<<

PharmacologicalReports Copyright©2013 2013,65,1–14 byInstituteofPharmacology ISSN1734-1140 PolishAcademyofSciences

Drugsneedtobedesignedwithdeliveryinmind TakeruHiguchi [70]

Review

Prodrugs:A challengeforthedevelopment

JolantaB.Zawilska1,2,JakubWojcieszak2,AgnieszkaB.Olejniczak1

1 InstituteofMedicalBiology,PolishAcademyofSciences,Lodowa106,PL93-232£ódŸ,Poland

2 DepartmentofPharmacodynamics,MedicalUniversityofLodz,Muszyñskiego1,PL90-151£ódŸ,Poland

Correspondence: JolantaB.Zawilska,e-mail:[email protected]

Abstract: It is estimated that about 10% of the approved worldwide can be classified as prodrugs. Prodrugs, which have no or poor bio- logical activity, are chemically modified versions of a pharmacologically active agent, which must undergo transformation in vivo to release the active drug. They are designed in order to improve the physicochemical, biopharmaceutical and/or pharmacokinetic properties of pharmacologically potent compounds. This article describes the basic functional groups that are amenable to prodrug design, and highlights the major applications of the prodrug strategy, including the ability to improve oral absorption and aqueous , increase lipophilicity, enhance active transport, as well as achieve site-selective delivery. Special emphasis is given to the role of the prodrug concept in the design of new anticancer therapies, including antibody-directed prodrug therapy (ADEPT) andgene-directedenzymeprodrugtherapy(GDEPT).

Keywords: prodrugs,drugs’ ,blood-brainbarrier,ADEPT,GDEPT

Abbreviations: AADC – decarboxylase, ACE – logically active compounds by the implementation of angiotensin converting enzyme, ADEPT – antibody-directed en- a prodrug strategy. It is estimated that currently about zyme prodrug therapy, BBB – blood-brain barrier, CNS – central 10% of worldwide marketed drugs can be classified nervous system, COMT – catechol-O-methyltransferase, GDEPT – gene-directed enzyme prodrug therapy, GPAT – gene prodrug as prodrugs. Moreover, in 2008, one third of all ap- activation therapy, LAT1 – large neutral amino acid transporter, proved small molecular weight drugs were prodrugs L-DOPA – L-dihydroxyphenylalanine, LHRH – luteinizing [35]. The first compound fulfilling the classical crite- hormone-releasing hormone, MCT – monocarboxylic acid trans- ria of a prodrug was acetanilide, introduced (under the porter, PEPT – peptide transporter, PMSA – prostate membrane name of Antifebrin®) into the medical practice by specific antigen, SDMT – sodium-dependent multivitamin trans- porter, VDEPT – virus-directed enzyme prodrug therapy Cahn and Hepp in 1867 as an antipyretic agent. In the body, acetanilide is hydroxylated (aromatic hydroxy- lation) to biologically active acetaminophen (para- cetamol), the compound endowed with both antipy- Introduction retic and analgesic activity. Acetaminophen can be also formed in the process of O-dealkylation of an- other analgesic drug – phenacetin (acetophenetidin), During the last two decades, there has been a steady introduced into clinical use in 1887 by von Mering. improvement in the physicochemical, biopharmaceu- Acetanilide and phenacetin were not originally de- tical and/or pharmacokinetic properties of pharmaco- signed as prodrugs, but their prodrug nature was de-

PharmacologicalReports, 2013,65,1–14 1 termined later on. Another example of a historical lism, to improve time profile, to increase organ/ prodrug is aspirin (acetylsalicylic acid), synthesized tissue-selectivedeliveryoftheactiveagent. in 1897 by Felix Hoffman (Bayer, Germany), and in- 3. Pharmacodynamic: to decrease toxicity and im- troduced into by Dreser in 1899 [12]. How- prove therapeutic index, to design single chemical en- ever, the prodrug concept was intentionally used for titiescombiningtwodrugs(co-drugsstrategy). the first time in the middle of the 20th century by the It should be noted that strategies to improve oral Parke-Davis company during studies on modification and achieve brain- and tumor-specific of chloramphenicol in order to improve the targeting have been the most important developments ’s bitter taste and poor solubility in water. As intheprodrugdesignduringthelastdecade. a result of this work, two prodrug forms of chloram- phenicol were synthesized: chloramphenicol sodium succinate with a good water solubility, and chloram- phenicol palmitate used in the form of suspension in children[71]. Structureandclassificationofprodrugs

There are two main classes of prodrugs: (1) carrier- Prodrugconcept linked prodrugs, and (2) bioprecursor prodrugs. In the carrier-linked prodrugs, the active molecule (the drug) is temporary linked to a carrier (also known as The basic aim of prodrug design is to mask undesir- a promoiety) through a bioreversible covalent linkage. able drug properties, such as low solubility in water or Once in the body, the carrier-linked prodrug under- lipid membranes, low target selectivity, chemical in- goes biotransformation, releasing the parent drug and stability, undesirable taste, irritation or pain after local the carrier. Ideally, the carrier should be nonimmuno- administration, presystemic metabolism and toxicity genic, easy to synthesize at a low cost, stable under [54, 70, 74]. In general, the rationale behind the use of the conditions of prodrug administration, and undergo prodrugs is to optimize the absorption, distribution, biodegradation to nonactive metabolites [35, 63, 69]. metabolism, , and unwanted toxicity (so- In so-called co-drugs (mutual prodrugs, multiple called ADMET properties) of the parent drugs [35]. prodrugs), a prodrug is formed from two pharmaco- Classically, the term prodrug, introduced in 1958 by logically active agents coupled together into a single Adrien Albert [3], relates to biologically inert deriva- molecule, and act as promoieties of each other. Exam- tives of drug molecules that undergo an enzymatic ples of co-drugs include sulfapyridine – 5-amino- and/or chemical conversion in vivo to release the salicylic acid, indomethacin – paracetamol, L-DOPA pharmacologically active parent drug. The active drug – enthacapone, gabapentin – pregabalin, 5-fluoro- is released from its inactive form before, during or , 5- after absorption of the prodrug. Some drugs are re- triamcinolone, ampicilin – sulbactram, sulfamethoxa- leased only after reaching targets of their actions [54, zole – nalidixic acid [20]. The major groups of 70]. A prodrug should increase the bioavailability and carrier-linked prodrugs are and ; other therapeutic effectiveness of a parent drug. Although groups include phosphates, carbamates, carbonates, the term prodrug is now standard, prodrugs have been oximes, imines and N-Mannich bases (Fig. 1). Bio- also referred to as reversible or bioreversible deriva- precursors do not contain a promoiety but result from tivesorbiolabiledrug-carrierconjugates. a molecular modification of the active compound it- According to Testa [74] there are three basic, over- self. The bioprecursor prodrug is transformed metab- lappingobjectivesinprodrugresearch: olically or chemically by hydration (e.g., 1. Pharmaceutical: to improve solubility, chemical such as some statins), oxidation (e.g., dexpanthenol, stability, and organoleptic properties; to decrease irri- nabumetone) or reduction (e.g., sulindac, plati- tation and/or pain after local administration, to reduce num(IV) complexes) to the active agent [30, 42, 54, problems related with the pharmaceutical technology 61,74]. oftheactiveagent. Based on the site of conversion into the pharmaco- 2. Pharmacokinetic: to improve absorption (oral and logically active agent, the prodrugs can be addition- by non-oral routes), to decrease presystemic metabo- allyclassifiedintotwogroups:

