Abstract
Phosphoryl prodrugs are key compounds in drug development. Biologically active phosphoryl compounds often have negative charges on the phosphoryl group, and as a result, frequently have poor pharmacokinetic (PK) profiles. The use of lipophilic moieties bonded to the phosphorus (or attached oxygen atoms) masks the negative charge of the phosphoryl group, cleavage releasing the active molecule. The use of prodrugs to improve the PK of active parent molecules is an essential step in drug development. This review highlights promising trends in terminal elimination half-life, Cmax, clearance, oral bioavailability, and cLogP in phosphoryl prodrugs. We focus on specific prodrug families: esters, amidates, and ProTides. We conclude that moderating lipophilicity is a key part of prodrug success. This type of evaluation is important for drug development, regardless of clinical application. It is our hope that this analysis, and future ones like it, will play a significant role in prodrug evolution.
Introduction
Phosphoryl moieties are commonly found in metabolic pathways ranging from isoprenoids to nucleotides.[1] Often, the active molecule will bear one or more negative charge at biological pH, and, as a result, this negative charge can inhibit uptake into cells and tissues, resulting in a poor PK profile, even if the molecule is shown to be impressively potent against the target[1, 2]. The use of prodrugs, a strategy to side-step use of the active/parent compounds, was first popularized by Adrien Albert in 1958[3]. Prodrugs are defined as inactive molecules that, when subjected to metabolism, are converted into the active molecule.[3] The goal of phosphoryl prodrugs is to mask the negative charge found on the active compound by increasing the lipophilicity of the molecule—ultimately improving the PK profile. Evaluation of the PK profile is typified by following parameters such as oral bioavailability, terminal elimination half-life, plasma Cmax, plasma clearance, and cLogP[4].
This review strives to compile and highlight PK data of a variety of phosphoryl prodrugs, illuminating the impact of the prodrug moiety on PK. Interestingly, but not surprising, these prodrugs have found a variety of uses, including the treatment of hepatitis B, diabetes mellitus, and several types of cancers. Figure 1 shows the general structures of the prodrugs discussed. The families of interest are the esters, amidates, and ProTides. Wherever possible, we discuss human PK data. If not available, we rely on primate or other higher order mammalian data for comparisons. With a history in phosphoryl—specifically phosphonate—chemistry([5–9]). We hope this review will serve as a guide to those interested in phosphoryl prodrugs, as well as those, like us, who are in pursuit of compounds with improved PK characteristics. Thus, this endeavor aims to aid in synthetic decision making. All cLogP values were calculated using the Consensus cLogP from SwissADME[10].
Figure 1.

Structures of the phosphonate prodrugs discussed: esters, amidates, and ProTides.
Esters
The ester family of prodrugs is comprised of molecules where the prodrug has phosphor(mono)ester or phosphodiester bonds connecting the pro-moiety to the rest of the molecule (Figure 1). In this section, we chose to examine PK data for Adefovir, Tenofovir, and Cidofovir (Table 1). These parent compounds have extensive PK data available in the literature, and researchers have taken great interest in improving the PK profile through prodrugs. The use of POM (pivaloyloxymethyl) and POC (propargyloxycarbonyl) groups (Figure 2) are popular and simple lipophilic moieties that produce profound changes in the PK parameters of the drugs, as highlighted in this review. Cidofovir, however, has a monoester prodrug Brincidofovir. Brincidofovir is a mono-alkoxyalkyl phosphoester with equally promising improvements in the PK profile. The PK data compiled for each parent compound and prodrug of the mono- and diesters is summarized in Table 1.
Table 1.
Mono and Diesters
| Compound | Structure | Half-lifea | Cmaxb | Clearancec | cLog Pd | % Fe |
|---|---|---|---|---|---|---|
| Adefovir |
|
38±15 hr[11] (HepG2 cells) | 0.28±0.33 L/kg[12] (Monkeys, PO) | −1.29 | 15.9±11.5%[13] (mice) 4.0%[12] (Monkeys) |
|
| Adefovir Dipivoxil |
|
< 5 mins[14] (Human serum) | 0.35±0.15 μg-eq/mL[15] (Monkeys, PO) | 1.77 | 52.8±11.4%[13] (Mice) 22.2±15.6[15] (Monkeys) |
|
| Tenofovir (TFV) |
|
6.64 hr[16] (Humans, IV) 7.31 hr [17] (Dogs, PO) |
2.50 μg/mL [16] (Humans, IV) 1.29 μg/mL [17] (Dogs, PO) |
217±55.8 mL/hr/kg [16] (Humans, IV) 0.28 6 0.05 liter/h/kg[17] (Dogs, PO) |
−1.01 | 18.5±6.79 %[17] (Dogs) |
| Tenofovir Bis-POM |
|
14.8±2.9[18] hr (dogs, PO) <10mins[19] (Dog plasma, PO) |
2.7±0.6[18] μg/mL (Dogs, PO) | 2.03 | 37.8±5.1[18] (Dogs) | |
| Tenofovir Bis-POC (Disoproxil Fumarate) |
|
12–13 hr[20] (Fasted Humans, PO) | 111 μg/L[20] (Fasting Humans, PO) | 247 mL/hr [20] (Fasted H uman, PO) | 1.49 | 20%[21] (Mice) 31% [22] (Monkeys) 25% (TFV)[20] (Fasted humans) |
| Cidofovir (CDV) |
|
63.0±11.0 hr[23] (Humans, PO) | 31.1±7.00 ng/mL[23] (Humans, PO) | 148±20 mL/hr/kg[24] (Humans, PO) | −2.01 |
2.2%[25] (Rats) <5.3±2.9%[24] (HIV patients) |
| Brincidofovir (BCV) |
|
24.0±0.705 hr[23] (Humans, PO) | 350±119 ng/mL[23] (Humans, PO) | 3.94 | 88%[26] (Mice) |
Plasma half-life: time required for the concentration of the drug in the plasma to be reduced to 50%,
Cmax: maximum concentration of the drug in plasma during a dosing interval,
clearance: renal clearance rate of drug,
cLogP: calculated partition coefficient,
systemic bioavailability in % after oral administration. [27]
Figure 2.

