`(SIV)
`and
`feline
`immunodeficiency virus (FIV), Visna virus and Moloney murine
`sarcoma virus in cell culture(52). An interesting structure-activity
`relationship (SAR) has been found with these compounds: first, the
`presence of an α-amino acid ester is essentially important for the
`biological activity. In contrast to α-amino acid bearing compounds,
`simple alkyl amine derivatives exhibited a complete loss of antiviral
`activity(53). The same was observed for derivatives bearing β-amino
`acid side groups. Among the natural α-amino acids L-alanine was found
`to be the most effective(54) while the enantiomeric D-alanine showed a
`30-fold decrease in bioactivity(55). This interesting SAR points to an
`
`the 4'-hydroxybenzyl phosphotriester 26. Again, a subsequent
`spontaneous fragmentation of 26 is induced by this enzymatic reaction
`the formation of phosphodiester 27 and
`resulting
`in
`the 4'-
`hydroxybenzyl cation 28 which
`is oxidized
`to yield
`the 4'-
`quinonemethide 29 or
`is quenched by water
`to yield 4'-
`hydroxybenzylalcohol 30. Diester 27 could then be degraded after a
`second esterase activation or a phosphodiester cleavage to yield the
`nucleotide AZTMP 9 (Figure 7). The mechanism of delivery has been
`studied using methylphosphonate as model compound(42). Obviously,
`these compounds are also tripartate prodrugs.
`
`S. Freeman et al. applied their approach to the delivery of AZTMP(39,41)
`and obtained in vitro antiviral activity against HIV-1 and SIV that was
`comparable to AZT 2. Unfortunately, she has not tested the compounds
`in thymidine kinase-deficient cells which could prove the direct
`intracellular delivery of AZTMP 9. Recently, S. Freeman applied their
`bis(AB) approach to the intracellular delivery of foscarnet (PFA) from
`various precursors(43). A possible limitation of the approach is the
`generation of the reactive benzylcation 28 in close vicinity of the active
`site of the enzyme because such cations are known to be potential
`alkylating agents that could react with side chains of amino acids(44).
`This possible problem was faced by the approach reported by A. Glazier
`using the bis(AB) derivatives 25 (Figure 7)(45). Although the basic
`principle of the nucleotide delivery from 25 is essentially the same
`(carboxyesterase-mediated activation to give 32 via 31) as from triesters
`24, the difference to the Freeman concept is the introduction of a
`methylmethoxycarbonyl group in the benzylic position of the AB
`residue. The rationale for this substitution at the benzylic position is the
`possibility of a fast elimination reaction via
`the fleeting 4'-
`quinonemethide 33 due to the electron-withdrawing methoxycarbonyl
`group that facilitates proton abstraction by decreasing the intrinsic
`barrier to proton removal. The transition state for proton removal is
`further stabilized by resonance delocalisation into the aromatic ring. The
`adjacent carbonyl group may also facilitate proton tautomerisation by
`acting by intramolecular general base catalysis. Firstly, this concept has
`bis(4'-acetoxy-α-methylmethoxycarbonyl-
`been
`proven
`with
`
`5
`
`

`

`238
`
`C. Meier
`
`SYNLETT
`
`This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.
`
`J. Balzarini et al. showed that the corresponding ddA 6 and d4A
`derivatives (shown in Figure 8) markedly increased the antiviral
`potency of the parent nucleosides in different cell lines(50). The most
`obvious interpretation of these data is the efficient delivery of the
`appropriate nucleotides ddAMP 8 and d4AMP, respectively, which
`leads to the ADA-bypass. It should be added that for the ddA and the
`d4A phosphoramidate derivatives an increase in the selectivity index
`compared to the free nucleosides was observed. Furthermore, both
`adenosine analogues were efficient as 3TC in inhibiting hepatitis-B
`virus replication in hepatocytes. As a consequence, these derivatives
`rank among the most potent HIV and HBV inhibitors reported so far in
`cell culture.
`
`Amino Acid Phosphoramidate Diesters
`Recently, C. Wagner et al. published an approach using
`phosphoramidates 39 bearing aromatic amino acids that are still
`negatively charged(58). The approach has been applied
`to
`the
`nucleosides d4T 1(58), AZT 2(59), 5-FdU 3(60), and 3'-fluoro-3'-
`deoxythymidine (FLT)(58) (Figure 9). In few cases the reported
`phosphoramidates exhibited greater potency than the parent nucleosides
`in vitro. As amino acid L-tryptophan was found to be the best choice: in
`vitro, the L-tryptophan-AZT phosphoramidate demonstrated a 8-fold
`increase in antiviral activity compared to free AZT 2 against HIV-1
`replication in peripheral blood mononuclear cells without toxicity(59).
`Furthermore, cells treated with the active AZT diester contained 4-fold
`more phosphorylated AZT than those treated only with AZT(59). Again,
`this high biological activity could not be transferred to the antitumor-
`active nucleoside 5-FdU 3. Evaluation of the corresponding L-
`tryptophan-5-FdU phosphoramidate demonstrated a pronounced
`decrease in antitumor activity of the prodrug compared to free 5-FdU in
`several cell lines. Although some of the activity was gained back by
`using permeabilized cells, even in these cells the activity was still lower
`than the activity of the parent nucleoside 3 in intact cells. However,
`incubation of cell-free extracts of CEM cells with radiolabelled L-
`tryptophan-5-FdU phosphoramidate resulted in the rapid production of
`5-FdUMP and a lag in the generation of 5-FdU 3(60). It should be added
`that this approach is the only reported case that successfully uses
`phosphodiesters as a prodrug concept. It has been shown that due to the
`remaining charge, the compounds are still considerably water soluble
`and furthermore, the compounds are indefinitely stable in human blood.
`The high stability was also obser

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