2 PharmacologicalReports, 2013,65,1–14 Prodrugs JoannaB.Zawilskaetal.

Fig. 1. Common functional groups on parent drugs applied in prodrug de- sign.Modifiedfrom[54]

• Type I – metabolized intracellularly. Type IA pro- carboxylesterases, arylesterases) which are present drugs (e.g., acyclovir, , 5-fluoro- throughout the body [35, 54]. Following the process uracil, L-DOPA, zidovudine) are metabolized at the of esterification, a desired chemical and biological cellular targets of their therapeutic actions. Type IB stability of prodrug esters can be achieved by an ap- prodrugs (e.g., carbamazepine, captopril, molsi- propriate manipulation of their electronic and steric domine, primidone) are converted to parent drugs by properties [63]. An ideal prodrug should have metabolictissues,namelybytheliver. chemical stability across a pH range, high aqueous • Type II – metabolized extracellularly. Type IIA – in solubility, exhibit good transcellular absorption, be re- the milieu of the gastrointestinal fluid (e.g., loperamide sistant to hydrolysis during the absorption phase, and oxide, sulfsalazine). Type IIB – within the circulatory undergo rapid and quantitative breakdown to yield system and/or other extracellular fluid compartments high circulating concentrations of the active compo- (e.g., aspirin, bambuterol, fosphenytoin). Type IIC – nentpostabsorption[10]. near or inside therapeutic target/cells (ADEPT, The main rationale underlying synthesis of prodrug GDEPT). Some of the prodrugs, called mixed-type prodrugs, esters is to (1) enhance lipophilicity, and thus the pas- belongtomorethanoneclass[79]. sive membrane permeability, of water soluble drugs, Esters of active agents with carboxyl, hydroxyl or (2) increase aqueous solubility (phosphate esters). To thiol functionalities, and phosphate esters of active increase the lipophilicity of hydroxyl group bearing agents with hydroxyl or functionalities are the pharmacologically active compounds, they can be most commonly used prodrugs. Approximately half acylated with aliphatic or aromatic carboxylic acids. of the prodrugs currently available on the market are Esterification of the hydroxyl group of the parent drug activated via enzymatic hydrolysis by ubiquitous es- with derivatives bearing additional terases (acetylcholinesterases, butyrylcholinesterases, functional groups, such as hydroxyl, amino or car-

PharmacologicalReports, 2013,65,1–14 3 boxyl, provides prodrug molecules with increased A hemisuccinate group can be conveniently used aqueoussolubility(hydrophilicity)[35,54]. to increase water solubility, as it contains a free car- Some esters are not suitable substrates for endogenous boxylic group, which is suitable for the formation of esterases, sulfatases or phosphatases, which, in turn, im- dissociated salts. A fine example of the utilization of pose unfavorable, too slow kinetics of the prodrug hy- hemisuccinate esters is cinazepam, a novel benzodi- drolysis. To optimize the rate of enzymatic hydrolysis, azepine anxiolytic drug suitable for intravenous injec- knowledge on the mechanisms of the specific processes tions[60]. involved is highly desirable. An increase in the rate of al- kaline hydrolysis can be achieved by the attachment of Prodrugswithincreasedlipidsolubility electron-withdrawing groups, while in the case of acidic hydrolysis, tethering of electron-donating groups to the In order to improve lipophilicity, and thus passive carboxylate part of esters is recommended. If the esters transport through biological membranes, compounds are too reactive, bulky substituents causing steric hin- containing polar or ionizable groups can be converted drance to hydrolysis or esters of long-chain fatty acids into ester prodrugs [26, 33, 36, 44, 54]. Examples of can be employed [71]. ester prodrugs with improved lipophilicity designed fororaladministrationarelistedinTable2. Prodrugs with increased lipophilicity are also de- signed for topical administration. For example, esters Prodrugapproachesforenhancing of ketolac (a non-steroidal anti-inflammatory drug with administration,permeability,absorption, potent analgesic activity) and fatty acids (stearic, lino- anddistributionofdrugs leic, oleic) allow the drug to accumulate in the skin with concomitant low skin permeation, leading to in- creased therapeutic efficiency and reduced side ef- Prodrugswithincreasedaqueoussolubility fects of the parent drug [13]. Additional examples are latanoprost and travoprost, prodrugs of prostaglandin

Poor aqueous solubility is considered as a serious F2a (PGF2a) analogs, applied as eye drops for the problem limiting the therapeutic use of numerous treatment of glaucoma. Naturally occurring prosta- drugs and drug candidates. Among various strategies glandins, including PGF2a, are molecules which are used to overcome this drawback is the prodrug ap- relatively polar and hydrophilic due to their carbox- proach. One frequently employed means of improving ylic acid moiety and several hydroxyl groups. They the aqueous solubility of a drug is by the use of esters penetrate through biological membranes poorly. This and amides of phosphoric acid, due to the ionic nature problem has been solved by a modification of the of the phosphate group. It should be noted that phos- chemical structure of PGF2a analogs. Latanoprost phate derivatives display high chemical stability, (Xalatan®) and travoprost (Travatan®), isopropyl es- often even higher than the parent compound. Under ters of latanoprost acid and travoprost acid, respec- physiological conditions phosphate prodrugs undergo tively, penetrate easily through the corneal epithelium rapid biotransformation by endogenous phosphatases, due to their increased lipophilicity, where they are such as , of the intestine, plasma, then hydrolyzed to active carboxylic acids (Fig. 2). and the liver [15, 26, 44, 48, 54, 65, 74]. Examples of The free acids of latanoprost and travoprost activate phosphoric acid ester prodrugs and their correspond- prostanoid FP receptors and reduce intraocular pres- ingparentdrugsareshowninTable1. sure by enhancing the uveoscleral and the trabecular Another approach to increase aqueous solubility is meshworkoutflowpathways[9]. esterification with amino acids. Examples of such ® prodrugs are valacyclovir (Valtrex ) and valgancy- ProdrugsassubstratesforGImembrane ® clovir (Valcyte ), which are valine esters of the antivi- transporters ral drugs acyclovir and gancyclovir, respectively [53, 67]. Both prodrugs are also good substrates of small Gabapentin enacarbil ([(±)-1-([a-isobutanoyloxyeth- peptide transporters (PEPT1) present in the intestine oxy)carbonyl]aminomethyl)-1-cyclohexane acetic acid; epithelium, a property that further increases their Horizant®), a prodrug of gabapentin (1-amino- bioavailability[32,48,77]. methyl)-cyclohexane acetic acid), is a substrate for