Molecular structure of Bis-POC and Bis-POM groups
Adefovir
Developed by Gilead Sciences, Adefovir is a reverse transcriptase inhibitor used to treat chronic hepatitis B. The active metabolite, Adefovir diphosphate, is formed after the parent compound Adefovir is phosphorylated twice [28]. The structure of Adefovir is an adenine analog with an ethyloxymethyl linker to the phosphoryl moiety. The PK profile of Adefovir is unfavorable. Despite a 38 hour half-life[11], the Cmax and oral bioavailability are only 0.28 L/kg and 4.0%, respectively, in monkeys[12]. Adefovir has a very low cLogP of −1.29.
The parent compound Adefovir was transformed to the bis-POM analog, Adefovir Dipivoxil[29]. The structure of Adefovir Dipivoxil is the identical to adefovir, except that the diacid has been esterified to create two pivaloyloxymethyl (POM) esters. This prodrug displays significant changes in the PK profile. Adefovir Dipivoxil has a half-life in human serum of less than 5 minutes[14], but a Cmax and oral bioavailability improvement to 0.35 μg-eq/mL and 22.2%, respectively, in monkeys[15]. The improvements in Cmax and oral bioavailability are correlated with a significant increase in cLogP from −1.29 to 1.77. The increase in lipophilicity likely contributes to the marked increase in these two PK parameters. Adefovir Dipivoxyl is quickly cleaved on first-pass metabolism in the intestines and the liver to form Adefovir. Any intact Adefovir Dipivoxyl that reaches cells is readily absorbed, rapidly cleaved, and ultimately phosphorylated twice to produce the active metabolite[15]. Optimization of Adefovir Dipivoxyl would allow for an increased plasma half-life and Cmax, which would ultimately allow for increased absorption and conversion to the active metabolite by cells. Despite Adefovir Dipivoxyl having extraordinarily lipophilic POM groups to promote cellular penetration, one drawback to the use of POM groups is how readily they are cleaved by esterases during metabolism[30].
Tenofovir
The reverse transcriptase inhibitor Tenofovir is used to treat chronic hepatitis B as well as treat and prevent AIDS/HIV[31]. Tenofovir is a nucleoside analog of adenine with an aliphatic hydrocarbon chain linking the nucleoside to the diacid phosphoryl group. The parent compound Tenofovir was found by Deeks et al. to have a half-life of 6.64±2.1 hours, a Cmax of 2.50 μg/mL, and a clearance of 217 mL/hr/kg in humans[16]. It was found to have an oral bioavailability of 17.1±1.88 % in dogs[17]. The cLogP of Tenofovir is −1.01.
The first Tenofovir prodrug we consider is Tenofovir bis-POM, or Tenofovir Dipivoxil. The modification to the structure of the parent drug to reach this prodrug was the esterification of the diacid to create a pivaloyloxymethyl ester. The bis-POM prodrug had a great increase in lipophilicity with a cLogP of 2.03 compared to the parent compound’s value of −1.01. In dogs, Tenofovir Dipivoxil had a half-life of 14.8 hours, resulted in a Cmax of Tenofovir of 2.7 μg/mL, and an oral bioavailability of Tenofovir of 37.8%[18]. Despite esterases being found throughout the blood and tissues of dogs, the highest esterase activity was found to be in the liver. This indicates that the liver may be the primary location of prodrug cleavage[18]. As a result, a drug with a longer half-life and greater Cmax value would be greatly beneficial to prevent all of the prodrug being eliminated in the liver before being distributed throughout the body, as the prodrug is more likely to penetrate cells. Intracellular enzymatic cleavage to the diacid followed by two phosphorylation events would result in the active metabolite formation[30].
A second Tenofovir prodrug is Tenofovir bis-POC, or Tenofovir Disoproxil Fumarate (TDF). The structure of TDF is the same as tenofovir, but instead of the diacid, the molecule is esterified to give the propargyloxycarbonyl (POC) diester. Dosing with TDF resulted in a 12–13-hour half-life of the parent compound Tenofovir. The Cmax was found to be 111 μg/mL of Tenofovir after dosing with TDF in humans, and Tenofovir clearance was found to be 247 mL/hr after a single dose of TDF. Finally, in a fasted state, oral bioavailability was determined to be 25%[20]. Similar to Tenofovir Dipivoxil, TDF had a great increase in lipophilicity with a cLogP of 1.49. Preliminary in vitro studies found that TDF was metabolized first by non-specific carboxylesterase enzymes found in the blood, organs, and tissues with the monoester as the major circulating metabolite[20]. A long half-life and high Cmax are found in human serum, which are essential in increasing the effectiveness of the active metabolite. Ultimately, any circulating TDF, monoester, or TFV would enter the cells, and the esters, if present, would be fully cleaved. The TFV is then phosphorylated twice to produce the active metabolite. Not only was TDF found to have a favorable half-life and Cmax in the serum, but was also found to have a long intracellular half-life[32]. A long intracellular half-life is favorable as HIV inhibition will remain high long after drug administration is discontinued.
