Quick viewing(Text Mode)

Enzyme-Mediated Hydrolytic Activation of Prodrugs

Enzyme-Mediated Hydrolytic Activation of Prodrugs

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector Acta Pharmaceutica Sinica B 2011;1(3):143–159

Institute of Materia Medica, Chinese Academy of Medical Sciences Chinese Pharmaceutical Association

Acta Pharmaceutica Sinica B

www.elsevier.com/locate/apsb www.sciencedirect.com

REVIEW -mediated hydrolytic activation of prodrugs

Yan-hui Yang, Herve Aloysius, Daigo Inoyama, Yu Chen, Long-qin Hun

Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA

Received 15 July 2011; revised 4 August 2011; accepted 10 August 2011

KEY WORDS Abstract Prodrug design is an important part of drug discovery. Prodrugs can offer many advantages over parent drugs such as increased , enhanced stability, improved bioavail- Prodrugs; Hydrolytic activation; ability, reduced side effects, and better selectivity. Many prodrugs have been used successfully in ; the clinic; examples include oseltamivir in anti-influenza therapy, enalapril in anti-hypertension ; therapy, in cancer therapy, and omeprazole in the treatment of peptic ulcer. A key step in prodrug design is the incorporation of an activation mechanism that can convert the prodrug into the active species in an efficient and/or controlled manner to meet the needs of a given medical application. Prodrug activation can be achieved through enzyme-mediated hydrolytic or oxido- reductive processes while activation of some prodrugs may proceed through pure chemical nonenzymatic processes. This review focuses on the hydrolytic that have been used in prodrug activation, including , , and .

& 2011 Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical Association. Production and hosting by Elsevier B.V. All rights reserved.

nCorresponding author. Tel.: þ1 732 445 5291; fax: þ1 732 445 6312. E-mail address: [email protected] (Long-qin Hu).

2211-3835 & 2011 Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical Association. Production and hosting by Elsevier B.V. All rights reserved.

Peer review under responsibility of Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical Association. doi:10.1016/j.apsb.2011.08.001 144 Yan-hui Yang et al.

1. Introduction specificity of CEs is, in part, determined by con- straints enforced by the dimensions of, and access to, the active With the advance of new technologies such as combinatorial site10. and computational chemistry, more and more compounds are The carboxylesterases are involved in the activation of being identified with extremely potent in vitro activity but are various antiviral, anticancer, and prodrugs. Several found to be inactive in vivo. They may have the optimal representative esterase-activated prodrugs are shown in Fig. 1. configuration and conformation needed to interact with their (1, CPT-11) is converted to its active metabolite, target or enzyme, but they do not necessarily possess 7-ethyl-10-hydroxycamptothecin, which is an inhibitor of the best molecular form and physicochemical properties topoisomerase I8,11; temocapril (2), a long-acting ACE inhibi- needed for their delivery to the site of action. Some of the tor, is quickly metabolized to the active dicarboxylic acid12; problems often encountered include (i) limited solubility and dipivefrin (3), available as an ophthalmic solution for the poor chemical stability preventing the drug from being ade- treatment of glaucoma, is hydrolyzed to epinephrine on quately formulated, (ii) low or variable bioavailability due to penetrating cornea13; lovastatin (4), and simvastain (5), used incomplete absorption across biological membranes or exten- to lower in patients with hypercholesterolemia, sive first-pass , and (iii) lack of site specificity. were hydrolyzed by hCE1 to form the hydroxy acid forms, Prodrugs have been designed and used clinically to solve these which inhibit the activity of HMG-CoA reductase14. problems. A prodrug is inactive or much less active and has to be activated in vivo to form the active drug . 2.2. Acetylcholinesterase Prodrug design has proven to work for many pharmacologi- cally active compounds or drugs in improving their physicochem- Acetylcholinesterase (AChE, EC 3.1.1.7) can rapidly hydro- ical and biological properties and their target selectivity. Currently, lyze the neurotransmitter acetylcholine at cholinergic synapses 5–7% of the drugs approved worldwide can be classified as and neuromuscular junctions. AChE is an extrinsic mem- prodrugs, and approximately 15% of all new drugs approved each brane-bound enzyme that projects into the synapse. It is year are prodrugs1. Prodrugs have been used in a wide variety of responsible for the elimination of ACh released from the therapeutic areas including anti-influenza, anti-hypertension, anti- presynaptic nerve process in response to an action potential biotics, antiulcer, anticoagulant, anti-inflammatory, antifungal, and is critical for maintaining normal cholinergic function15,16. anesthetic, and anticancer. Many prodrugs have gained huge AChE is widely expressed in many tissues throughout the body. clinical success. Activation of prodrugs can involve many enzymes including skeletal muscle, many types of neurons in the central and including like CYP450 and DT-diaphorase as peripheral nervous systems, and numerous other types that are well as hydrolytic enzymes like carboxylesterase and b-glucuroni- targets of cholinergic nerves such as endocrine and exocrine dase2–6. This review will provide an overview of the enzymes glands, smooth and cardiac muscle cells, and sensory organs involved in the hydrolytic activation of prodrugs. embedded in epithelia17. In , AChE is also expressed in non-attached cells such as erythrocytes and lymphocytes. Several prodrugs activated by AChE have been reported. For example, 2. Enzymes butyryl ACV (6, Fig. 2) is an AChE-activated ester prodrug of 2.1. Carboxylesterase acyclovir (ACV), which is a synthetic purine nucleoside analog of and clinically used in the treatment of herpes simplex virus infections because of its affinity for the viral thymidine kinase18. Carboxylesterases (CEs, EC 3.1.1.1) are members of the hydrolase superfamily and they can efficiently hydrolyze a variety of ester-, -, and carbamate-containing xenobio- 2.3. Butyrylcholinesterase tics to their respective free acids. They act as an effective biological barrier to limit the distribution of substrates that Butyrylcholinesterase (BChE, EC 3.1.1.8) is relatively abun- might be toxic and facilitate their elimination by turning them dant in plasma and is sometimes referred to as the serum into more polar . cholinesterase. However, its physiological role remains Although carboxylesterases show varying activity and sub- unclear. BChE can degrade a large number of ester-containing strate specificity, depending on species and location, they are compounds, including ester prodrugs19. The overall 3-D present virtually throughout the body, including intestines, structure of BChE is very similar to that of AChEs. However, blood, brain, skin, and tumor. There are several different BChE does not form the same kind of dimer as observed in isozymes of carboxylesterases; based on sequence homology structures of Torpedo californica AChE, mouse AChE, and and similarity of characteristics, the isozymes have recently AChE15,19. BChE has a broader substrate specificity been classified into five subfamilies, CES1, CES2, CES3, CES4, than AChE, and this difference in specificity can be explained and CES57. However, other names such as human liver by BChE’s more spacious acyl binding pocket at the carboxylesterase 1 (hCE1), carboxylesterase 2 (hCE2), intest- and differences in residues19. inal carboxylesterase (hiCE), and human brain carboxylester- BChE exists predominantly in plasma as well as in various ase (hBr2) continue to be used in the literature. Biochemical tissues such as brain, muscle, , intestine, retina, and analysis indicated that CEs are efficient at hydrolyzing small placenta. Several BChE-activated prodrugs have been developed. substrates, but show vast differences with increasingly larger Bambuterol (7) is slowly converted by BChE to terbutaline, a 20,21 substrates. For example, the human liver hCE1 was 100- to b2-adrenoceptor agonist . Methylprednisolone acetate (8)can 1000-fold less efficient at metabolizing CPT-11 than rabbit liver be hydrolyzed by BChE to its active form—methylprednisolone, a rCE, even though the two enzymes have over 80% amino acid anti-inflammatory agent22. Isosorbide diaspirinate (9), the identity8,9. Analysis of the crystal structures indicates that the diester of isosorbide, is stable towards aqueous hydrolysis Enzyme-mediated hydrolytic activation of prodrugs 145

O O

O N O N CE O O OH N OH N O OH CH3 CH N 3 O N CPT-11 (1) 7-ethyl-10-hydroxycamptothecin

O O

HO O HO O N N NH CE NH S S S S O O O OH Temocapril (2) Temocapril diacid

O OH OH H H HO N O N CE CH O CH3 3 HCl O HO epinephrine Dipivefrin (3)

O OH OOH OH O O HO O H C O 3 H CE H3C R CH H C O 3 3 H H3C R CH3 H3C H C Lovastatin (4) R = H 3 5 hydroxy acid form Simvastatin ( ) R = CH3

Figure 1 Carboxylesterase-mediated activation of representative ester and carbamate prodrugs.

O O

N NH N NH O AChE N N NH N N NH O 2 O 2 O HO Butyryl ACV (6) Acyclovir (ACV)

Figure 2 Acetylcholine esterase-mediated activation of butyryl ACV.