4 PharmacologicalReports, 2013,65,1–14 Prodrugs JoannaB.Zawilskaetal.

Tab.1. Phosphateestersasprodrugsforimprovedaqueoussolubility.Modifiedfrom[54]

A. Fororaladministration

Prodrugname;functionalgroup Structure Action Fosamprenavir(Telzir®);phosphateester Bioconvertedbyalkaline ofamprenavir phosphatasestoamprenavir, aHIVproteaseinhibitor. Antiviral,HIVinfections.

Fludarabinephosphate(Fludara®);phosphate NH Bioconvertedbyalkalinephosphatases N esterof N tofludarabine.Fludarabineundergoes transformationto2-fluoro-9-b-D-arabino- N N F OH furanosyladenine,whichafteruptakeintocells OH O P isconvertedtoactive2-fluoro-9-b-D-arabino- O O furanosyladenine5’-triphosphate.Antiviral.

HO OH

® O Estramustinephosphate(Emcyt );phosphate – + Bioconvertedbyalkalinephosphatases O P O Na esterofestramustine O–Na+ toestramustine,whichisfurthertransformed intoestromustine.Antimitotic.

O Cl N O

Cl

Prednisolonephosphate;phosphateesterof O Bioconvertedbyalkalinephosphatases ® prednisolone(Inflamase ,OrapredODT®) O – + toprednisolone.Anti-inflammatory,antiallergic. O PO Na O–Na+ OH OH

O

B. Forparenteraladministration Fosfluconazole(Procif®);phosphateester N O Bioconvertedbyalkalinephosphatases PO–Na+ offluconazole N – tofluconazole.Antifungal. N O O Na+

F N N

N

F

Fosphenytoin Rapidlybioconvertedbyalkalinephosphatases (Pro-dilatin®,Pro-epanutin®,Cerebryx®); tophenytoin.Anticonvulsant. phosphateesterofphenytoin O O – + O P O Na N – + N O Na H O Propofolphosphate;phosphateester O Rapidlybioconvertedbyalkalinephosphatases ofpropofol Na+O– P O–Na+ topropofol.Anaesthetic. O

PharmacologicalReports, 2013,65,1–14 5 Tab.2. Prodrugswithimprovedlipophicilityfororaladministration.Modifiedfrom[54]

Prodrugname;functionalgroup Structure Action

Enalapril(Innovace®,Vasotec®,Renitc®), O O Intheliverbioconvertedbyesterasesto monoethylesterofenalaprilat enalaprilat,anangiotensin-convertingenzyme N inhibitor.Usedinthetreatmentofhypertension, NH ischemicheartdisease. O COOH

® Oseltamivir(Tamiflu ),ethylester O Bioconvertedbycarboxylesterase-1to ofoseltamivircarboxylate oseltamivircarboxylate–selectiveinhibitorof O O viralneuroamidaseglycoproteintypeAandB. Antiviral(anti-influenza). HN

O NH2 Famciclovir(Famvir®),dimethylester Bioconvertedbyesterasesandaldehyde ofpenciclovir N oxidasetopenciclovir–inhibitorof Herpes N O O DNAsynthesis.Antiviral.

H N N N

O

O

Adefovirdipivoxil(Viread®), NH Bioconvertedbyesterasesand

bis-(pivaloyloxymethyl)esterofadenofovir N phosphodiesterasestotenofovir.In N O lymphocytesTtenofovirinconvertedtoactive

N metabolite,tenofovirdiphosphate,aninhibitor N O ofHIVvirusreversetranscriptase.Antiviral O O O O P (anti-HIV). O O O

O Pivampicillin(Alphacyllin®),pivaloylmethyl Bioconvertedbyesterasestoampicillin esterofampicillin (b-lactamantibiotic).

monocarboxylic acid transporter-1 (MCT-1) and Several groups of prodrugs, such as ACE inhibitors sodium-dependent multivitamin transporter (SMVT), (e.g., captopril, enalapril, fosinopril), antiviral (e.g., distributed throughout the intestine. Gabapentin en- valcyclovir and valgancyclovir), and anticancer (e.g., acarbil is stable at physiological pH and rapidly con- ester analogs of fluxiridine and ) are sub- verted into gabapentin. In vitro and in vivo studies stratesforPEPT1[32,48,58,66,76,77]. have demonstrated that gabapentin enacarbil is en- dowed with better absorption, bioavailability, and pharmacokinetic properties compared with gabapen- tin [78]. Gabapentin is approved for the treatment of ProdrugapproachesfortheCNSdelivery epilepsy and postherpetic neuralgia. Phase II and III clinical studies indicate that gabapentin enacarbil ad- One of the major difficulties in the development of ministered in the form of the prodrug could be effec- drugs acting at the central nervous system (CNS) is tive in the treatment of restless leg syndrome [80]. the inability of many therapeutical compounds to

6 PharmacologicalReports, 2013,65,1–14 Prodrugs JoannaB.Zawilskaetal.

HO HO

COOCH(CH3)2 COOH Fig. 2. Chemical of prosta- glandin PGF2a analog prodrugs de- signed for improved ophthalmic deliv- HO HO ery.Modifiedfrom[9] OH OH