Cidofovir
Cidofovir (CDV) is a broad-spectrum antiviral agent used against herpesviruses, papillomaviruses, and poxviruses[33]. The structure of cidofovir is that of a cytosine with an aliphatic linker with a hydroxymethyl group on the second carbon of the linker. The cytosine is linked to the phosphonic acid moiety. In humans, Cidofovir was found to have a half-life of 63 hours, a Cmax of 31.1 ng/mL, a clearance of 148 mL/hr/kg, an oral bioavailability of <5%, and a very low cLogP of −2.01[23, 34]. The prodrug of interest for Cidofovir is Brincidofovir. The modification to the structure is addition of a lipid alkoxyalkyl ester. This prodrug was found to have a half-life 24 hours and a Cmax of 150 μM, as well as an oral bioavailability of 88% and a cLogP of 3.94[23, 26]. This impressive change in bioavailability is even more impressive due to the fact that Brincidofovir is a monoester, compared with the diester prodrugs above. BVC is taken into cells using endogenous lipid uptake pathways where BVC resembles the naturally occurring lipid lysophosphatidylcholine. Once inside the cell, the lipid moiety can be cleaved by esterases to release CDV which is phosphorylated twice to produce the active metabolite. A high serum half-life and Cmax is advantageous with BCV as cleavage of BCV in the serum will result in the potentially nephrotoxic CDV [35]. Therefore, BCV is able to accumulate in high concentrations, remaining in the body long enough to be transported into the cells where cleavage and phosphorylation can occur.
Amidates
Another option for masking the negative charges of the phosphoryl group is to use phosphoramide bonds, usually where the phosphorus atom is bonded to an amino acid (Table 2). Bisamidates have not been as popular as esters (or ProTides, discussed below) as a prodrug strategy. However, it is a powerful technique when applied to particular molecules and has been shown to have lower toxicity and higher plasma stability while still masking the charged phosphonate, similar to esters or ProTides[36].
Table 2.
Bisamidates
| Compound | Structure | Half-life | Cmax | Clearance | cLog P | % F |
|---|---|---|---|---|---|---|
| Adefovir |
|
1.6±0.5 hr[37] (Humans, IV) | 10.6±2.27 mg/L[37] (Humans, IV) | 220±52 mL/hr/kg[37] (Humans, IV) | −1.29 | <12%[37] (Humans) |
| GS-9219 |
|
<30 min[38] (Dogs, IV) 0.33±0.20 hr[39] (Dogs, IV) |
4280±1550 nmol/L[39] (Dogs, IV) | 2.25±0.23 L/hr/kg[38] (Dogs, IV) | 1.04 | |
| MB05032 |
|
1.19 | 2%[40] (Rats) | |||
| CS-917 |
|
>7 days [41] pH 3.0–7.4 (t90) | 2.72 | 22%[40] (Rats) |
Adefovir
As previously mentioned, Adefovir is a reverse transcriptase inhibitor used to treat chronic hepatitis B. Developed by Gilead Sciences, the adenine analog has a terminal half-life of 1.6±0.5 hours, a Cmax of 10.6±2.27 mg/L, and a clearance of 220±52 mL/hr/kg in humans. Adefovir has a low cLog P value of −1.29 which is accompanied by a low oral bioavailability of <12% in humans[37]. A bisamidate prodrug of an Adefovir analog is GS-9219. Also developed by Gilead, this prodrug is Adefovir with an N6-cyclopropyl group and a phosphoryl group added to the primary alcohol. The phosphoryl group has two amide bonds, each bonded to an ethyl ester of (L)-alanine. It was found that in dogs, GS-9219 had a half-life of about 30 minutes, a Cmax of 4280±1550 nmol/L, and a clearance of 2.25±0.23 L/hr/kg. The cLog P is 1.04. With such a great increase in lipophilicity, we would expect to also see an overall trend of increasing oral bioavailability. The clearance of GS-9219 was relatively high, however. For any GS-9219 that did penetrate the cells, the molecule was quickly hydrolyzed to cPrPMEDAP and then deaminated to PMEG. Phosphorylation of PMEG twice results in the active metabolite PMEGpp[38]. The high clearance and short half-life of GS-9219 are concerning. Optimization of GS-9219 would seek to maintain lipophilicity and Cmax, but increase the half-life and decrease the clearance rate so that GS-9219 has more time to penetrate cells.
MB05032
MB05032 and its prodrug CS-917 are used to inhibit fructose 1,6-bisphosphatase, an enzyme that is crucial for gluconeogenesis. Therefore, inhibition of fructose 1,6-bisphosphatase is an attractive enzymatic target for small molecule treatment of type 2 diabetes mellitus[41]. The structure of MB05032 is comprised of a thiazole ring with 2-amino and 5-methylpropyl groups branching and connected to the phosphonic acid through a furanyl ring. MB05032 has extremely low oral bioavailability in rats of only 2% when dosed at 30 mg/kg, and the polar parent molecule has a cLogP value of 1.19. CS-917 is a bisamidate of MB05032 bearing two phosphoramidate substituents: ethyl esters of L-alanine. Prodrug CS-917 displays an 11-fold increase in bioavailability, up to 22%. Such a large jump in bioavailability is expected as the cLogP increased by roughly 1.5 log units, to 2.72. In addition, CS-917 was found to be extremely stable at pH2.5–7 at 37°C with a t90 of greater than 7 days. Erion et al. also found that the ethyl esters of L-alanine are quickly cleaved from the phosphoryl group in liver s9 fractions of rats and humans, but did not report quantitative values for this experiment[40, 41]. Although half-life, Cmax, and clearance data on CS-917 and its parent MB05032 are limited, the increase in lipophilicity of CS-917 allows for greater penetration into cells as shown by the increased bioavailability. Once inside the cell, the prodrug moieties can be cleaved to produce the active metabolite MB05032 to inhibit fructose 1,6-bisphosphatase[41].