OAc O OH O HO H O H O BChE O O OAc OH + O O H H O OH salicylic acid isosorbide Isosorbide Diaspirinate (9)

Figure 3 Activation of isosorbide diaspirinate by BChE. and in the presence of a-chymotrypsin23. However, it undergoes esterase that can catalyze the hydrolysis of various arylesterase rapid hydrolysis in the presence of BChE as shown in Fig. 3. and organophosphates. PON2 and PON3 share 60% sequence identity with PON1, but, they lack or have very limited arylesterase activities. On the other hand, PON2 and PON3 2.4. Paraoxonase exhibit high lactonase activity. Human PON1 is synthesized in the liver and secreted into the blood, while PON3 is similarly Paraoxonase (PON, EC 3.1.8.1) is a family of hydrolases that expressed predominantly in the liver and at low levels in the include PON1, PON2, and PON3. PON1 is an important kidney24. In recent years, PON1 and its two known Q and R 146 Yan-hui Yang et al. isoenzymes have been shown to possess both arylesterase and require proteolytic processing to produce their active forms that organophosphatase activities, and play important roles in drug usually consist of three main domains, namely a propeptide metabolism and in the prevention of atherosclerosis25. domain, a catalytic domain, and a hemopexin/vitronectin-like Prulifloxacin (10), a lipophilic prodrug of ulifloxacin, is a domain. Several MMPs, including MMP-1, MMP-2, MMP-8, new oral fluoroquinolone with a broad spectrum of in vitro MMP-13, MMP-14, MMP-18, and MMP-22, catalyze the first activity against various Gram-positive and Gram-negative committed step in extracellular matrix degradation by unwinding microorganisms. Its structure contains the skeletal quinolone and cleaving the triple helix of interstitial collagens (type with a four-membered ring in the 1,2-position, including a I–III)29–34. On the other hand, MMP-3 and MMP-9 only possess sulfur atom to increase antibacterial activity and an oxodiox- the ability to bind to single-strand type I collagen. The collagen olenylmethyl group in the 7-piperazine ring to improve its oral cleavage site is similar for most MMPs, and the general motif absorption; it is immediately and quantitatively transformed preceding the scissile bond (between Gly and Ile/Leu) has been by hPON1 into the active metabolite ulifloxacin (Fig. 4)26,27. characterized as a four-triplet region rich in proline with one Lactone-containing drugs, Lovastatin and Simvastatin, are all always at the P3 position. It is important to recognize the ability substrates of hPON315,24,28. to unwind and cleave interstitial collagen in the triple helix form as the main determinant of substrate specificity, rather than the primary structure33,34. 2.5. Matrix metalloproteinase Because of their significant role in tumor angiogenesis, invasion, and metastasis, MMPs have been targeted in poten- Matrix metalloproteinases (MMPs), also known as matrixins, are tial therapeutic strategies for a number of disease conditions -dependent endopeptidases consisting of more than 25 family including cancer, arthritis, glomerulonephritis, periodontal members classified based on domain structure and substrate disease, and ulcers34. Among them, MMP-2 and MMP-9 have specificity (Table 1). They play important roles in the degrada- been used in the targeted activation of potential prodrugs35–38. tion of extracellular matrix components, such as collagen, Model prodrugs have been reported to release, upon activation laminin, fibronectin, and elastin. MMPs also cleave a variety by MMP-9, anthraquinone-type cytotoxic agents that are often of , such as growth factor receptors and cell adhesion used in combination with conventional therapies for the treatment 37 molecules, which may be important for tumor growth and of multiple myeloma . L-Prolyl-N-(3-aminopropyl)-1-aminoan- survival. Synthesized initially as inactive proenzymes, MMPs thraquinone, an adduct known to possess dual topoisomerase

H C H C O 3 O O 3 H3C N N HN O S S S O N N hPON1 O N N N N CH3 O OH H OH OH F F F O O O O O O (10) Ulifloxacin

Figure 4 Activation of prulifloxacin by hPON1.

Table 1 Matrix metalloproteinases and their substrates.

Enzyme Alternative name EC number Substrates

MMP-1 Interstitial collagenase 3.4.24.7 Collagens (I, II, III, VII, VIII, and X); gelatin; aggrecan; L-selectin; IL-1beta; ; entactin; ovostatin; MMP-2; MMP-9 MMP-2 Gelatinase A 3.4.24.24 Collagens (IV, V, VII, and X); gelatin MMP-3 Stromelysin-1, Proteoglycanase 3.4.24.17 Collagens (III, IV, V, and IX); gelatin; aggrecan; perlecan; decorin; laminin; elastin; caesin; osteonectin; ovostatin; entactin; plasminogen; MBP; IL-1beta; MMP-2/TIMP-2; MMP-7; MMP-8; MMP-9; MMP-13 MMP-8 Neutrophil collagenase 3.4.24.34 Collagens (I, II, III, V, VII, VIII, and X); gelatin; aggrecan; fibronectin MMP-9 Gelatinase B 3.4.24.35 Collagens (IV, V, VII, X, and XIV); gelatin; entactin; aggrecan; elastin; fibronectin; osteonectin; plasminogen; MBP; IL-1beta MMP-13 Collagenase-3 – Collagens (I, II, III, IV, IX, X, and XIV); gelatin; plasminogen; aggrecan; perlecan; fibronectin; osteonectin; MMP-9 MMP-14 MT1-MMP 3.4.24.80 Collagens (I-III); gelatin; casein; fibronectin; laminin; vitronectin; entactin; proteoglycans; MMP-2; MMP-13 MMP-18 Xenopus Collagenase-4 – Not known MMP-22 Chicken MMP (C-MMP) – Not known Enzyme-mediated hydrolytic activation of prodrugs 147 inhibitory activity in vivo, was coupled to an optimized substrate class of compounds, including , proteins, and ofMMP-9asshowninEV1(11)37. Cleavage of EV1 by MMP-9 alkaloids. AP is ubiquitously expressed in many tissues with occurs between glycine and norvaline, resulting in the liberation of the highest expression in the liver. The differential expression an active inhibitor of topoisomerases. of various AP isozymes in liver and bone has been used as a Selective delivery of to tumor tissues was diagnostic marker for various cancers and renal dysfunction39. investigated using its conjugates as prodrugs activated AP is believed to be responsible for the activation of several by the combined proteolytic action of MMP-2, MMP-9, and clinically used phosphate prodrugs including fosfluconazole, MMP-14. Doxorubicin is a potent used for the fosphenytoin, fosprepitant, amifostine, phosphate, treatment of various types of cancer. It causes cytotoxicity , and phosphate. Most of these through topoisomerase II-mediated DNA breaks. Coupling of prodrugs were developed to overcome the solubility problems of doxorudicin to several MMP-cleavable generated a parent drugs and are for parental administration while some are series of prodrugs that were readily cleaved by MMP-2, for the targeted treatment of cancer. Amifostine (WR-2721, S-2 MMP-9, and MMP-14. For example, the prodrug Ac–Glu– [3-aminopropylamino]-ethylphosphorothioic acid, 13) was devel- Pro–Cit–Gly–homo-Phe–Tyr–Leu–Dox (12) was cleaved by oped as a protective agent against radiation- and - MMP-2, MMP-9, and MMP-14 to generate a tripeptide–Dox induced cellular injury40. It is selectively activated in normal cells conjugate, which was subsequently degraded by extracellular by AP via dephosphorylation to afford the free WR-1065, proteases to release Leu–doxorubicin as shown in Fig. 5. Leu– which acts a free-radical scavenger (Fig. 6). In addition to free- doxorubicin was known to release doxorubicin upon further radical scavenging, the protective effect of amifostine has been proteolytic cleaveage. Prodrug testing in the fibrosarcoma cell postulated to originate from DNA repair through hydrogen line HT1080 expressing multiple MMPs and HT1080 xeno- donation from the free thiol group, and inhibition of DNA grafts in mice demonstrated the selective conversion of the damage. The protective effects of amifostine have been demon- doxorubicin prodrug to free doxorubicin36. The prodrug also strated in preclinical species in the presence of platinum exhibited a 10-fold increase in tumor/heart distribution ratio, compounds such as . Following incubation of cisplatin greater efficacy and less toxicity as compared to doxorubicin. in salmon sperm DNA in the presence of 50-fold molar excess of amifostine, WR-1065 and related compounds, a maximal 49% 2.6. Alkaline phosphatase reduction in platinum–DNA adduct formation was observed with WR-106541. Clinically, the protective effects of amifostine Alkaline phosphatase (AP, EC 3.1.3.1) is a metalloenzyme administration 30 min prior to chemotherapy have proven (contains zinc and ) with broad substrate specificity. effective although no proof-of-concept could be obtained in It catalyzes the removal of phosphate groups from a diverse in vivo animal studies.

O O OH O OH O CH2OH CH2OH OH OH

OCH3O OH O OCH3 O OH O HO HO

H3C O H3C O MMPs O O O OH H H H O O OH N N N NH H N N N H2N N NH H H H N H3C N O O O H O O O HN MMPs OOHONH2

peptide-doxorubicin prodrug (12) Protease

O OH O CH OH O 2 O OH OH Protease CH2OH OH OCH3O OH O

OCH3O OH O H3C O

H3C O O OH H2N NH OH NH2 Doxorubicin

Figure 5 Activation of a peptide–doxorubicin prodrug designed to be activated by MMPs. 148 Yan-hui Yang et al.