Latanoprost Latanoprost acid

HO HO

COOCH(CH3)2 COOH O O HO OH HO OH CF3 CF3 Travoprost Travoprost acid

cross the blood-brain barrier (BBB). The BBB is (GLUT1), peptide transporters (PEPT), monocarbox- formed by endothelial cells of brain microvessels con- ylic acid transporters (MCT), organic cation trans- nected by extensive tight junctions. Combined with porters (OCT), organic anion transporters (OAT), an absence of fenestrae and an extremely low pinocy- concentrative nucleoside and transporters totic activity, transport of molecules across the BBB (CNT) [29, 31]. It is suggested that prodrugs with is achieved either through diffusion as a passive pro- a very close structural resemblance to endogenous cess, or with the aid of special carrier systems involv- substrates of the BBB influx transporters will be rec- ing intrinsic transporter localized on the lu- ognized by them and transported across the BBB into minal (blood side) and abluminal (brain side) sides of the brain, where the release of an active drug will take the epithelial cells. The BBB is necessary to provide place. L-DOPA, a dopamine prodrug used in the treat- an optimal chemical environment for brain function. ment of Parkinson’s disease, is an example of com- It restricts the passage of blood-borne drugs, neuro- pounds transported into the brain with the aid of in- toxic substances and peripheral immune cells from flux transporters. Although dopamine is unable to entering the brain, while selectively facilitating the cross the BBB due to its hydrophilic nature, L-DOPA, transport of nutrients into the brain. In addition, it pre- its amino acid analog and precursor, crosses the BBB vents central and neuromodulators via LAT1. The drug is then decarboxylated by amino fromreachingthecirculatorysystem[1]. acid decarboxylase (AADC) to dopamine. Addition- Three basic prodrug strategies have been used to ally, catechol-O-methyltransferase (COMT) converts facilitate the transport of drugs into the CNS: (1) in- L-DOPA to 3-O-methyl DOPA. To minimize the me- creasing passive diffusion by masking polar groups tabolism of L-DOPA outside the CNS and to increase on the parent drug (so-called lipidization of mole- its half-life time, the drug is given in combination cules), (2) increasing carrier-mediated or receptor- with peripheral inhibitors of AADC (carbidopa and mediated transport through the BBB, and (3) decreas- benserazide) and COMT (tolcapone and entacapone) ing the active efflux of the drug from the brain into [23,39]. the blood. In order to achieve therapeutically effective doses in the CNS, the process of prodrug biotransfor- mation should be slow in the peripheral tissues but fast in the brain itself. Furthermore, taking into ac- Howcanprodrugsimprovetargetability count the anatomical complexity of the CNS, the bio- andefficacyofchemotherapeutic transformation should be limited to selected brain agents? structures[50,51,55]. Several specific endogenous influx transporters have been identified at the brain capillary endothe- is a primary treatment for . During lium which forms the BBB, including large neutral the last several decades major progress has been made in amino acid transporters (LAT1), glucose transporters the field of chemotherapy of various . The vast

PharmacologicalReports, 2013,65,1–14 7 majority of anticancer drugs currently in use exert rylase in tumors to highly cytotoxic 5’-fluorouracil their oncostatic actions by inhibiting proliferation or [47]. by arresting cycle at a certain phase. As oncos- Anticancer prodrugs can be designed to target spe- tatic drugs are endowed with poor selectivity, they af- cific molecules (, peptide transporters, anti- fect not only neoplastic cells but also rapidly prolifer- gens) that are overexpressed in tumor cells in com- ating normal cells, such as, to name just a few, bone parison to normal cells. The new promising chemo- marrow, gut epithelia, hair follicles, lymphatic cells therapeuticprodrugsinclude: and gametes. The lack of selectivity of anticancer 1. Enzyme-activated prodrugs – antibody-directed en- drugs, and associated toxicity, hampers their effec- zyme prodrug therapy (ADEPT) and gene-directed tiveness and long term use. Hence, not surprisingly, enzymeprodrugtherapy(GDEPT). thereisanurgentneedtoimprovetheirselectivity. 2. Targeting-ligand conjugated prodrugs – antibody- One of the promising approaches to achieve this drug conjugates, peptide-drug conjugates, aptamer- goal is prodrug technology. An anticancer prodrug drugconjugates,andfolicacid-drugconjugates. should be transported to neoplastic cells, where it will 3.Enzyme-cleavableprodrugs. undergo transformation to a cytotoxic parent drug by 4.Membranetransporter-associatedprodrugs. native or recombinant enzymes [45]. 5.Polymericprodrugs. (5’-deoxy-5-fluorocytidine carbamate; Xeloda®), a tri- ple prodrug of 5-fluorouracil approved for the therapy Enzyme-activatedprodrugs of solid tumors including breast cancer and , may be considered a pioneer of chemotherapy One approach toward improving the specificity of prodrugs [37, 43]. Following oral administration, chemotherapy is enzyme-activated prodrug therapy in capecitabine is rapidly and extensively absorbed. After which a non-toxic drug is converted into a cytotoxic adsorption, capecitabine is first metabolized by he- agent by an enzyme either extracellularly, at a site of patic carboxyesterases, CES1 and CES2. 5’-Deoxy- tumor (ADEPT), or intracellularly in neoplastic cells 5-fluorocytidine formed is then converted to 5’-de- (GDEPT). Both strategies utilize precursors of two oxy-5-fluorouridine by cytidine deaminase in the liver groups of oncostatic drugs, i.e., and and tumor tissues, and finally by phospho- alkylating agents [59]. GDEPT is also known in the literature under other names, such as suicide gene therapy (SGT), virus-directed enzyme prodrug ther- Tab.3. Examplesofenzyme-activatedprodrugs apy (VDEPT), and gene prodrug activation therapy (GPAT) [4, 64]. Examples of enzyme-activated pro- Antibody-directedenzymeprodrugtherapy(ADEPT) drugsarelistedinTable3. The principle of ADEPT is to use an antibody di- Antibody Prodrug Drug Tumortarget rected at a tumor-associated antigen which localizes L6 MitomycinC MitomycinC Lungadenocarci- the enzyme in the vicinity of the tumor. A non-toxic phosphate noma prodrug, a substrate for the enzyme, is then given in- BW413 phosphate Etoposide Coloncarcinoma travenously and converted to a cytotoxic drug only at L6 Doxorubicin Lungadenocarci- the tumor site where the enzyme is localized, result- phosphate noma ing in tumor cell death (Fig. 3). For ADEPT to be ef- fective and safe the following criteria should be ful- Gene-directedprodrugtherapy(GDEPT) filled: Enzyme Prodrug Drug • The prodrug should be easily dissolved in water, and marginallytakenupbycells. Cytochromep450 Cyclophosphamide, Phosphamide • The drug should be efficiently and rapidly trans- mustard,acrolein portedintocancercells. deaminase 5-Fluorocytosine 5-Fluorouridine 5-Fluorouracyl • The antibody-enzyme complex should bind selec- Nitroreductase 5-(Aziridin-1-yl)-2,4- tivelytocancercells. dinitrobenzamide 5-(Aziridin-1-yl)-4- •Theenzymeshouldhavehighintrinsicactivity. hydroxylamino-2- • The enzyme cannot be inactivated by a prodrug or nitrobenzamide drug.