ProTides
ProTides were first invented and launched to stardom by Christopher McGuigan from Cardiff University in Wales[42]. The name comes from the structure—a prodrug form of a nucleotide. ProTide prodrugs consist of a mixed ester and phosphoryl amidate typically using an aryloxy ester and an esterified amino acid[42]. ProTides were found to be an extremely effective prodrug strategy and have resulted in several refined ProTides becoming FDA approved, some of which will be explored in this review[43, 44]. Sofosbuvir and Tenofovir Alafenamide are two FDA approved ProTides. These molecules were chosen to discuss because of their prevalence in medicine, and because of the amount of PK data available. More recently, research groups are seeking to make ProTide versions of active drugs in order to improve the PK profile. The data is summarized in Table 3.
Table 3.
ProTides
| Compound | Structure | Half-life | Cmax | Clearance | cLog P | % F |
|---|---|---|---|---|---|---|
| Tenofovir (TFV) |
|
6.64 hr[16] (Humans, IV) 7.31hr[17] (Dogs, PO) |
2.50 μg/mL [16] (Humans, IV) 1.29 μg/mL[17] (Dogs, PO) |
217±55.8 mL/hr/kg [16] (Humans, IV) 0.28 6 0.05 liter/h/kg[17] (Dogs, PO) |
−1.01 | 18.5±6.79 %[17] (Dogs) |
| Tenofovir Alafenamide (TAF) |
|
35.95 hr[45] (Humans, as TVF, PO) | 5.74±0.62 μM[46] (Dogs, PO) | 1.79 | 17% of TVF and >70% of TAF[47] (Dogs) | |
| PSI-6130 |
|
5.64±1.13 hr[48] (Monkeys, PO) | 9.64±1.13 μM[48] (Monkeys, PO) | −0.59 | 24.0%±14.3[48] (Monkey) | |
| PSI 6206 |
|
|||||
| Sofosbuvir (PSI7977) |
|
0.4 hr[49] (Humans, PO) | 622±56.1 ng/mL[49] (Humans, PO) | 1.24 | 9.89 %[50] (Dogs) | |
| Gemcitabine |
|
32.72±2.28 min[51] (Mice) 13.4±4.0 min[52] (Humans, IV) |
2.3 ng/mL [53] (Humans, IV) 124μM[54] (Humans, IV) |
1.173±0.059 mL/min[51] (Mice, IV) | −0.64 | 10%[53] (Human) |
| Acelarin |
|
139 min[55] (Human hepatocytes) 8.3 hr[54] (Humans, IV) |
26,910 μM[54] (Humans, IV) | 1.79 |
Tenofovir
As described above, Tenofovir is a nucleoside analog of adenine used to treat chronic hepatitis B and treat/prevent AIDS and HIV. Tenofovir is the parent compound of the Tenofovir ProTide: Tenofovir Alafenamide Fumarate (TAF). TAF has the same structure as tenofovir except that it has been prodrugged by the modification of the phosphonic acid to a phosphoramidate. The phosphoramidate consists of a phenoxy ester and an amide bond to L-alanine that has been esterified with isopropanol. TAF is used to treat chronic Hepatitis B and HIV by acting as a reverse transcriptase inhibitor[45, 56]. Cundy et al. found that, in dogs at 10 mg/kg, Tenofovir has a half-life of 7 hours in plasma, a Cmax of 1 μg/mL, plasma clearance of 0.28 L/hr/kg, and oral bioavailability of 17%[17]. As mentioned previously, Tenofovir has a cLog P of −1.01. The cLog P for TAF jumps to 1.79. Such an increase in lipophilicity was accompanied by a plasma half-life of TAF of 0.30 hours, a Cmax of 4.51 μM, and resulted in an oral bioavailability of greater than 70% for TAF[56]. TAF metabolism is unique. Once the lipophilic TAF has been absorbed by the intestines and eventually goes from the blood to cells, the prodrug is hydrolyzed by cathepsin A (CatA) or carboxylesterase 1 to produce TFV-Alanine. The molecule is then further hydrolyzed in lysosomes to produce TFV. TFV is finally phosphorylated twice to produce the active metabolite TFV-DP[57]. TAF has a long half-life and relatively high Cmax which affords high oral bioavailability and time for TAF to penetrate cells and continue on to the active metabolite.