AP-mediated prodrug activation has been applied to cancer acid-containing , such as , imaging and therapy. Such radio-detection of solid tumors . and dermatan sulfate43. The function of in vivo was demonstrated using the radioactive quinazolinone human b-glucuronidase is essential for the restructuring of the 125 phosphate prodrug IQ2-P, the ammonium salt of extracellular matrix components and in the b- 2-(20-phosphoryloxyphenyl)-6-[125I]iodo-4-(3H)-quinazolinone glucuronidase has been shown to cause a rare lysosomal (14). As shown in Fig. 6, the prodrug is hydrolyzed to a much storage disease called mucopolysaccharidosis type-VII (Sly 125 44 less -soluble, radiolabeled quinazolinone IQ2-OH by syndrome) . b-Glucuronidase is a tetrameric glycoprotein AP, which is overexpressed on the exterior surfaces of tumor with four identical subunits of 77 kDa and is localized in cell plasma membranes42. The quinazolinone molecule preci- lysosomes of the cell. The optimal pH for the activity of pitates and is concentrated and permanently trapped within the human b-glucuronidase is around 4, while the physiological extracellular spaces of targeted solid tumors. The prolonged pH of 7.4 leads to decreased enzyme activity43. Since endo- residence time of the quinazolinone molecule should permit the genous extracellular b-glucuronidase is present at high levels in noninvasive detection and therapy of tumors. necrotic tumors, glucuronide prodrugs could potentially be used as a monotherapy. Moreover, in the case of non-necrotic 2.7. b-Glucuronidase areas of tumors where the enzyme concentration is low, exogenous b-glucuronidases can be delivered to these tumor 45 Human b-glucuronidase (EC 3.2.1.31) is a lysosomal enzyme tissues using ADEPT and GDEPT strategies . that plays an important role in the degradation of glucuronic Theactivesitecavityofhumanb-glucuronidase consists of three amino acid residues, Glu540,Glu451,andTyr504. Among them, Glu540 acts as a , while Glu451 acts as an acid– S 2- AP SH 46,47 504 HN PO4 HN base catalyst or the proton donor .Tyr is also important, NH NH2 2 although its role is unclear. b-Glucuronidase have a broader Amifostine (13, WR-2721) WR-1065 substrate specificity; almost any aglycone in a b-linkage can be hydrolyzed to glucuronic acid43. One possible mechanism pro- O O posed for the hydrolytic action by b-glucuronidase is illustrated 125I NH 125I 48 NH in Fig. 7 . N AP Tumor tissues, including breast, lung and gastrointestinal tract N carcinomas, and melanomas have been shown to have increased NH4 O O expression of b-glucuronidase as compared to normal tissues. P HO NH O O The high expression of the b-glucuronidase enzyme is localized 4 43 125 14 125 mainly in necrotic areas of the malignancies . The liberation of IQ2-P ( ) IQ2-OH lysosomal b-glucuronidase extracellularly leads to the accumula- 125 Figure 6 Activation of amifostine and IQ2-P by AP. tion of the enzyme in the extracellular matrix of the malignant

Glu 451 Glu 451 Glu 451 Enz Enz Enz HO R

O O O O OO HOOC HOOC δ+ HOOC H δ− H HO O HO O δ+ O HO O HO O HOHO R R OH OH OH δ− O O O

O O O Enz Enz Enz Glu 540 Glu 540 Glu 540 Enzyme glucuronylation Glucuronyl-Enzyme intermediate

Glu 451 Glu 451 Enz Enz

O O OO HOOC HOOC O H O H H HOHO OH HOHO O OH OH O O

O O Enz Enz Glu 540 Glu 540

Figure 7 The catalytic cycle of b-glucuronidase. The R group shown in red is released upon hydrolysis of the . Enzyme-mediated hydrolytic activation of prodrugs 149 tissues. Cancer tissues have been targeted by a wide variety of acyclovir (ACV), increases the oral bioavailability of ACV glucuronide prodrugs exploiting the presence of high concentra- 3- to 5-fold53. In addition to activating valacyclovir and tions of b-glucuronidase as the enzyme is not present in the valganciclovir, VACVase has been shown to hydrolyze the general circulation. BHAMG (15), a synthetic glucuronide prodrugs of a broad range of antiviral and anticancer nucleoside prodrug of aniline mustard, can be activated by analogs such as zidovudine, floxuridine, and gemcitabine52. b-glucuronidase49. Pyrrolo[2,1-c][1,4]--b-glucuro- nide (16), a prodrug of DNA minor groove binder, has been shown to be activated by b-glucuronidase50. HMR1826 (17), 2.9. Plasmin where the glucuronide group is linked to doxorubicin as showed in Fig. 8,hasaVmax of 635 mmol/(min mg) and a Km of Plasmin (EC 3.4.21.7) is an important present 1.3 mmol/L as a substrate of b-glucuronidase43. Upon b-glucur- in blood that degrades many blood plasma proteins, most onidase-catalyzed hydrolysis, the 4-hydroxybenzyl carbamate- notablely, fibrin clots. In humans, the plasmin is doxorubicin intermediate undergoes self-immolative 1,6-elimina- encoded by the PLG gene. Plasmin is derived from plasmino- tion to release the active doxorubicin drug. gen, a circulating glycoproteins (91 kDa) present in plasma at a concentration of 1.5 mmol/L54. Plasmin is believed to be involved in migration, invasion, 2.8. Human valacyclovirase and metastasis of tumor cells by catalyzing the breakdown of extracellular matrix proteins55,56. Plasmin is predominantly Human valacyclovirase (VACVase) is a highly specific a- present in its inactive pro-enzyme form plasminogen in the amino acid ester hydrolase. VACVase catalyzes the hydrolytic body. Active plasmin is formed locally at or near the surface activation of two clinically important antiviral nucleoside of tumor cells from cell-bound plasminogen activated by cell- prodrugs, valacyclovir and valganciclovir, to their correspond- bound urokinase-type plasminogen activator (u-PA), pro- ing active drugs, acyclovir and ganciclovir51. duced by cancer or stroma cells57. In the general circulation, VACVase is expressed at a high level in human liver and urokinase and plasmin are inactivated by protease inhibitors kidney and lower levels in intestines, heart, and skeletal such as PAI-1 and a2-antiplasmin present in the blood. Many muscle52. Nucleoside analogs valacyclovir (18, VACV) and tumor cell lines and tumor tissues have a significantly higher valganciclovir (19, VGCV) are two typical prodrugs activated u-PA level than their normal counterparts58, and u-PA is a by VACVase. Valacyclovir, the valyl ester prodrug of strong prognostic factor for reduced survival and increased

O2N HOOC HO O O Cl HO OO HO O H OH N O CH2 O N N H 5 H HOOC Cl O HO O N OMe MeO N HO HO O O BHAMG (15) Pyrrolo[2,1-c][1,4]-benzodiazepine--glucuronide (16)

O OH O O OH O OH OH OH OH -glucuronidase

H3CO O OH O H3CO O OH O O H C O H3C HOOC 3 HOOC HO HN O O HOHN O HO O O HO OH HO HO O OH O OH HMR1826 (17)

O OH O OH OH OH + CO2 H3CO O OH O O H3C O NH HO 2 Doxorubicin

Figure 8 Activation of doxorubicin–glucuronide conjugate (HMR1826) by b-glucuronidase followed by 1,6-elimination of the self- immolative linker. 150 Yan-hui Yang et al. relapse in many types of tumors59,60. Therefore, plasmin is and primary prostate cancer, where 80–90% of PSA is considered a promising enzyme for the tumor-specific activa- enzymatically active72. However, intracellular PSA is enzyma- tion of anticancer prodrugs. Characteristic substrate tripep- tically inactive because of the presence of high concentration of tides, D-Ala–Phe–Lys and D-Val–Leu–Lys, with a N-terminal zinc . Normally, PSA is present in blood at very low levels D-amino acid have been used in the design of potential (o4.0 ng/mL). However, increased serum PSA levels are often prodrugs for activation by plasmin. Examples include N- seen in prostate cancer patients, which are believed to be the nitroso-urea-based prodrug (20) with a substrate tripeptide result of cellular PSA leak caused by the absence of a basement coupled via a cyclization linker61. As shown in Fig. 9,an membrane barrier that normally separates the prostate gland ethylene diamine and a mono-methylated ethylene diamine from the surrounding stoma; this lack of confinement is the serve to link the tripeptide substrates to N-nitroso-urea. In the hallmark of invasive prostate cancer73,74. There is evidence that presence of plasminogen, the tripeptide segment is cleaved off serum PSA levels correlate well with the number of malignant by plasmin and subsequent intramolecular cyclization leads to prostate cells, and high serum PSA levels suggest a high risk of the formation of imidazolidin-2-one with concomitant expul- metastatic prostate cancer75. PSA has been approved by FDA sion of the free parent drug. as a clinically important serological biomarker for the screen- Albumin-binding prodrug was reported by ing of prostate cancer. Most importantly, PSA present in conjugation of albumin with the maleimide-linked peptide bloodstream lacks enzymatic activity because it forms an 62 methotrexate 21 . Plasmin cleavage between D-Ala–Phe–Lys inactive complex with protease inhibitors such as a1-antic- 76–78 tripeptide and the lysine linker releases gMTX–eLys–OH, hymotrpsin and a2-macroglobulin . Serum concentrations which is an active cytotoxic agent. Anthracycline-based pro- of these protease inhibitors are 105–106 fold higher than serum drug (22), formed by insertion of a cleavable linker between the PSA levels and, therefore, are able to trap any active PSA tripeptide substrate and doxorubicin, was shown to be stable in present74,79. The loss of enzymatic activity of serum PSA does Tris buffer and bovine serum, and is readily cleaved by not affect the use of PSA as a serological marker because its plasmin63. prodrug (23) with a carbonate linker immunoreactivity is still maintained. These characteristics of was also designed to be selectively activated by plasmin64. The PSA, including specific production in the prostate tissue, paclitaxel-20-carbonate prodrug is stable in Tris buffer, and the enzymatically active form only in extracellular environment, release of free paclitaxel through 1,6-elimination process was enzymatically inactive form in serum and intracellular environ- shown to be instantaneous upon proteolysis by plasmin. ment, and high concentration of PSA in malignant prostate cancer cells, suggest that PSA can be used as a prodrug- converting enzyme to activate anticancer prodrugs to achieve 2.10. Prostate-specific antigen selective therapeutic effects in prostate-derived cancer tissues. Peptide-based prodrugs linking a substrate peptide sequence Prostate-specific antigen (PSA, EC 3.4.21.77) was first identi- to an anticancer drug have been reported for proteolytic fied in human seminal plasma in 1969 (Fig. 11). PSA is a activation by PSA. Upon proteolytic cleavage by PSA, the glycoprotein composed of a single polypeptide chain with a anticancer agent is then released locally in the microenviron- total mass of 33–34 kDa containing approximately 7% carbo- ment that surrounds prostate cancer cells. The anticancer hydrate65. PSA is primarily produced by prostate ductal and selectivity is, therefore, achieved through the site-specific acinar epithelium and is secreted into the lumen, where it activation of PSA. The peptide should be cleaved specifically cleaves semenogelin I and II in the seminal coagulum66. PSA by PSA, and resist the hydrolytic actions of other enzymes; has been shown to activate urokinase-type plasminogen acti- thus, activation of the prodrugs will be localized in the prostate vator, which is thought to be involved in cancer invasion and and prostate-derived cancer tissues where 80–90% of PSA is metastasis67. PSA could also affect tumor spread by proteolytic enzymatically active. Several short peptide sequences have been modulation of cell adhesion receptors68. Furthermore, PSA is identified as PSA-specific, including Ser–Lys–Leu–Gln–, His– able to cleave insulin-like growth factor binding protein 3 Ser–Ser–Lys–Leu–Gln–, and glutaryl-Hyp–Ala–Ser–Chg–Gln–, 80,81 (IGFBP-3) causing the release of active IGF-I, which could all with a unique residue at P1 site . These enhance tumor growth69. However, other studies indicated that unique PSA substrate sequences were attached to anticancer PSA may inhibit tumor growth by generating antiangiogenic drugs with the dipeptide Ser–Leu or Leu as the linker to angiostatin from plasminogen70,71. improve the rate of PSA cleavage (e.g., glutaryl-Hyp–Ala–Ser– The concentration of PSA in the prostate extracellular fluid Chg–Gln–Ser–Leu–Dox (24), N-morpholinocarbonyl-His–Ser– is around 1.6–2.1 mmol/L (50–68 mg/mL) in normal prostate Ser–Lys–Leu–Gln–Leu–Drug). The peptide–drug conjugates