8 PharmacologicalReports, 2013,65,1–14 Prodrugs JoannaB.Zawilskaetal.

ADEPT GDEPT

Tumor cell Tumor specific regulatory element Gene encoding enzyme PRODRUG CYTOTOXIC DRUG

Transgene expression

Gene expression vector is Antibody Prodrug activating enzyme directly delivered to tumor cell Enzyme

CYTOTOXIC PRODRUG PRODRUG DRUG

Administration of enzyme-antibody conjugate Cell death

Fig. 3. Schematic diagram illustrating principles of the antibody-directed enzyme prodrug therapy (ADEPT) and gene-directed enzyme prodrug therapy (GDEPT). In the ADEPT approach, the activating enzyme is specifically delivered to tumor cells using a tumor-specific antibody, followed by administration of a prodrug. The prodrug is then activated by the conjugated enzyme to the active cytotoxic parent drug. In GDEPT, a gene expres- sion vector encoding an activating enzyme is delivered to tumor cells. Next, a nontoxic prodrug is administered systematically and converted by the enzyme to the toxic parent drug. Modified from [55]

•Enzymeimmunogenicityshouldbelow. cleoside phosphorylase (PNP), and nitroreductase Endogenously expressed human enzymes cannot (NR) from Escherichia coli. The second category be utilized for ADEPT, since the prodrug will be con- consists of enzymes of human origin that are absent or verted to a cytotoxic compound not only in the vicin- are expressed only at low concentrations in tumor ity of the tumor, but also at sites where endogenous cells, such as deoxycytidine kinase (dCK) and cyto- enzyme is expressed, causing systemic toxicity. Non- chrome P450. The main disadvantage of nonmammal- human enzymes which are currently used in ADEPT ian enzymes is their potential immunogenicity. En- technology could, however, induce immune re- zymes of the second category are unlikely to induce sponses, which, in turn, will limit their therapeutic immune responses, but their use will in most cases usefulness[2,5,75]. lead to some prodrug activation in normal cells [4, 6, GDEPT, like ADEPT, is a two-step process. In the 17]. Many viral and non viral gene delivery systems first step, the gene for a foreign enzyme is delivered have been employed to deliver the therapeutic DNA to tumor cells. In the second step, a non-toxic agent is into cancer cells. The most widely used are retrovirus administered systematically and converted by the en- vectors. To increase tumor selectivity of GDEPT, zyme to its cytotoxic metabolite (Fig. 3) (e.g., [40, a tumor-specific regulatory element is incorporated in 46]). Enzymes used in GDEPT catalyze specific reac- the gene expression vector. An example of such ap- tions that are not catalyzed by endogenous enzymes, proach is using prostate specific antigen (PSA)-spe- and should be delivered in sufficiently high amounts cific promoter or the prostate membrane specific anti- to result in prodrug conversion. They can be divided gen (PMSA)-specific promoter/enhancer to target into two categories. The first comprises foreign en- GDEPT virus vector to prostate cancer cells. An im- zymes of nonmammalian origin (viral, bacterial, portant feature of GDEPT is so-called bystander or yeast), with or without human homologues. Examples neighboring cell killing effect, which can be divided are thymidine kinase (TK) from Herpes simplex or into two categories: (1) local – the active toxic drug Varicella zoster viruses, cytosine deaminase (CD), spreads by passive, gap-junction independent diffu- uracil phosphoribosyl transferase (UPRT), purine nu- sion to surrounding cells, including those that do not

PharmacologicalReports, 2013,65,1–14 9 express the enzyme, and kills them; and (2) distant – conjugated to a chemotherapic agent either directly or dying cells can stimulate systemic anticancer effects by a linker. Peptides have many advantages over anti- through the host immune responses mediated by natural bodies as tumor-targeting moieties: particularly small killer cells, T cells and macrophages [19, 34]. A strong molecular weight, very good cell permeability, and bystander effect could be seen even when only 1 in ease of production [45]. So far, promising results have 100–1000 cells expresses the transgene [27]. Prodrugs been obtained with luteinizing hormone-releasing for GDEPT must be considerably less cytotoxic than hormone (LHRH) conjugated to via their corresponding active drugs, and must be suitable polyethylene glycol (PEG) as the spacer. LHRH is substrates for the activating enzyme under physiologi- used as a targeting moiety to LHRH receptors that are cal conditions. In addition, they should have good overexpressed in the plasma membrane of breast, pharmacological and pharmacokinetic properties, be ovarian, and prostate cancer cells and are not ex- chemically stable and diffuse readily in the tumor in- presseddetectablyinnormalvisceralorgans[22]. terstitium. The formed cytotoxic drug should be me- Aptamers are single-stranded DNA or RNA oligo- tabolically stable, and its retention time must be long that bind to specific target molecules with enough to achieve desired therapeutic effects [40, 68]. high specificity and affinity. In general, they are sta- ble, nonimmunogenic, and easy to synthesize and im- mobilize on chemical devices. Several types of ap- Targeting-ligandconjugatedprodrugs tamers have been selected against cancer-related pro- teins, such as -derived growth factor (PDGF), Tumor-specific monoclonal antibodies (or fragments vascular endothelium growth factor (VEGF), type 3 of antibodies) are conjugated to oncostatic drugs such receptor for epidermal growth factor (HER3), nuclear as , , and vinca alka- factor kB (NFkB), tenascin-C or PSMA, by an in vi- loids. The antibody delivers the therapeutic agent to tro process known as cell systematic evolution of tumor cells. After reaching its target, the conjugate is ligands by exponential enrichment (cell-SELEX) internalized through a receptor-mediated pinocytosis, [82]. There are now ongoing studies on aptamer- and the pharmacologically active compound is re- cytotoxic agent conjugates targeted against human leased in the cell. Tumor-specific antigens include, acute lymphoblastic cells, prostate and among others, CD19, CD22, CD33, CD56, MUC16, breastcancercells[21,41,81,83]. TAG-72, PMSA, epidermal growth factor receptor-2 (HER-2) and aV integrin. It should be emphasized Prodrugsinanticancerphotodynamictherapy that some of these antigens are preferably expressed (PDT) at high levels in specific cancer types, e.g., MUC16 in ovarian cancer, PMSA in prostate cancer, TAG-72 in (PDT) is a clinically approved human adenocarcinomas [45]. Major problems related method for the treatment of several types of cancers. with this prodrug strategy that still have to be resolved The advantages of PDT over other conventional che- include: immunogenicity, selectivity and permeability motherapies are its low systemic toxicity and ability into the solid tumors [45, 72, 73]. Although at present, to selectively destroy tumors accessible to light. PDT around twenty antibody-drug conjugates are in clini- combines a drug with photosensitizing properties, cal trials, so far, only one such prodrug, i.e., Mylo- a or photosensitizing agent, with targ® (Weyth), which consists of calicheamicin linked a specific type of light to kill the photosentitized cells. to the human anti-CD33 monoclonal antibody (gen- Following administration of the photosensitizer and tuzumab) via a hydrolyzable bifunctional linker has its accumulation in a malignant tissue, light of the ap- been approved by FDA for the treatment of cancer propriate wavelength is directed onto the tissue. The () [62]. However, due to se- subsequent activation of the photosensitizer by light vere side effects, the drug was withdrawn from the results in the transfer of its energy to molecular oxy- marketin2010[45]. gen. This, in turn, generates reactive oxygen , Peptide-conjugated prodrugs for cancer therapy responsibleforcytotoxicityofthecells[38]. utilize peptide ligands designed to bind with a tumor- derivatives are well known photosensi- specific antigen or a peptide transporter which is tizers but their therapeutical use is limited by a low overexpressed in neoplastic cells. These ligands are bioavailability and difficulties in administration [49].