PSI-6130
The compound PSI-6130 is the parent compound of PSI-6206. PSI-6130 was found to be readily deaminated to PSI-6206. A prodrug of PSI-6206 is the clinically approved Sofosbuvir. Sofosbuvir, originally discovered by Pharmasett (then acquired by Gilead), is a ProTide used to treat chronic hepatitis C, working as a polymerase inhibitor[58]. PSI-6130, PSI-6202 and Sofosbuvir have fluoro and methyl groups on the 5’ carbon of the deoxyribose ring. However, PSI-6130 has a cytosine while PSI-6206 and Sofosbuvir are uridine analogs. The prodrug Sofosbuvir has a phosphoryl group attached to the hydroxyl of the 5’ carbon containing a phenyl ester as well as an amide of (L)-alanine isopropyl ester. PSI-6130 was dosed in monkeys at 33.3 mg/kg orally and found to have a half-life of 5 hours and a Cmax of 9 μM with a cLog P of −0.59, and oral bioavailability of 24%[48]. On the other hand, Sofosbuvir was found by Wang et al. to have a half-life of 1.9 hours, and a Cmax 0.058 μM in monkeys at 5 mg/kg PO[59]. These changes in half life and Cmax are accompanied by a cLog P increase to 1.24. Once Sofosbuvir is taken into cells, it is first hydrolyzed by human carboxylesterase 1 or CatA to remove the phenoxy moiety of the prodrug. Then, histidine triad nucleotide-binding protein 1 (HINT1) cleaves the amide bond to release the diacid-Sofosbuvir metabolite. Finally, this diacid is phosphorylated twice to produce the active metabolite[60]. The unique metabolic pathway of Sofosbuvir is best supported with long plasma half-life and Cmax to allow for as much time as possible for Sofosbuvir to penetrate cells and begin its breakdown.
Gemcitabine
Gemcitabine is an anti-tumor medication used for many different cancers including ovarian, pancreatic, and bladder cancer[61]. This parent compound is an analog of deoxycytidine with two fluorine atoms attached to the 2’-carbon of the deoxyribose ring. Gemcitabine is typically administered intravenously, and the half-life and Cmax values shown in Table 3 are from intravenous administration. From a study in which Gemcitabine was administered orally, oral bioavailability was reported as 10%[53]. Gemcitabine is not particularly lipophilic with a cLog P of −0.64. It also has a short half-life of 13 minutes[52] and Cmax of 64 μmol/L[54]. Comparatively, the prodrug Acelarin is much more lipophilic. This prodrug has the same structure as Gemcitabine except that it has a prodrugged phosphoramide group attached. This ProTide is comprised of a phenyl ester and an amide bond to (L)-alanine benzyl ester. The prodrug moiety is attached to the 5’-carbon of the deoxycytidine ring by a phosphorester bond. The prodrug has a cLog P of 1.79, a half-life of 8.3 hours and a Cmax of 26,910 μM, which is a 217-fold increase in Cmax from gemcitabine[54]. Acelarin is able to avoid the equilibrative nucleoside transporter 1 (hENT1), which is normally how gemcitabine enters the cell, because of its lipophilic nature. Once inside the cell, esterases remove the ester and amide linked moieties of the prodrug to release gemcitabine, which is ultimately phosphorylated twice to release the active metabolite[54]. Acelarin has a relatively long half-life and high Cmax which is advantageous as the prodrug has ample time to enter cells and undergo its unique metabolism to produce the active metabolite.
Concluding Remarks
The compilation of molecules in this review are meant to highlight the PK parameters that are essential to an effective phosphoryl drug. Increasing the lipophilicity of the molecule, as reflected in cLogP, through the use of pro-moieties bound by phosphoester, phosphoramide, or a mixture of phosphoester/phosphoramide (aka ProTide) is key to the drug’s success. Using molecules with a variety of clinical applications has shown the power of phosphoryl prodrugs and cemented their use in drug development. Phosphoryl prodrug design can be continued through the use of this review as it tabulates, and therefore, emphasizes where and how increasing lipophilicity can directly relate to improvements in half-life, Cmax, and oral bioavailability.
References
- 1.Wiemer AJ, Wiemer DF (2015) Prodrugs of phosphonates and phosphates: crossing the membrane barrier. Top Curr Chem 360:115–160 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hecker SJ, Erion MD (2008) Prodrugs of Phosphates and Phosphonates. J Med Chem 51:2328–2345 [DOI] [PubMed] [Google Scholar]
- 3.Albert A (1958) Chemical aspects of selective toxicity. Nature 182:421–422 [DOI] [PubMed] [Google Scholar]
- 4.Toon S (1990) The Assessment of Pharmacokinetics. In: O’Grady J, Linet OI (eds) Early Phase Drug Eval. Man Macmillan Education; UK, London, pp 229–242 [Google Scholar]