O O + RHN Cl plasmin Cl NN H2N Cl NN NN OH R' NO R' NO N 20 -nitroso-urea-based prodrugs ( ) O

R = D-Ala-Phe-Lys, D-Val-Leu-Lys HN NR' N2 R' = H, CH3 O Cl Cl Cl H , ,

Figure 9 Plasmin activation of N-nitroso-urea-based prodrugs. Enzyme-mediated hydrolytic activation of prodrugs 151 were shown to have increased therapeutic index in preclinical while high molecular mass homohexamers, mainly found in prostate cancer models, validating the use of PSA as a bacteria, accept as substrates both 6-oxo- and/or 6-aminopur- prodrug-converting enzyme82. ines and their nucleosides95. In mammals it is located especially Various anticancer agents, including doxorubicin81,83, thapsi- in the liver, brain, thyroid, kidney, and spleen. The highest gargin84,85, vinblastine86,87, 5-fluoro-20-deoxyuridine88, 3-hydroxy- concentrations of the enzyme are found in the kidney, periph- lapapchone, diazeniumdiolate89,nitrogenmustard90,protoxin91, eral lymphocytes, and granulocytes39. and paclitaxel92,havebeenusedinprodrugsthatshowedselective The selective cleavage of purine nucleoside analogs by E. coli cytotoxicity against PSA-expressing cells. The peptide– PNP is being evaluated as a possible suicide gene therapy doxorubicin conjugate 24 was evaluated in phase I clinical trials82. strategy (GDEPT) for the treatment of solid tumors. Excellent Another design of PSA substrate peptide conjugates of antitumor activity has been demonstrated with 9-(2-deoxy-b-D- anticancer drugs is to incorporate cleavable linkers between ribofuranosyl)-6-methylpurine (MeP-dR, 27) against tumors PSA substrate peptide and anticancer drugs84,86,93,94.As that express E. coli PNP96. The active metabolite 6-methylpur- shown in Fig. 10, the parent drug FUDR could be released ine (MeP), can readily diffuse across cell membranes and kill through cyclization upon proteolytic cleavage of prodrug 25, adjacent tumor cells that do not express E. coli PNP (Fig. 12)96. thus maintaining the potency and physiological property of 2-Fluoro-20-deoxyadenosine (F-dAdo, 28) is another nucleoside the parent drug. analog that is an excellent substrate for E. coli PNP97.Itis Spontaneous conversion of 4-aminocyclophosphamide to better tolerated by mice than MeP-dR, and the active form is phosphoramide mustard under physiologiocal conditions was F-Ade, which is active against human tumor cells that do not demonstrated and exploited towards the design PSA-activated express E. coli PNP due to its conversion to toxic metabolites prodrugs94. A series of 4-aminocylophosphamide prodrugs and the subsequent inhibition of DNA synthesis97. (26) were synthesized by conjugating 4-aminocylophospha- mide to the tetrapeptide Cbz–Ser–Ser–Phe–Tyr sequence, a PSA substrate. The stereoselectivity of PSA-mediated cleavage CH3 of the prodrugs was found to favor the cis-(2R,4R)-conjugate, CH3 N N which has a t1/2 of 12 min in the presence of PSA (Fig. 11). N E. coli PNP N HO N N O N N H 2.11. Purine-nucleoside phosphorylase OH MeP-dR (27) MeP Purine-nucleoside phosphorylase (PNP, EC 2.4.2.1) catalyzes the cleavage of the glycosidic bond of ribo- and deoxyribonu- NH2 cleosides, in the presence of inorganic orthophosphate (Pi) as a NH N 2 second substrate, to generate the purine base and (deox- N N E. coli PNP N yribose)-1-phosphate. For the natural substrates, the reaction is HO N N F 95 O N N F reversible . PNPs are found in a broad range of organisms, H and divided into two main categories based on size, low OH molecule mass homotrimers (about 80–100 kDa) and high F-dAdo (28) F-Ade molecular mass homohexamers (about 110–160 kDa)95.Low molecular mass homotrimers, mainly isolated from mammals, Figure 12 The bioconversion of MeP-dR and F-dAdo by E. coli are specific for of 6-oxopurines and their nucleosides, PNP in GDEPT.

O O O F O NH2 F F O HN O HN HN H N PSA HN Mu-His-Ser-Ser-Lys-Leu-Gln N O O N O O N HO O N H O O O O OH OH OH Mu-His-Ser-Ser-Lys-Leu-Gln-OH peptide-Fluorodeoxyuridine conjugate (25) 5-Fluoro-2'-deoxyuridine

Figure 10 Example of a peptide–fluorodeoxyuridine conjugate and its release mechanism.

OH O O O O HO H H O PSA O O N N P P P Cbz N N N * N N(CH2CH2Cl)2 H2N * N N(CH CH Cl) H H H H H 2 2 2 H2N N(CH CH Cl) O O 2 2 2 OH 26 Rate of proteolytic activation: OH cis-(2R,4R)-26 > trans-(2S,4R)-26 >> trans-(2S,4S)-26 > cis-(2R,4S)-26

Figure 11 Stereoselectivity of PSA cleavage of peptide 4-aminocyclophosphamide conjugates as potential prodrugs of phosphoramide mustard activated by PSA. 152 Yan-hui Yang et al.

2.12. Carboxypeptidase G2 2.13. Penicillin amidase

Glucarpidase under development as a chemoprotective agent Penicillin amidase (PA), also known as penicillin acylase, is to neutralize the toxic effects of methotrexate is a recombinant widely expressed in various microorganisms including, yeast, bacterial carboxypeptidase G2 (CPG2, EC 3.4.17.11). This fungi, and bacteria. It is a member of the N-terminal- CPG2 from Pseudomonas RS-16 is a 42 kDa exopeptidase in nucleophile hydrolase family, a group of enzymes sharing a the aminoacylase-1/M20 family of enzymes that is not found characteristic fold proximal to their active sites as well as a in mammals. The natural substrate of CPG2 is folic acid, but nucleophilic residue at their N-terminus. The PA family it is capable of hydrolyzing glutamate from amidic98,99, members hydrolyze a series of penicillins, and are subdivided urethanic and ureidic bonds100,101, which allows it to be used into three subtypes based on their substrate specificity, namely to activate synthetic prodrugs in GDEPT and ADEPT as penicillin V amidases (PVA, Type I; substrate: penicillin V, represented by the reaction shown in Fig. 13. phenoxymethylpenicillin), penicillin G amidase (PGA, Type A series of CPG2-activated mustard prodrugs has II; substrate: penicillin G, benzylpenicillin) and ampicillin been developed with various cytotoxic nitrogen mustards linked to amidases (Type III; substrate: ampicillin). The E. coli isozyme glutamate directly98,99,102–104 or through a self-immolative lin- (EC 3.5.1.11) is a well-characterized, inexpensive and com- ker105,106. CPG2 catalyzes the hydrolytic activation of these mercially available Type II amidase commonly used for the prodrugs, releasing and the parent cytotoxins, production of semisynthetic penicillins. including benzoic acid nitrogen mustards (BANM), aniline nitro- PGA is known to have broad substrate specificity; several gen mustards (ANM) and phenol nitrogen mustards (PNM). targeted prodrugs activated by PGA have been reported112,113. 4-[(2-mesyloxylethyl)(2-chloroethyl)amino]benzoyl-L-glutamic acid The potent cytotoxin palytoxin exhibits activity against a wide (CMDA, 29) was the first ADEPT prodrug evaluated clinically107. spectrum of cell types including fibroblasts, lymphocytes, A phase I ADEPT clinical trial of CDMA showed promising erythrocytes and epithelial cells. It is cytotoxic against cells results, with one patient showing a partial response and six expressing the Naþ–Kþ-ATPase associated toxin receptor. with stable disease for 4 months. However, the activated drug, The N-acyl palytoxin derivative, N-(40-hydroxyphenylacetyl)- 4-[(2-mesyloxylethyl)-(2-chloroethyl)amino]benzoic acid, had a palytoxin (NHPAP, 32) was developed as a targeted prodrug long half-life that resulted in dose-limiting myelosuppression due for PGA activation via ADEPT112. PGA was covalently to diffusion of the activated drug back into the circulation. linked to the monoclonal antibody L6 mAb. Compared to The pyrrolo[2,1-c][1,4] (PBDs) are a family palytoxin, the prodrug was 1000 times less toxic to carcinoma of antitumor produced by Streptomyces species108. and lymphoma cells lacking PGA expression. NHPAP was They exert their cytotoxicity by covalently bonding to the efficiently converted to palytoxin in H2981 cells expressing the exocyclic C2–NH2 group of guanine residues in the minor L6 antigen, and exhibited comparable cytotoxic activity to the groove of DNA through their N10–C11 imine functionality. free drug (IC50 values were 0.06 and 0.01 nmol/L for NHPAP The more stable carbamate prodrugs are good substrates for and palytoxin, respectively). CPG2. The prodrug 30 was completely converted into the The L6-PGA conjugate has also been used in the ADEPT- cytotoxic parent PBD within 50 min of incubation with the assisted activation of doxorubicin and melphalan113. The enzyme in vitro. prodrugs, N-(phenylactemido)doxorubicin (33) and N-(pheny- Methotrexate conjugates with amino acids or peptides lacetyl) (34) were found to be 10- and 20-fold less covalently linked to the a-carboxyl group of methotrexate cytotoxic than free drugs in H2981 cancer lines lacking PGA. (MTX) are prodrug forms of the parent antifolate109–111. Co-incubation with PGA increased the cytotoxicity of both When tested against UCLA-P3 human lung carcinoma cells prodrugs due to PGA activation. For example, the IC50 values in vitro, the IC50 values for methotrexate–amino acid con- of prodrug 33 were 2 mmol/L in the absence of PGA and jugates in the absence of CPG2 were in the micromolar range 0.2 mmol/L in the presence of PGA, whereas the IC50 of (about 7.0 mmol/L). This value was much higher than the IC50 doxorubicin was 0.2 mmol/L in the same assay. for MTX, which was in nanomolar range (28 nmol/L). When incubated with UCLA-P3 human lung adenocarcinoma cells treated with KS1/4-CPG2 conjugate (KS1/4 is a carcinoma- 2.14. b-Lactamase specific monoclonal antibody), MTX–Phe (31) produced an IC50 of 63 nmol/L. KS1/4–CPG2 conjugate treatment alone b-Lactamase (EC 3.5.2.6) is a serine protease produced by produced no cytotoxicity and, in the absence of KS1/4-CPG2 various bacteria; it catalyzes the hydrolysis of the b-lactam conjugate, cells were much less susceptible to MTX–Phe moiety in penicillin and other similar b-lactam antibiotics to (IC50 ¼2.2 mmol/L). b-amino acid. Based on primary sequences and metal ion