10 PharmacologicalReports, 2013,65,1–14 Prodrugs JoannaB.Zawilskaetal.

To overcome these drawbacks, a prodrug strategy Concludingremarks based on the administration of 5- (ALA) was introduced in the 1990s [14, 25, 52]. ALA is an endogenous substrate used in the multistep syn- The prodrug strategy is one of the most promising ap- thesis of . In vivo, catalytic synthesis of proaches to enhance the therapeutic efficacy and/or ALA is the slowest step of the biosynthetic reduce the adverse effects of the pharmacologically pathway, thus intake of exogenous ALA leads to accu- active agents via different mechanisms, including in- mulation of protoporphirin IX in cancer cells. Fur- creased solubility, stability, improved permeability thermore, ALA can be administered by various routes, and bioavailability, prolonged biological half-life e.g., orally, intravenously or transdermally [49]. To time, and tissue-targeted delivery. Hence, not surpris- increase ALA bioavailability even more, researchers ingly, prodrugs are becoming an integral part of the designed cascade prodrugs: double prodrugs, prodrug paradigm. Their importance is sup- of a prodrug. Lipophilicity, and thus bioavailability of ported by the increasing percentage of approved new ALA, can be improved by esterification with long drug entities that are, in fact, prodrugs. Despite the re- chain such as hexanol or octanol [28], or by markable progress made in the field of prodrug de- synthesis of dendritic derivatives [7, 8, 24, 56] which sign, more studies are clearly needed, especially at break down in vivo to the parent compound. Another early stages of the drug discovery, for prodrugs to approach is glycosidation of ALA, which gives achieve the desired state of art and take their place in a more cell-friendly derivative, free from potentially modernpharmacotherapy. toxic carriers [18]. A further modification of the ALA molecule are short peptide conjugates, an important advantage of which is that enzymes and peptide trans- Acknowledgment: porters may be upregulated in cancer cells, thus in- SupportedbytheEuropeanRegionalDevelopmentFundunder theOperationalProgramInnovativeEconomy,grantPOIG.01.01.02 creasing the efficiency of therapy and reducing the re- -10-107/09. quiredconcentrationofthedrug[11,57].

Ironchelatorsincancertherapy References: Iron plays the role of both cancer initiator and pro- moter. Free iron ions participate in the Haber-Weiss 1. AbbottNJ,PatabendigeAA,DolmanDE,YusofSR, and Fenton reactions leading to the formation of reac- BegleyDJ:Structureandfunctionoftheblood-brain tive oxygen species (ROS) that play an important role barrier.NeurobiolDis,2010,37,13–25. 2. in carcinogenesis. Moreover, iron availability is nec- AfsharS,AsaiT,MorrisonSL,HumanizedADEPT comprisedofanengineeredhumanpurinenucleoside essary for rapidly proliferating tumors to convert pro- phosphorylaseandatumortargetingpeptidefortreat- toporphyrin IX into hemoglobin. Therefore, reducing mentofcancer.MolCancerTher,2009,8,185–193. the pool of free iron ions using chelators inhibits ROS 3. AlbertA:Chemicalaspectsofselectivetoxicity.Nature, synthesis and facilitates protoporphyrin IX accumula- 1958,182,421–422. 4. AltanerC:Prodrugcancergenetherapy.CancerLett, tion. This, in turn, increases the efficiency of ALA 2008,270,191–201 and its prodrugs. As iron bound within chelators can 5. BagshaweKD:Targeting:theADEPT storysofar.Curr also produce ROS to some degree, chelators should DrugTargets,2009,10,152–157. not compete with proteins naturally binding iron in 6. BaldwinA,HuangZ,JounaidiY,WaxmanDJ:Identifi- vivo [49]. This particular goal has been achieved by cationofnovelenzyme-prodrugcombinationsforusein cytochromeP450-basedgenetherapyforcancer.Arch Charkoudian [16], who designed a prochelator by BiochemBiophys,2003,409,197–206. condensing isonicotinic acid hydrazide with pinacol 7. BattahSH,CheeCE,NakanishiH,GerscherS, ester of boronic acid. The synthesized compound is MacRobertAJ, Edwards C: Synthesis and biological inactive in situ and does not compete with iron bind- studies of 5-aminolevulinic acid-containing dendrimers ing proteins. On the other hand, hydrogen peroxide for photodynamic therapy. Bioconjugate Chem, 2001, 12, 980–988. formed during oxidative stress activates the prochela- 8. BattahS,O’NeillS,EdwardsC,BalaratnamS,Dobbin tor to its active form: salicyl aldehyde isonicotinoyl P,Mac-RobertAJ:Enhancedporphyrinaccumulationus- hydrazone. ingdendriticderivativesof5-aminolaevulinicacidfor