- 5.Uh E, Jackson ER, San Jose G, Maddox M, Lee RE, Lee RE, Boshoff HI, Dowd CS (2011) Antibacterial and antitubercular activity of fosmidomycin, FR900098, and their lipophilic analogs. Bioorg Med Chem Lett 21:6973–6976 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jackson ER, San Jose G, Brothers RC, Edelstein EK, Sheldon Z, Haymond A, Johny C, Boshoff HI, Couch RD, Dowd CS (2014) The effect of chain length and unsaturation on Mtb Dxr inhibition and antitubercular killing activity of FR900098 analogs. Bioorg Med Chem Lett 24:649–653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.San Jose G, Jackson ER, Haymond A, et al. (2016) Structure-Activity Relationships of the MEPicides: N-Acyl and O-Linked Analogs of FR900098 as Inhibitors of Dxr from Mycobacterium tuberculosis and Yersinia pestis. ACS Infect Dis 2:923–935 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wang X, Edwards RL, Ball H, et al. (2018) MEPicides: α,β-Unsaturated Fosmidomycin Analogues as DXR Inhibitors against Malaria. J Med Chem 61:8847–8858 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Heidel KM, Dowd CS (2019) Phosphonate prodrugs: an overview and recent advances. Future Med Chem 11:1625–1643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Daina A, Michielin O, Zoete V (2017) SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep 7:42717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.As R, Je V, L O, A F (2004) Effective metabolism and long intracellular half life of the anti-hepatitis B agent adefovir in hepatic cells. Biochem Pharmacol 68:1825–1831 [DOI] [PubMed] [Google Scholar]
- 12.Cundy KC, Shaw JP, Lee WA (1994) Oral, subcutaneous, and intramuscular bioavailabilities of the antiviral nucleotide analog 9-(2-phosphonylmethoxyethyl) adenine in cynomolgus monkeys. Antimicrob Agents Chemother 38:365–368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Naesens L, Balzarini J, Bischofberger N, De Clercq E (1996) Antiretroviral activity and pharmacokinetics in mice of oral bis(pivaloyloxymethyl)-9-(2-phosphonylmethoxyethyl)adenine, the bis(pivaloyloxymethyl) ester prodrug of 9-(2-phosphonylmethoxyethyl)adenine. Antimicrob Agents Chemother 40:22–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Benzaria S, Pélicano H, Johnson R, Maury G, Imbach J-L, Aubertin A-M, Obert G, Gosselin G (1996) Synthesis, in Vitro Antiviral Evaluation, and Stability Studies of Bis(S-acyl-2-thioethyl) Ester Derivatives of 9-[2-(Phosphonomethoxy)ethyl]adenine (PMEA) as Potential PMEA Prodrugs with Improved Oral Bioavailability. J Med Chem 39:4958–4965 [DOI] [PubMed] [Google Scholar]
- 15.Cundy KC, Fishback JA, Shaw JP, Lee ML, Soike KF, Visor GC, Lee WA (1994) Oral bioavailability of the antiretroviral agent 9-(2-phosphonylmethoxyethyl)adenine (PMEA) from three formulations of the prodrug bis(pivaloyloxymethyl)-PMEA in fasted male cynomolgus monkeys. Pharm Res 11:839–843 [DOI] [PubMed] [Google Scholar]
- 16.Deeks SG, Barditch-Crovo P, Lietman PS, Hwang F, Cundy KC, Rooney JF, Hellmann NS, Safrin S, Kahn JO (1998) Safety, pharmacokinetics, and antiretroviral activity of intravenous 9-[2-(R)-(Phosphonomethoxy)propyl]adenine, a novel anti-human immunodeficiency virus (HIV) therapy, in HIV-infected adults. Antimicrob Agents Chemother 42:2380–2384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cundy KC, Sueoka C, Lynch GR, Griffin L, Lee WA, Shaw J-P (1998) Pharmacokinetics and Bioavailability of the Anti-Human Immunodeficiency Virus Nucleotide Analog 9-[(R)2-(Phosphonomethoxy)Propyl]Adenine (PMPA) in Dogs. Antimicrob Agents Chemother 42:687–690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shaw JP, Sueoko CM, Oliyai R, Lee WA, Arimilli MN, Kim CU, Cundy KC (1997) Metabolism and pharmacokinetics of novel oral prodrugs of 9-[(R)-2-(phosphonomethoxy)propyl]adenine (PMPA) in dogs. Pharm Res 14:1824–1829 [DOI] [PubMed] [Google Scholar]
- 19.Synthesis, in Vitro Biological Evaluation and Oral Bioavailability of 9-[2-(Phosphonomethoxy)Propyl]Adenine (PMPA) Prodrugs - Dimensions. https://app.dimensions.ai/details/publication/pub.1085722504. Accessed 20 Jan 2021
- 20.Kearney BP, Flaherty JF, Shah J (2004) Tenofovir disoproxil fumarate: clinical pharmacology and pharmacokinetics. Clin Pharmacokinet 43:595–612 [DOI] [PubMed] [Google Scholar]
- 21.Naesens L, Bischofberger N, Augustijns P, Annaert P, Van den Mooter G, Arimilli MN, Kim CU, De Clercq E (1998) Antiretroviral Efficacy and Pharmacokinetics of Oral Bis(isopropyloxycarbonyloxymethyl)9-(2-Phosphonylmethoxypropyl)adenine in Mice. Antimicrob Agents Chemother 42:1568–1573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Durand-Gasselin L, Van Rompay KKA, Vela JE, Henne IN, Lee WA, Rhodes GR, Ray AS (2009) Nucleotide analogue prodrug tenofovir disoproxil enhances lymphoid cell loading following oral administration in monkeys. Mol Pharm 6:1145–1151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Painter W, Robertson A, Trost LC, Godkin S, Lampert B, Painter G (2012) First pharmacokinetic and safety study in humans of the novel lipid antiviral conjugate CMX001, a broad-spectrum oral drug active against double-stranded DNA viruses. Antimicrob Agents Chemother 56:2726–2734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cundy KC (1999) Clinical Pharmacokinetics of the Antiviral Nucleotide Analogues Cidofovir and Adefovir. Clin Pharmacokinet 36:127–143 [DOI] [PubMed] [Google Scholar]