Figure 13 The mechanism of mustard prodrugs activated by CPG2. Enzyme-mediated hydrolytic activation of prodrugs 153 requirement, b-lactamases have been grouped into four As shown in Fig. 14, opening of the b-lactam ring in classes. Classes A, C, and D utilize a serine residue in the cephalosporin is initiated by the nucleophillic attack from the active site, whereas class B enzymes require an additional hydroxyl group of the Ser70 side chain at the carbonyl catalytic zinc atom for activity. All of these enzymes contain (CQO) of the b-lactam ring. The drug attached to the C30 the active site serine residue as well as a conserved triad of position represented as a leaving group (LG) is released by the KS(T)G between the active site serine and the C terminus. The movement of electrons towards the thiazine ring of the cephem class A b-lactamases are largely plasmid-encoded and are core. It has been noted that the propensity for the drug release widely distributed in both Gram-positive and Gram-negative depends on the leaving group ability of the drug molecule and bacteria. The b-lactamases produced by Gram-positive organ- the stability of the linkage to the cephalosporin. isms are usually secreted enzymes. b-Lactamase may be Vinca alkaloid derivative, LY233425, was also conjugated clinically beneficial when orally administered to preserve the to C30 position of cephalosporin through an alkyl thioether, natural intestinal flora during the parenteral administration of antibiotics. NH2 O The advantages of using b-lactamase in prodrug activation F F N HN in ADEPT are that the enzyme is not present in human cells deaminase and that it is highly selective to the b-lactam containing O N O N H H compounds. Cephalosporin has been widely utilized as the 5-Fluorocytosine (39) 5- core of b-lactamase-activated prodrugs due to its ability of releasing the drug from C30 position following the cleavage of Figure 16 The activation of 5-fluorocytosine by cytosine deami- b-lactam ring by b-lactamase. nase in GEDPT.

O O O R NH R NH S R NH S O S O N LGN LG O N + LG Ser O 3' O 70 Ser70 OOH OOH OOH

Ser70 O + H2O -H

O O Enzyme R NH R NH O S O S OH OH N O N Ser70

OOH OOH

Figure 14 The mechanism of activation of cephalosporin prodrugs by b-lactamases114.

O N O N O N

H Cl H Cl H3C H3C H H NH NH N N NH -galactosidase Winstein-cyclization N O O O HO HO O O OH O HO OH prodrug of duocarmycin analog (37) seco-drug drug

O OH O O OH O OH OH OH OH O HO OCH O OH O -galactosidase H 3 O N OCH3 O OH O HO Dox O H C O O HO O 3 O H3C O OH OH NH OH O NH2 + CO2 O Doxorubicin glycoside prodrug of doxorubicin (38)

Figure 15 Activation of glycoside prodrugs by b-galactosidase. 154 Yan-hui Yang et al.

Figure 17 The structures of other compounds. Enzyme-mediated hydrolytic activation of prodrugs 155

OH HO OH O OH OH OH CH3 OH CH3 O O H3C O N N OH O H H CH OH OH OH OH OH H3C 3

OH OH OH CH OH OH OH 3 O O O OH OH OH OH OH HO OH HO OH OH OH OH

OH HO

OH OH HO O O O OH CH3 OH R O HO OH HN OH OH Palytoxin (PTX) R = H

N-(4'-hydroxyphenylacetyl) palytoxin (NHPAP, 32) R = HO CH2 C O

O OH O

CH2OH Cl Cl OH N

OCH3O OH O

H3C O H N OH NH O HOOC O N-(phenylactemido)doxorubicin (33) N-(phenylactemido)melphalan (34)

Et N OH

H N N OAc S O N Et O OH S O H H H COOMe N O N O MeOOC O O O MeO OH O S NH N H O OH S Ph O HO H Me BzOAcO O NHBz O N S NH O N H O OH LY233425 cephalosporin prodrug (35) Cephalosporin taxol (Protax, 36)

Figure 17 (continued)

which would leave as a thiol upon hydrolysis of the b-lactam taxol did not readily react with the p-nitrophenyl carbonate ring115. This vinca alkaloid prodrug 35 was shown to be 5 fold attached to cephalosporin at C30, a GABA linker was used to less toxic than the free drug; but, it was equally potent as the conjugate the C20-hydroxyl group of taxol with cephalosporin. parent drug in the presence of b-lactamase. Since the C20-hydroxyl group is required for biological A carbamate linker was used in a cephalosporin-taxol activity, the prodrug will only be activated after the C20 linker prodrug, Protax (36)116. Because the C20 hydroxyl group of is removed. 156 Yan-hui Yang et al.