PharmacologicalReports, 2013,65,1–14 11 photodynamictherapy:an invitro study.IntJBiochem 25. DivarisDX,KennedyJC,PottierRH:Phototoxicdam- CellBiol,2006,38,1382–1392. agetosebaceousglandsandhairfolliclesofmiceafter 9. BeanGW,CamrasCB:Commerciallyavailableprosta- systemicadministrationof5-aminolevulinicacidcorre- glandinanalogsforthereductionofintraocularpressure: lateswithlocalizedprotoporphyrinIXfluorescence. similaritiesanddifferences.SurvOphthalmol,2008,53, AmJPathol,1990,136,891–897. Suppl1,S69–84. 26. FasinuP,PillayV,NdesendoVMK,deToitLC,Choon- 10. BeaumontK,WebsterR,GardnerI,DackK,Designof araYE:Diverseapproachesfortheenhancementoforal esterprodrugstoenhanceoralabsorptionofpoorlyper- drugbioavailability.BiopharmDrugDispos,2011,32, meablecompounds:challengestothediscoveryscientist. 185–209. CurrDrugMetab,2003,4,461–485. 27. GadiVK,AlexanderSD,KudlowJE,AllanP,Parker 11. BergerY:Evaluationofdipeptide-derivativesof WB,SorscherEJ: Invivo sensitizationofovariantumors 5-aminolevulinicacidasprecursorsfor tochemotherapybyexpressionof E.coli purinenucleo- inphotodynamictherapy.BioorgMedChem,2003,11, sidephosphorylaseinasmallfractionofcells.Gene 1343–1351. Ther,2000,7,1738–1743. 28. 12. BertoliniA,FerreriA,GuerzoniS,TacchiR,LeoneS: GaullierJ,BergK,PengQ,AnholtH,SelboP,MaL, Paracetamol:Newvistaofanolddrug.CNSDrugRev, MoanJ:Useof5-aminolevulinicacidesterstoimprove 2006,12,250–275. photodynamictherapyoncellsinculture.JCancerRes, 13. BhandariKH,NewaM,YoonSI,KimJS,JangKY, 1997,57,1481–1486. 29. KimJA,YooBKetal.:Evaluationofphysicochemical GirardinF:Membranetransporterproteins:achallenge properties,skinpermeationandaccumulationprofiles forCNSdrugdevelopment.DialoguesClinNeurosci, ofketorolacfattyesterprodrugs.BiolPharmBull,2007, 2006,8,311–321. 30. 30,2211–2216. GrafN,LippardSJL:Redoxactivationofmetal-based 14. BissonnetteR,TremblayJ,JuzenasP,BoushiraM, prodrugsasastrategyfordrugdelivery.AdvDrugDeliv LuiH:Systemicphotodynamictherapywithaminolevu- Rev,2012,64,993–1004. 31. linicacidinducesinlesionalT lymphocytesof GyntherM,LaineK,RopponenJ,LeppänenJ,Mannila psoriaticplaques.JInvestDermatol,2002,119,77–83. A,NevalainenT,SavolainenJetal.:Largeneutralamino acidtransporterenablesbraindrugdeliveryviaprodrugs. 15. BobeckDR,SchinaziRF,CoatsSJ:Advancesinnucleo- JMedChem,2008,51,932–936. sidemonophosphateprodrugsasanti-HCV agents. 32. HatanakaT,HaramuraM,FeiYJ,MiyauchiS,Bridges AntivirTher,2010,15,935–950. CC,GanapathyPS,SmithSBetal.:Transportofamino 16. CharkoudianLK,PhamDM,FranzKJ:A prochelator acid-basedprodrugsbytheNa+-andCl–-coupledamino triggeredbyhydrogenperoxideinhibitsiron-promoted acidtransporterATB0,+ andexpressionofthetransporter hydroxylradicalformation.JAmChemSoc,2006,128, intissuesamenablefordrugdelivery.JPharmacolExp 12424–12425. Ther,2004,308,1138–1147. 17. ChenL,WaxmanDJ:CytochromeP450gene-directed 33. HeG,MassarellaJ,WardP:Clinical enzymeprodrugtherapy(GDEPT)forcancer.Curr oftheprodrugoseltamiviranditsactivemetabolite PharmDes,2002,8,1405–1416. Ro64-0802.ClinPharmacokinet,1999,37,471–484. 18. ChenX,WuB,WangPG:Glucuronidesinanti-cancer 34. HughesBW,KingSA,AllanPW,ParkerWB,Sorscher therapy.CurrMedChemAnti-cancerAgents,2003,3, EJ:Celltocellcontactisnotrequiredforbystandercell 139–150. killingby Escherichiacoli purinenucleosidephosphory- 19. DachsGU,HuntMA ,SyddallS,SingletonDC,Patter- lase.JBiolChem,1998,273,2322–2328. sonAV:Bystanderornobystanderforgenedirecteden- 35. HuttunenKM,RaunioH,RautioJ:Prodrugs–fromser- zymeprodrugtherapy.Molecules,2009,14,4517–4545. endipitytorationaldesign.PharmacolRev,2011,63, 20. DasN,DhanawatM,DashB,NagarwalRC,Shrivastava 750–771. SK:Codrug:Anefficientapproachfordrugoptimiza- 36. JanaS,MandlekarS,MaratheP:Prodrugdesigntoim- tion.EurJPharmSci,2010,41,571–588. provepharmacokineticanddrugdeliveryproperties: 21. DharS,GuFX,LangerR,FarokhzadOC,LippardSJ: challengestothediscoveryscientists.CurrMedChem, Targeteddeliveryoftoprostatecancercellsby 2010,17,3874–3908. aptamerfunctionalizedPt(IV)prodrug-PLGA-PEG 37. JohnstonPG,KayeS:Capecitabine:anovelagentfor nanoparticles.ProcNatlAcadSciUSA,2008,105, thetreatmentofsolidtumors.AnticancerDrugs, 2001, 17356–17361. 12,639–646. 22. DharapSS,WangY,ChandnaP,KhandareJJ,QiuB, 38. JuarranzA,JaénP,Sanz-RodríguezF,CuevasJ, GunaseelanS,SinkoPJetal.:Tumor-specifictargeting GonzálezS:Photodynamictherapyofcancer.Basic ofananticancerdrugdeliverysystembyLHRHpeptide. principlesandapplications.ClinTranslOncol,2008,10, ProcNatlAcadSciUSA,2005,102,12962–12967. 148–154. 23. DiStefanoA,SozioP,CerasaLS:Antiparkinsonian 39. KaakkolaS:Clinicalpharmacology,therapeuticuseand prodrugs.Molecules,2008,13,46–68. potentialofCOMT inhibitorsinParkinson’sdisease. 24. DiVenosaGM,CasasAG,BattahS,DobbinP,Fukuda Drugs,2000,59,1233–1250. H,MacrobertAJ,BatlleA:Investigationofanovelden- 40. KhatriA,RussellPJ:Targeted,gene-directedenzyme driticderivativeof5-aminolaevulinicacidforphotody- prodrugtherapiestotacklediversityandaggressionof namictherapy.IntJBiochemCellBiol,2006,38,82–91. latestageprostatecancer.DiscovMed,2007,7,39–45.