- 25.Eriksson†‡ Ulrika, Peterson† Larryn W., Kashemirov† Boris A., Hilfinger§ John M., Drach‡∥ John C., Borysko∥ Katherine Z., Breitenbach∥ Julie M., Kim§ Jae Seung, Mitchell§ Stefanie, Kijek§ Paul, and McKenna*† Charles E. Serine Peptide Phosphoester Prodrugs of Cyclic Cidofovir: Synthesis, Transport, and Antiviral Activity | Molecular Pharmaceutics. https://pubs.acs.org/doi/10.1021/mp8000099. Accessed 19 Jan 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ciesla SL, Trahan J, Wan WB, Beadle JR, Aldern KA, Painter GR, Hostetler KY (2003) Esterification of cidofovir with alkoxyalkanols increases oral bioavailability and diminishes drug accumulation in kidney. Antiviral Res 59:163–171 [DOI] [PubMed] [Google Scholar]
- 27.Pharmacokinetic terms: symbols and units - Baggot - 2001 - Journal of Veterinary Pharmacology and Therapeutics - Wiley Online Library. https://onlinelibrary.wiley.com/doi/pdf/10.1046/j.1365-2885.2001.00340.x. Accessed 11 Jun 2021 [DOI] [PubMed] [Google Scholar]
- 28.Marcellin P, Chang T-T, Lim SG, et al. (2003) Adefovir dipivoxil for the treatment of hepatitis B e antigen-positive chronic hepatitis B. N Engl J Med 348:808–816 [DOI] [PubMed] [Google Scholar]
- 29.Jones DJ, O’Leary EM, O’Sullivan TP (2019) An improved synthesis of adefovir and related analogues. Beilstein J Org Chem 15:801–810 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Schultz C (2003) Prodrugs of biologically active phosphate esters. Bioorg Med Chem 11:885–898 [DOI] [PubMed] [Google Scholar]
- 31.Wassner C, Bradley N, Lee Y (2020) A Review and Clinical Understanding of Tenofovir: Tenofovir Disoproxil Fumarate versus Tenofovir Alafenamide. J Int Assoc Provid AIDS Care JIAPAC 19:2325958220919231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Anderson PL, Kiser JJ, Gardner EM, Rower JE, Meditz A, Grant RM (2011) Pharmacological considerations for tenofovir and emtricitabine to prevent HIV infection. J Antimicrob Chemother 66:240–250 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Andrei G, Snoeck R (2010) Cidofovir Activity against Poxvirus Infections. Viruses 2:2803–2830 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Buller RM, Owens G, Schriewer J, Melman L, Beadle JR, Hostetler KY (2004) Efficacy of oral active ether lipid analogs of cidofovir in a lethal mousepox model. Virology 318:474–481 [DOI] [PubMed] [Google Scholar]
- 35.Chittick G, Morrison M, Brundage T, Nichols WG (2017) Short-term clinical safety profile of brincidofovir: A favorable benefit-risk proposition in the treatment of smallpox. Antiviral Res 143:269–277 [DOI] [PubMed] [Google Scholar]
- 36.Česnek M, Jansa P, Šmídková M, Mertlíková-Kaiserová H, Dračínský M, Brust TF, Pávek P, Trejtnar F, Watts VJ, Janeba Z (2015) Bisamidate Prodrugs of 2-Substituted 9-[2-(Phosphonomethoxy)ethyl]adenine (PMEA, adefovir) as Selective Inhibitors of Adenylate Cyclase Toxin from Bordetella pertussis. ChemMedChem 10:1351–1364 [DOI] [PubMed] [Google Scholar]
- 37.Cundy KC, Barditch-Crovo P, Walker RE, Collier AC, Ebeling D, Toole J, Jaffe HS (1995) Clinical pharmacokinetics of adefovir in human immunodeficiency virus type 1-infected patients. Antimicrob Agents Chemother 39:2401–2405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Reiser Hans 1, Wang Jianying, Lee Chong, Watkins William J, Ray Adrian S, Shibata Riri, Birkus Gabriel, Cihlar Tomas, Wu Sylvia, Li Bei, Liu Xiaohong, Henne Ilana N, Wolfgang Grushenka H I, Desai Manoj, Rhodes Gerald R, Fridland Arnold, Lee William A, Plunkett William, Vail David, Thamm Douglas H, Jeraj Robert, Tumas Daniel B GS-9219--a novel acyclic nucleotide analogue with potent antineoplastic activity in dogs with spontaneous non-Hodgkin’s lymphoma - PubMed. https://pubmed.ncbi.nlm.nih.gov/18451250/. Accessed 28 Jan 2021 [DOI] [PubMed]
- 39.Vail DM, Thamm DH, Reiser H, et al. (2009) Assessment of GS-9219 in a pet dog model of non-Hodgkin’s lymphoma. Clin Cancer Res Off J Am Assoc Cancer Res 15:3503–3510 [DOI] [PubMed] [Google Scholar]
- 40.Dang Q, Kasibhatla SR, Reddy KR, et al. (2007) Discovery of Potent and Specific Fructose-1,6-Bisphosphatase Inhibitors and a Series of Orally-Bioavailable Phosphoramidase-Sensitive Prodrugs for the Treatment of Type 2 Diabetes. J Am Chem Soc 129:15491–15502 [DOI] [PubMed] [Google Scholar]
- 41.Erion MD, van Poelje PD, Dang Q, Kasibhatla SR, Potter SC, Reddy MR, Reddy KR, Jiang T, Lipscomb WN (2005) MB06322 (CS-917): A potent and selective inhibitor of fructose 1,6-bisphosphatase for controlling gluconeogenesis in type 2 diabetes. Proc Natl Acad Sci U S A 102:7970–7975 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mehellou Y, Balzarini J, McGuigan C (2009) Aryloxy phosphoramidate triesters: a technology for delivering monophosphorylated nucleosides and sugars into cells. ChemMedChem 4:1779–1791 [DOI] [PubMed] [Google Scholar]