2.15. b-Galactosidase deliver active drug molecules to a specific target tissue or organ, non-endogenous enzymes have been employed for site- E. coli b-galactosidase (EC 3.2.1.23), encoded by lacZ, has specific activation of prodrugs. These novel approaches been used in ADEPT. This enzyme is often used for histolo- include ADEPT, GDEPT, and VDEPT strategies for the gical localization with 5-brom-4-chloro-3-indolyl-beta-D- treatment of cancer. Enzymes used in these strategies include galactopyranoside (X-Gal) as the substrate. A glycoside some of the hydrolytic enzymes discussed above. These prodrug of duocarmycin analog (37) consists of the approaches are very promising but are also very challenging. (1S,10R)-methyl-seco-CBI-skeleton as effector, a dimethyla- There are still many problems to solve. With the advance of minoethoxyindole (DMAI) side chain for binding to the minor new technologies, it is very likely we will see some of these 117 groove of DNA and b-D-galactose as a promoiety . This approaches become effective tools against various diseases. prodrug has a rather low cytotoxicity with an IC50 of 3600 nmol/L but a very high cytotoxicity upon activation by b-galactosidase to give a highly cytotoxic duocarmycin analog 118 (Fig. 15) . Another example is the glycoside prodrug 38 of References doxorubicin, which was found to be stable in buffer and could 119 be readily converted to doxorubicin by b-galactosidase . 1. Rautio J, Kumpulainen H, Heimbach T, Oliyai R, Oh D, Jarvinen¨ T, et al. Prodrugs: design and clinical applications. 2.16. Cytosine deaminase Nat. Rev. Drug Discov. 2008;7:255–70. 2. Chen Y, Hu L. Design of anticancer prodrugs for reductive Cytosine deaminase (CD, EC 3.5.4.1) catalyzes the hydrolytic activation. Med. Res. Rev. 2009;29:29–64. deamination of cytosine to and 5-methylcytosine to 3. Hsieh PW, Hung CF, Fang JY. Curr. prodrug design for drug thymine. The enzyme has been found in bacteria and fungi, discovery. Curr. Pharm. Design 2009;15:2236–50. 4. Law B, Tung CH. Proteolysis: a biological process adapted in where it plays an important role in pyrimidine salvage120. drug delivery, therapy, and imaging. Bioconjugate Chem. However, no CD is present in mammalian cells, which utilize 2009;20:1683–95. the deaminase instead. Comparison of the active sites 5. Testa B. Prodrugs: bridging pharmacodynamic/pharmacokinetic of CD with other cytidine and deoxycytidylate deaminase gaps. Curr. Opin. Chem. Biol. 2009;13:338–44. family, reveals a very similar interaction network between the 6. Jana S, Mandlekar S, Marathe P. Prodrug design to improve attacking water molecule, the zinc ion, the zinc ligands, and pharmacokinetic and drug delivery properties: challenges to the the proton shuttle, suggesting a similar zinc-assisted deamina- discovery scientists. Curr. Med. Chem. 2010;17:3874–908. tion mechanism in the family120. 7. Satoh T, Hosokawa M. Structure, function and regulation of The yeast enzyme cytosine deaminase has also been widely carboxylesterases. Chem. Biol. Interact. 2006;162:195–211. studied for GDEPT in conjunction with the antifungal agent 8. Bencharit S, Morton CL, Howard-Williams EL, Danks MK, Potter PM, Redinbo MR. Structural insights into cpt-11 activa- 5-fluorocytosine (39), which is converted by CD to the toxic tion by mammalian carboxylesterases. Nat. Struct. Biol. thymidylate synthase inhibitor 5-fluorouracil as shown in 2002;9:337–42. Fig. 16. This freely diffusible effector, already a drug of choice 9. Wadkins RM, Morton CL, Weeks JK, Oliver L, Wierdl M, for treating colon cancer, has much better bystander effects. Danks MK, et al. Structural constraints affect the metabolism of GDEPT model studies with CD/5-fluorocytosine have focused 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothe- mainly on colon cancer models. Possible drawbacks of this cin (cpt-11) by carboxylesterases. Mol. Pharmacol. 2001;60: protocol include the relatively low potency of the effector, 355–62. coupled with its pronounced selectivity121,122. 10. Potter PM, Wadkins RM. Carboxylesterases–detoxifying enzymes and targets for drug therapy. Curr. Med. Chem. 2006;13:1045–54. 3. Concluding remarks 11. Rivory LP, Bowles MR, Robert J, Pond SM. Conversion of irinotecan (cpt-11) to its active metabolite, 7-ethyl-10-hydroxy- Prodrugs have been very effective in solving problems of the (sn-38), by human liver carboxylesterase. Biochem. parent molecules to provide the desired physicochemical and Pharmacol. 1996;52:1103–11. 12. Yasunari K, Maeda K, Nakamura M, Watanabe T, Yoshikawa J, pharmaceutical properties for improved oral bioavailability, Asada A. Pharmacological and clinical studies with temocapril, an patient acceptability, pharmacokinetic profiles, and target angiotensin converting that is excreted in the specificity. There are many examples of prodrugs that are bile. Cardiovasc. Drug Rev. 2004;22:189–98. used clinically. Enzymes play an important role in the 13. Nakamura M, Shirasawa E, Hikida M. Characterization of activation of many prodrugs although some prodrugs can be esterases involved in the hydrolysis of dipivefrin hydrochloride. activated solely through chemical reactions without involving Ophthalmic Res. 1993;25:46–51. enzymes. 14. Fleming CD, Bencharit S, Edwards CC, Hyatt JL, Tsurkan L, With the discovery of more compounds with desired Bai F, et al. Structural insights into drug processing by human activity but suboptimal pharmaceutical properties, prodrugs carboxylesterase 1: tamoxifen, mevastatin, and inhibition by have become an increasingly important approach that can be benzil. J. Mol. Biol. 2005;352:165–77. 15. Liederer BM, Borchardt RT. Enzymes involved in the biocon- used to solve some of these problems. The structures of version of ester-based prodrugs. J. Pharm. Sci. 2006;95:1177–95. compounds not in reactions were shown in Fig. 17. Knowing 16. Quinn DM. Acetylcholinesterase: enzyme structure, reaction the various enzymes that could potentially be used to activate dynamics, and virtual transition states. Chem. Rev. 1987; potential prodrugs will definitely help with the design process. 87:955–79. Most often endogenous enzymes can be effectively utilized 17. Rotundo RL. Expression and localization of acetylcholinesterase for the activation of prodrugs. With the recent shift of focus to at the neuromuscular junction. J. Neurocytol. 2003;32:743–66. Enzyme-mediated hydrolytic activation of prodrugs 157

18. Tak RV, Pal D, Gao H, Dey S, Mitra AK. Transport of 39. Rooseboom M, Commandeur JN, Vermeulen NP. Enzyme- acyclovir ester prodrugs through rabbit cornea and sirc-rabbit catalyzed activation of anticancer prodrugs. Pharmacol. Rev. corneal epithelial cell line. J. Pharm. Sci. 2001;90:1505–15. 2004;56:53–102. 19. Nicolet Y, Lockridge O, Masson P, Fontecilla-Camps JC, 40. van der Vijgh WJ, Peters GJ. Protection of normal tissues from Nachon F. Crystal structure of human butyrylcholinesterase the cytotoxic effects of chemotherapy and radiation by amifos- and of its complexes with substrate and products. J. Biol. Chem. tine (ethyol): Preclinical aspects. Semin. Oncol. 1994;21:2–7. 2003;278:41141–7. 41. Treskes M, Nijtmans LG, Fichtinger-Schepman AM, van der 20. Tunek A, Svensson LA. Bambuterol, a carbamate ester prodrug Vijgh WJ. Effects of the modulating agent wr2721 and its main of terbutaline, as inhibitor of cholinesterases in human blood. metabolites on the formation and stability of cisplatin-DNA Drug Metab. Dispos. 1988;16:759–64. adducts in vitro in comparison to the effects of thiosulphate 21. Waldeck B. Beta-adrenoceptor agonists and asthma—100 years of and diethyldithiocarbamate. Biochem. Pharmacol. 1992;43: development. Eur. J. Pharmacol. 2002;445:1–12. 1013–9. 22. Meyers C, Lockridge O, La Du BN. Hydrolysis of methylpred- 42. Chen K, Wang K, Kirichian AM, Al Aowad AF, Iyer LK, nisolone acetate by human serum cholinesterase. Drug Metab. Adelstein SJ, et al. In silico design, synthesis, and biological Dispos. 1982;10:279–80. evaluation of radioiodinated quinazolinone derivatives for 23. Gilmer JF, Moriarty LM, Lally MN, Clancy JM. Isosorbide- alkaline phosphatase-mediated cancer diagnosis and therapy. based aspirin prodrugs. Ii. Hydrolysis kinetics of isosorbide Mol. Cancer Ther. 2006;5:3001–13. diaspirinate. Eur. J. Pharm. Sci. 2002;16:297–304. 43. de Graaf M, Boven E, Scheeren HW, Haisma HJ, Pinedo HM. 24. Draganov DI, La Du BN. Pharmacogenetics of paraoxonases: a Beta-glucuronidase-mediated drug release. Curr. Pharm. Design brief review. Naunyn Schmiedebergs Arch. Pharmacol. 2002;8:1391–403. 2004;369:78–88. 44. Vogler C, Barker J, Sands MS, Levy B, Galvin N, Sly WS. 25. Harel M, Aharoni A, Gaidukov L, Brumshtein B, Khersonsky Murine mucopolysaccharidosis vii: impact of therapies on the O, Meged R, et al. Structure and evolution of the serum phenotype, clinical course, and pathology in a model of a paraoxonase family of detoxifying and anti-atherosclerotic lysosomal storage disease. Pediatr. Dev. Pathol. 2001;4:421–33. enzymes. Nat. Struct. Mol. Biol. 2004;11:412–9. 45. Alaoui AE, Saha N, Schmidt F, Monneret C, Florent JC. New 26. Tougou K, Nakamura A, Watanabe S, Okuyama Y, Morino A. taxol (paclitaxel) prodrugs designed for adept and pmt strategies Paraoxonase has a major role in the hydrolysis of prulifloxacin in cancer chemotherapy. Bioorg. Med. Chem. 2006;14:5012–9. 46. Jain S, Drendel WB, Chen ZW, Mathews FS, Sly WS, Grubb (nm441), a prodrug of a new antibacterial agent. Drug Metab. JH. Structure of human beta-glucuronidase reveals candidate Dispos. 1998;26:355–99. lysosomal targeting and active-site motifs. Nat. Struct. Biol. 27. Matera MG. Pharmacologic characteristics of prulifloxacin. 1996;3:375–81. Pulm. Pharmacol. Ther. 2006;19(Suppl 1)20–9. 47. Islam MR, Tomatsu S, Shah GN, Grubb JH, Jain S, Sly WS. 28. Billecke S, Draganov D, Counsell R, Stetson P, Watson C, Hsu Active site residues of human beta-glucuronidase. Evidence for C, et al. Human serum paraoxonase (pon1) isozymes q and r glu(540) as the nucleophile and glu(451) as the acid-base residue. hydrolyze lactones and cyclic carbonate esters. Drug Metab. J. Biol. Chem. 1999;274:23451–5. Dispos. 2000;28:1335–42. 48. Wong AW, He S, Grubb JH, Sly WS, Withers SG. Identification 29. Farr M, Pieper M, Calvete J, Tschesche H. The n-terminus of of glu-540 as the catalytic nucleophile of human beta-glucur- collagenase mmp-8 determines superactivity and inhibition: a onidase using electrospray mass spectrometry. J. Biol. Chem. relation of structure and function analyzed by biomolecular 1998;273:34057–62. interaction analysis. Biochemistry 1999;38:7332–8. 49. Chen BM, Chan LY, Wang SM, Wu MF, Chern JW, Roffler 30. Diaz N, Suarez D. Peptide hydrolysis catalyzed by matrix SR. Cure of malignant ascites and generation of protective metalloproteinase 2: a computational study. J. Phys. Chem. B immunity by monoclonal antibody-targeted activation of a 2008;112:8412–24. glucuronide prodrug in rats. Int. J. Cancer 1997;73:392–402. 31. Botos I, Meyer E, Swanson SM, Lemaitre V, Eeckhout Y, 50. Kamal A, Tekumalla V, Raju P, Naidu VG, Diwan PV, Sistla R. Meyer EF. Structure of recombinant mouse collagenase-3 Pyrrolo[2,1-c][1,4]benzodiazepine-beta-glucuronide prodrugs (mmp-13). J. Mol. Biol. 1999;292:837–44. with a potential for selective therapy of solid tumors by pmt 32. Briknarova K, Gehrmann M, Banyai L, Tordai H, Patthy L, and adept strategies. Bioorg. Med. Chem. Lett. 2008;18:3769–73. Llinas M. Gelatin-binding region of human matrix metallopro- 51. Lai L, Xu Z, Zhou J, Lee KD, Amidon GL. Molecular basis of teinase-2: Solution structure, dynamics, and function of the col- prodrug activation by human valacyclovirase, an alpha-amino 23 two-domain construct. J. Biol. Chem. 2001;276:27613–21. acid ester hydrolase. J. Biol. Chem. 2008;283:9318–27. 33. Fields GB. A model for interstitial collagen by 52. Kim I, Song X, Vig BS, Mittal S, Shin HC, Lorenzi PJ, et al. A mammalian collagenases. J. Theor. Biol. 1991;153:585–602. novel nucleoside prodrug-activating enzyme: substrate specificity 34. Lauer-Fields JL, Juska D, Fields GB. Matrix metalloproteinases of biphenyl hydrolase-like protein. Mol. Pharmacol. 2004;1: and collagen catabolism. Biopolymers 2002;66:19–32. 117–27. 35. Watermann I, Gerspach J, Lehne M, Seufert J, Schneider B, 53. Kim I, Chu XY, Kim S, Provoda CJ, Lee KD, Amidon GL. Pfizenmaier K, et al. Activation of cd95l fusion protein prodrugs Identification of a human valacyclovirase: biphenyl hydrolase- by tumor-associated proteases. Cell Death Differ. 2007;14:765–74. like protein as valacyclovir hydrolase. J. Biol. Chem. 36. Albright CF, Graciani N, Han W, Yue E, Stein R, Lai Z, et al. 2003;278:25348–56. Matrix metalloproteinase-activated doxorubicin prodrugs inhibit 54. Novokhatny V. Structure and activity of plasmin and other direct ht1080 xenograft growth better than doxorubicin with less thrombolytic agents. Thromb. Res. 2008;122(Suppl 3):S3–8. toxicity. Mol. Cancer Ther. 2005;4:751–60. 55. Quax PH, de Bart AC, Schalken JA, Verheijen JH. Plasminogen 37. Van Valckenborgh E, Mincher D, Di Salvo A, Van Riet I, Young L, activator and matrix metalloproteinase production and extra- Van Camp B, et al. Targeting an mmp-9-activated prodrug to cellular matrix degradation by rat prostate cancer cells in vitro: multiple myeloma-diseased bone marrow: a proof of principle in the correlation with metastatic behavior in vivo. Prostate 1997;32: 5t33mm mouse model. Leukemia 2005;19:1628–33. 196–204. 38. Kridel SJ, Chen E, Kotra LP, Howard EW, Mobashery S, 56. Quax PH, van Muijen GN, Weening-Verhoeff EJ, Lund LR, Smith JW. Substrate hydrolysis by matrix metalloproteinase-9. Dano K, Ruiter DJ, et al. Metastatic behavior of human J. Biol. Chem. 2001;276:20572–8. melanoma cell lines in nude mice correlates with urokinase-type 158 Yan-hui Yang et al.

plasminogen activator, its type-1 inhibitor, and urokinase- serum as a screening test for prostate cancer. New Eng. J. Med. mediated matrix degradation. J. Cell Biol. 1991;115:191–9. 1991;324:1156–61. 57. Hewitt R, Dano K. Stromal cell expression of components of 76. Christensson A, Laurell CB, Lilja H. Enzymatic activity of matrix-degrading protease systems in human cancer. Enzyme prostate-specific antigen and its reactions with extracellular Protein 1996;49:163–73. serine proteinase inhibitors. Eur. J. Biochem. 1990;194:755–63. 58. Quax PH, van Leeuwen RT, Verspaget HW, Verheijen JH. 77. Otto A, Bar J, Birkenmeier G. Prostate-specific antigen forms Protein and messenger levels of plasminogen activators and complexes with human alpha 2-macroglobulin and binds to the inhibitors analyzed in 22 human tumor cell lines. Cancer Res. alpha 2-macroglobulin receptor/ldl receptor-related protein. 1990;50:1488–94. J. Urol. 1998;159:297–303. 59. Schmitt M, Harbeck N, Thomssen C, Wilhelm O, Magdolen V, 78. Lilja H. A kallikrein-like serine protease in prostatic fluid cleaves Reuning U, et al. Clinical impact of the plasminogen activation the predominant seminal vesicle protein. J. Clin. Invest. system in tumor invasion and metastasis: prognostic relevance 1985;76:1899–903. and target for therapy. Thromb. Haemostasis 1997;78:285–96. 79. Lilja H, Christensson A, Dahlen U, Matikainen MT, Nilsson O, 60. Ganesh S, Sier CF, Griffioen G, Vloedgraven HJ, de Boer A, Pettersson K, et al. Prostate-specific antigen in serum occurs Welvaart K, et al. Prognostic relevance of plasminogen activa- predominantly in complex with alpha 1-antichymotrypsin. Clin. tors and their inhibitors in colorectal cancer. Cancer Res. Chem. 1991;37:1618–25. 1994;54:4065–71. 80. Denmeade SR, Lou W, Lovgren J, Malm J, Lilja H, Isaacs JT. 61. Eisenbrand G, Lauck-Birkel S, Tang W. An approach towards Specific and efficient peptide substrates for assaying the proteo- more selective anticancer agents. Synthesis 1996:1246–58. lytic activity of prostate-specific antigen. Cancer Res. 1997;57: 62. Warnecke A, Fichtner I, Sass G, Kratz F. Synthesis, cleavage 4924–30. profile, and antitumor efficacy of an albumin-binding prodrug of 81. Garsky VM, Lumma PK, Feng DM, Wai J, Ramjit HG, methotrexate that is cleaved by plasmin and cathepsin b. Arch. Sardana MK, et al. The synthesis of a prodrug of doxorubicin Pharm. 2007;340:389–95. designed to provide reduced systemic toxicity and greater target 63. de Groot FM, de Bart AC, Verheijen JH, Scheeren HW. efficacy. J. Med. Chem. 2001;44:4216–24. Synthesis and biological evaluation of novel prodrugs of anthra- 82. DiPaola RS, Rinehart J, Nemunaitis J, Ebbinghaus S, Rubin E, cyclines for selective activation by the tumor-associated protease Capanna T. Characterization of a novel prostate-specific anti- plasmin. J. Med. Chem. 1999;42:5277–83. gen-activated peptide-doxorubicin conjugate in patients with 64. de Groot FM, van Berkom LW, Scheeren HW. Synthesis and prostate cancer. J. Clin. Oncol. 2002;20:1874–9. biological evaluation of 20-carbamate-linked and 20-carbonate- 83. DeFeo-Jones D, Garsky VM, Wong BK, Feng DM, Bolyar T, linked prodrugs of paclitaxel: selective activation by the tumor- Haskell K, et al. A peptide-doxorubicin ‘prodrug’ activated by associated protease plasmin. J. Med. Chem. 2000;43:3093–102. prostate-specific antigen selectively kills prostate tumor cells 65. Wang MC, Valenzuela LA, Murphy GP, Chu TM. Purification of positive for prostate-specific antigen in vivo. Nat. Med. a human prostate specific antigen. Invest. Urol. 1979;17:159–63. 2000;6:1248–52. 66. Lilja H, Oldbring J, Rannevik G, Laurell CB. Seminal vesicle- 84. Denmeade SR, Jakobsen CM, Janssen S, Khan SR, Garrett ES, secreted proteins and their reactions during gelation and lique- Lilja H, et al. Prostate-specific antigen-activated thapsigargin faction of human semen. J. Clin. Invest. 1987;80:281–5. prodrug as targeted therapy for prostate cancer. J. Nat. Cancer 67. Yoshida E, Ohmura S, Sugiki M, Maruyama M, Mihara H. Inst. 2003;95:990–1000. Prostate-specific antigen activates single-chain urokinase-type 85. Jakobsen CM, Denmeade SR, Isaacs JT, Gady A, Olsen CE, plasminogen activator. Int. J. Cancer 1995;63:863–5. Christensen SB. Design, synthesis, and pharmacological evaluation 68. Killian CS, Corral DA, Kawinski E, Constantine RI. Mitogenic response of osteoblast cells to prostate-specific antigen suggests of thapsigargin analogues for targeting apoptosis to prostatic an activation of latent tgf-beta and a proteolytic modulation of cancer cells. J. Med. Chem. 2001;44:4696–703. cell adhesion receptors. Biochem. Biophys. Res. Commun. 86. Brady SF, Pawluczyk JM, Lumma PK, Feng DM, Wai JM, 1993;192:940–7. Jones R, et al. Design and synthesis of a pro-drug of 69. Cohen P, Graves HC, Peehl DM, Kamarei M, Giudice LC, targeted at treatment of prostate cancer with enhanced efficacy Rosenfeld RG. Prostate-specific antigen (psa) is an insulin-like and reduced systemic toxicity. J. Med. Chem. 2002;45:4706–15. growth factor binding protein-3 protease found in seminal 87. DeFeo-Jones D, Brady SF, Feng DM, Wong BK, Bolyar T, plasma. J. Clin. Endocrinol. Metab. 1992;75:1046–53. Haskell K, et al. A prostate-specific antigen (psa)-activated 70. Fortier AH, Nelson BJ, Grella DK, Holaday JW. Antiangio- vinblastine prodrug selectively kills psa-secreting cells in vivo. genic activity of prostate-specific antigen. J. Nat. Cancer Inst. Mol. Cancer Ther. 2002;1:451–9. 1999;91:1635–40. 88. Mhaka A, Denmeade SR, Yao W, Isaacs JT, Khan SRA. 5- 71. Heidtmann HH, Nettelbeck DM, Mingels A, Jager R, Welker fluorodeoxyuridine prodrug as targeted therapy for prostate HG, Kontermann RE. Generation of angiostatin-like fragments cancer. Bioorg. Med. Chem. Lett. 2002;12:2459–61. from plasminogen by prostate-specific antigen. Br J Cancer 89. Tang X, Xian M, Trikha M, Honn KV, Wang PG. Synthesis of 1999;81:1269–73. peptide-diazeniumdiolate conjugates: towards enzyme activated 72. Denmeade SR, Sokoll LJ, Chan DW, Khan SR, Isaacs JT. antitumor agents. Tetrahedron Lett. 2001;42:2625–9. Concentration of enzymatically active prostate-specific antigen 90. Jones GB, Mitchell MO, Weinberg JS, D’Amico AV, Bubley GJ. (psa) in the extracellular fluid of primary human prostate cancers Towards enzyme activated antiprostatic agents. Bioorg. Med. and human prostate cancer xenograft models. Prostate 2001; Chem. Lett. 2000;10:1987–9. 48:1–6. 91. Williams SA, Merchant RF, Garrett-Mayer E, Isaacs JT, 73. Castellani WJ. Cancer (prostate-specific antigen). Anal. Chem. Buckley JT, Denmeade SR. A prostate-specific antigen-activated 1995;67:399–403. channel-forming toxin as therapy for prostatic disease. 74. McCormack RT, Rittenhouse HG, Finlay JA, Sokoloff RL, J. Nat. Cancer Inst. 2007;99:376–85. Wang TJ, Wolfert RL, et al. Molecular forms of prostate-specific 92. Kumar SK, Williams SA, Isaacs JT, Denmeade SR, Khan SR. antigen and the human kallikrein gene family: a new era. Modulating paclitaxel bioavailability for targeting prostate Urology 1995;45:729–44. cancer. Bioorg. Med. Chem. 2007;15:4973–84. 75. Catalona WJ, Smith DS, Ratliff TL, Dodds KM, Coplen DE, 93. Wong BK, Defeo-Jones D, Jones RE, Garsky VM, Feng DM, Yuan JJ, et al. Measurement of prostate-specific antigen in Oliff A, et al. Psa-specific and non-psa-specific conversion of a Enzyme-mediated hydrolytic activation of prodrugs 159