- 43.Jordheim LP, Durantel D, Zoulim F, Dumontet C (2013) Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. Nat Rev Drug Discov 12:447–464 [DOI] [PubMed] [Google Scholar]
- 44.De Clercq E, Li G (2016) Approved Antiviral Drugs over the Past 50 Years. Clin Microbiol Rev 29:695–747 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ruane PJ, DeJesus E, Berger D, et al. (2013) Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of tenofovir alafenamide as 10-day monotherapy in HIV-1-positive adults. J Acquir Immune Defic Syndr 1999 63:449–455 [DOI] [PubMed] [Google Scholar]
- 46.Babusis D, Phan TK, Lee WA, Watkins WJ, Ray AS (2013) Mechanism for effective lymphoid cell and tissue loading following oral administration of nucleotide prodrug GS-7340. Mol Pharm 10:459–466 [DOI] [PubMed] [Google Scholar]
- 47.Lee WA, He G-X, Eisenberg E, Cihlar T, Swaminathan S, Mulato A, Cundy KC (2005) Selective Intracellular Activation of a Novel Prodrug of the Human Immunodeficiency Virus Reverse Transcriptase Inhibitor Tenofovir Leads to Preferential Distribution and Accumulation in Lymphatic Tissue. Antimicrob Agents Chemother 49:1898–1906 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Asif G, Hurwitz SJ, Shi J, Hernandez-Santiago BI, Schinazi RF (2007) Pharmacokinetics of the antiviral agent beta-D-2’-deoxy-2’-fluoro-2’-C-methylcytidine in rhesus monkeys. Antimicrob Agents Chemother 51:2877–2882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kirby BJ, Symonds WT, Kearney BP, Mathias AA (2015) Pharmacokinetic, Pharmacodynamic, and Drug-Interaction Profile of the Hepatitis C Virus NS5B Polymerase Inhibitor Sofosbuvir. Clin Pharmacokinet 54:677–690 [DOI] [PubMed] [Google Scholar]
- 50.Babusis D, Curry MP, Denning J, Wang T, Kirby B, Murakami E,, et al. (2014) Translational studies to understand the mechanism of liver delivery by sofosbuvir [oral presentation]. [Google Scholar]
- 51.Immordino ML, Brusa P, Rocco F, Arpicco S, Ceruti M, Cattel L (2004) Preparation, characterization, cytotoxicity and pharmacokinetics of liposomes containing lipophilic gemcitabine prodrugs. J Control Release Off J Control Release Soc 100:331–346 [DOI] [PubMed] [Google Scholar]
- 52.Reid JM, Qu W, Safgren SL, Ames MM, Krailo MD, Seibel NL, Kuttesch J, Holcenberg J (2004) Phase I trial and pharmacokinetics of gemcitabine in children with advanced solid tumors. J Clin Oncol Off J Am Soc Clin Oncol 22:2445–2451 [DOI] [PubMed] [Google Scholar]
- 53.Veltkamp SA, Jansen RS, Callies S, et al. (2008) Oral Administration of Gemcitabine in Patients with Refractory Tumors: A Clinical and Pharmacologic Study. Clin Cancer Res 14:3477. [DOI] [PubMed] [Google Scholar]
- 54.Blagden SP, Rizzuto I, Suppiah P, et al. (2018) Anti-tumour activity of a first-in-class agent NUC-1031 in patients with advanced cancer: results of a phase I study. Br J Cancer 119:815–822 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Slusarczyk M, Lopez MH, Balzarini J, Mason M, Jiang WG, Blagden S, Thompson E, Ghazaly E, McGuigan C (2014) Application of ProTide technology to gemcitabine: a successful approach to overcome the key cancer resistance mechanisms leads to a new agent (NUC-1031) in clinical development. J Med Chem 57:1531–1542 [DOI] [PubMed] [Google Scholar]
- 56.Murakami E, Wang T, Park Y, Hao J, Lepist E-I, Babusis D, Ray AS (2015) Implications of Efficient Hepatic Delivery by Tenofovir Alafenamide (GS-7340) for Hepatitis B Virus Therapy. Antimicrob Agents Chemother 59:3563–3569 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Birkus G, Bam RA, Willkom M, Frey CR, Tsai L, Stray KM, Yant SR, Cihlar T (2015) Intracellular Activation of Tenofovir Alafenamide and the Effect of Viral and Host Protease Inhibitors. Antimicrob Agents Chemother 60:316–322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Keating GM (2014) Sofosbuvir: a review of its use in patients with chronic hepatitis C. Drugs 74:1127–1146 [DOI] [PubMed] [Google Scholar]
- 59.Wang T, Babusis D, Park Y, et al. (2020) Species differences in liver accumulation and metabolism of nucleotide prodrug sofosbuvir. Drug Metab Pharmacokinet 35:334–340 [DOI] [PubMed] [Google Scholar]
- 60.Denning J, Cornpropst M, Flach SD, Berrey MM, Symonds WT (2013) Pharmacokinetics, Safety, and Tolerability of GS-9851, a Nucleotide Analog Polymerase Inhibitor for Hepatitis C Virus, following Single Ascending Doses in Healthy Subjects. Antimicrob Agents Chemother 57:1201–1208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.(2017) FDA Approval for Gemcitabine Hydrochloride - National Cancer Institute. https://web.archive.org/web/20170405134518/https://www.cancer.gov/about-cancer/treatment/drugs/fda-gemcitabine-hydrochloride. Accessed 25 Feb 2021