psa-targeted peptide conjugate of doxorubicin to its active 108. Thurston DE. Advances in the study of pyrrolo[2,1-c][1,4]- metabolites. Drug Metab. Dispos. 2001;29:313–8. benzodiazepine (pbd) antitumor antibiotics. In: Neidle S, Waring 94. Jiang Y, Dipaola RS, Hu L. Synthesis and stereochemical MJ, editors. Mol. Aspects Anticancer Drug–DNA Interact. preference of peptide 4-aminocyclophosphamide conjugates as London: The Macmillan Press Ltd.; 1993. p. 54–88. potential prodrugs of phosphoramide mustard for activation by 109. Palmer DH, Milner AE, Kerr DJ, Young LS. Mechanism of cell prostate-specific antigen (psa). Bioorg. Med. Chem. Lett. death induced by the novel enzyme-prodrug combination, nitro- 2009;19:2587–90. reductase/cb1954, and identification of synergism with 5-fluor- 95. Bzowska A, Kulikowska E, Shugar D. Purine nucleoside ouracil. British J. Cancer 2003;89:944–50. phosphorylases: properties, functions, and clinical aspects. 110. Sirotnak FM, Chello PL, Piper JR, Montgomery JA, DeGraw Pharmacol. Ther. 2000;88:349–425. JI. Structural specificity of analog transport and binding to 96. Parker WB, King SA, Allan PW, Bennett LL, Secrist JA, dihydropholate reductase in murine tumor and normal Montgomery JA, et al. In vivo gene therapy of cancer with e. cells: relevance to therapeutic efficacy. In: Kisliuk RL, Brown Coli purine nucleoside phosphorylase. Hum. Gene Ther. GM, editors. Chem. Biol. Pteridines. New York: Elsevier; 1979. 1997;8:1637–44. p. 597–608. 97. Parker WB, Allan PW, Hassan AE, Secrist JA, Sorscher EJ, 111. Kuefner U, Lohrmann U, Montejano YD, Vitols KS, Huenne- Waud WR. Antitumor activity of 2-fluoro-20-deoxyadenosine kens FM. Carboxypeptidase-mediated release of methotrexate against tumors that express Escherichia coli purine nucleoside from methotrexate alpha-peptides. Biochemistry 1989;28:2288–97. phosphorylase. Cancer Gene Ther. 2003;10:23–9. 112. Bignami GS, Senter PD, Grothaus PG, Fischer KJ, Humphreys 98. Springer CJ, Antoniw P, Bagshawe KD, Searle F, Bisset GM, T, Wallace PM. N-(40-hydroxyphenylacetyl)palytoxin: a palytoxin Jarman M. Novel prodrugs which are activated to cytotoxic prodrug that can be activated by a monoclonal antibody- alkylating agents by carboxypeptidase g2. J. Med. Chem. penicillin g amidase conjugate. Cancer Res. 1992;52:5759–64. 1990;33:677–81. 113. Vrudhula VM, Senter PD, Fischer KJ, Wallace PM. Prodrugs of 99. Springer CJ, Niculescu-Duvaz I, Pedley RB. Novel prodrugs of doxorubicin and melphalan and their activation by a monoclonal alkylating agents derived from 2-fluoro- and 3-fluorobenzoic antibody-penicillin-g amidase conjugate. J. Med. Chem. acids for antibody-directed enzyme prodrug therapy. J. Med. 1993;36:919–23. Chem. 1994;37:2361–70. 114. Minasov G, Wang X, Shoichet BK. An ultrahigh resolution 100. Springer CJ, Dowell R, Burke PJ, Hadley E, Davis DH, Blakey structure of tem-1 beta-lactamase suggests a role for glu166 as the DC, et al. Optimization of alkylating agent prodrugs derived general base in acylation. J. Am. Chem. Soc. 2002;124:5333–40. from phenol and aniline mustards: a new clinical candidate 115. Jungheim LN, Shepherd TA. Design of antitumor prodrugs: prodrug (zd2767) for antibody-directed enzyme prodrug therapy substrates for antibody targeted enzymes. Chem. Rev. (adept). J. Med. Chem. 1995;38:5051–65. 1994;94:1553–66. 101. Dowell RI, Springer CJ, Davies DH, Hadley EM, Burke PJ, 116. Rodrigues ML, Carter P, Wirth C, Mullins S, Lee A, Blackburn Boyle FT, et al. New mustard prodrugs for antibody-directed BK. Synthesis and beta-lactamase-mediated activation of a enzyme prodrug therapy: alternatives to the amide link. J. Med. cephalosporin-taxol prodrug. Chem. Biol. 1995;2:223–7. Chem. 1996;39:1100–5. 117. Tietze LF, Herzig T, Fecher A, Haunert F, Schuberth I. Highly 102. Springer CJ. Cmda, antineoplastic prodrug. Drugs Fut. selective glycosylated prodrugs of cytostatic cc-1065 analogues 1993;18:212–5. for antibody-directed enzyme tumor therapy. Chembiochem 2001; 103. Niculescu-Duvaz I, Scanlon I, Niculescu-Duvaz D, Friedlos F, 2:758–65. Martin J, Marais R, et al. Significant differences in biological 118. Tietze LF, Schuster HJ, Krewer B, Schuberth I. Synthesis and parameters between prodrugs cleavable by carboxypeptidase g2 biological studies of different duocarmycin based glycosidic that generate 3.5-difluoro-phenol and -aniline nitrogen mustards prodrugs for their use in the antibody-directed enzyme prodrug in gene-directed enzyme prodrug therapy systems. J. Med. Chem. therapy. J. Med. Chem. 2009;52:537–43. 2004;47:2651–8. 119. Devalapally H, Navath RS, Yenamandra V, Akkinepally RR, 104. Davies LC, Friedlos F, Hedley D, Martin J, Ogilvie LM, Scanlon Devarakonda RK. Beta-galactoside prodrugs of doxorubicin IJ, et al. Novel fluorinated prodrugs for activation by carboxypep- for application in antibody directed enzyme prodrug therapy/ tidase g2 showing good in vivo antitumor activity in gene-directed prodrug monotherapy. Arch. Pharmacal Res. 2007;30:723–32. enzyme prodrug therapy. J. Med. Chem. 2005;48:5321–8. 120. Ko TP, Lin JJ, Hu CY, Hsu YH, Wang AH, Liaw SH. Crystal 105. Niculescu-Duvaz D, Niculescu-Duvaz I, Friedlos F, Martin J, structure of yeast cytosine deaminase. Insights into enzyme Spooner R, Davies L, et al. Self-immolative mechanism and evolution. J. Biol. Chem. 2003;278:19111–7. prodrugs for suicide gene therapy. J. Med. Chem. 1998;41: 121. Denny WA. Tumor-activated prodrugs–a new approach to 5297–309. cancer therapy. Cancer Invest. 2004;22:604–19. 106. Niculescu-Duvaz D, Niculescu-Duvaz I, Friedlos F, Martin J, 122. Huber BE, Austin EA, Richards CA, Davis ST, Good SS. Lehouritis P, Marais R, et al. Self-immolative nitrogen mustards Metabolism of 5-fluorocytosine to 5-fluorouracil in human prodrugs cleavable by carboxypeptidase g2 (cpg2) showing large colorectal tumor cells transduced with the cytosine deaminase cytotoxicity differentials in gdept. J. Med. Chem. 2003;46:1690–705. gene: significant antitumor effects when only a small percentage 107. Denny WA. Prodrug strategies in cancer therapy. European of tumor cells express cytosine deaminase. Proc. Nat. Acad. Sci. J. Med. Chem. 2001;36:577–95. USA 1994;91:8302–6.