12 PharmacologicalReports, 2013,65,1–14 Prodrugs JoannaB.Zawilskaetal.

41. KimE,JungY,ChoiH,YangJ,SuhJS,HuhYM,Kim 57. RogersLM,McGivernPG,ButlerAR,MacRobertAJ, K,HaamS:Prostatecancercelldeathproducedbythe EgglestonIM:Anefficientsynthesisof5-amino- co-deliveryofBcl-xL shRNA anddoxorubicinusingan laevulinicacid(ala)-containingpeptidesforuseinphoto- aptamer-conjugatedpolyplex.Biomaterials,2010,31, dynamictherapy.Tetrahedron,2005,61,6951–6918. 4592–4599. 58. SalvettiA:NewerACEinhibitors.A lookatthefuture. 42. KokilGR,RewatkarPV:Bioprecursorprodrugs:mo- Drugs,1990,40,800–828. lecularmodificationoftheactiveprinciple.MiniRev 59. SchellmannN,DeckertPM,BachranD,FuchsH, MedChem,2010,10,1316–1330. BachranC:Targetedenzymeprodrugtherapies.Mini 43. KoukourakisGV,KoulouliasV,KoukourakisMJ, RevMedChem,2010,10,887–904. ZachariasGA,ZabatisH,KouvarisJ:Efficacyofthe 60. SchukinSI,ZinkovskyVG,ZhukOV:Eliminationkinet- oralfluorouracilpro-drugcapecitabineincancertreat- icsofthenovelprodrugcinazepampossessingpsycho- ment:areview.Molecules,2008,13,1897–1922. tropicactivityinmice.PharmacolRep,2011,63, 44. LiF,MaagH,AlfredsonT:Prodrugsofnucleosideana- 1093–1100. loguesforimprovedoralabsorptionandtissuetargeting. 61. ShiY,LiuS-A,KerwoodDJ,GoodismanJ,Dabrowiak JPharmSci,2008,97,1109–1134. JC:Pt(IV)complexesasprodrugsforcisplatin.JInorg 45. MahatoR,TaiW,ChengK:Prodrugsforimprovingtu- Biochem,2012,107,6–14. mortargetabilityandefficiency.AdvDrugDelivRev, 62. SieversEL:Antibody-targetedchemotherapyofacute 2011,63,659–670. myeloidleukemiausinggemtuzumabozogamicin 46. Martiniello-WilksR,WangXY,VoeksDJ,DaneA, (Mylotarg).BloodCellsMolDis,2003,31,7–10. ShawJM,MortensenE,BothGW,RussellPJ:Purine 63. SilvermanR.:TheOrganicChemistryofDrugDesign nucleosidephosphorylaseandfludarabinephosphate andDrugAction.2ndedn.,ElsevierAcademicPress, gene-directedenzymeprodrugtherapysuppressespri- Burlington,2004. marytumourgrowthandpseudo-metastasesinamouse 64. SinghPP,JoshiS,RussellPJ,VermaND,WangX, modelofprostatecancer.JGeneMed,2004,6, KhatriA:Molecularchemotherapyandchemotherapy: 1343–1357. anewfrontagainstlate-stagehormone-refractorypros- 47. MiwaM,UraM,NishidaM,SawadaN,IshikawaT, tatecancer.ClinCancerRes,2011,17,4006–4018. MoriK,ShimmaNetal.:Designofanoveloralfluoro- 65. SofiaMJ:NucleotideprodrugsforHCV therapy.Antivir carbamate,capecitabine,whichgenerates ChemChemother,2011,22,23–49. 5-fluorouracilselectivelyintumoursbyenzymescon- 66. SongX,LorenziPL,LandowskiCP,VigBS,Hilfinger centratedinhumanliverandcancertissue.EurJCancer, JM,AmidonGL:Aminoacidesterprodrugsoftheanti- 1998,34,1274–1281. canceragentgemcitabine:synthesis,bioconversion, 48. MüllerCE:Prodrugadministrationforenhancingthe metabolicbioevasion,andhPEPT1-mediatedtransport. bioavailabilityofdrugswithlowmolecularsolubility. MolPharm,2005,2,157–167. ChemDiver,2009,6,2071–2083. 67. Soul-LawtonJ,SeaberE,OnN,WoottonR,RolanP, 49. MusiolR,SerdaM,PolanskiJ:Prodrugsinphotody- PosnerJ:Absolutebioavailabilityandmetabolicdisposi- namicanticancertherapy.CurrPharmDes,2011,17, tionofvalaciclovir,theL-valylesterofacyclovir,fol- 3548–3559. lowingoraladministrationtohumans.Antimicrob 50. PardridgeWM:Braindrugdevelopmentandbraindrug AgentsChemother,1995,39,2759–2764. targeting.PharmRes,2007,24,1729–1732. 68. SpringerCJ,Niculescu-DuvazI.:Prodrug-activatingsys- 51. PatelMM,GoyalBR,BhadadaSV,BhattJS,AminAF: temsinsuicidegenetherapy.JClinInvest, 2000,105, Gettingintothebrain:approachestoenhancebraindrug 1161–1167. delivery.CNSDrugs,2009,23,35–58. 69. SrinivasNR:Therationalityforusingprodrugapproach 52. PengQ,WarloeT,BergK,MoanJ,KongshaugM: indrugdiscoveryprogramsfornewxenobiotics:oppor- 5-Aminolevulinicacid-basedphotodynamictherapy: tunitiesandchallenges.EurJDrugMetabPharmacoki- clinicalresearchandfuturechallenges.Cancer,1997,79, net,2011,36,49–59. 2282–2308. 70. StellaVJ:Prodrugs:somethoughtsandcurrentissues. 53. PescovitzMD,RabkinJ,MerionRM,PayaCV,PirschJ, JPharmSci,2010,99,4755–4765. FreemanRB,O’GradyJetal.:Valganciclovirresultsin 71. StellaVJ,BurchardtRT,HagemanMJ,OliyaiR,Maah improvedoralabsorptionofganciclovirinlivertrans- H,TilleyJW:Prodrugs:ChallengesandRewards.Part1, plantrecipients.AntimicrobAgentsChemother,2000; Springer,NewYork,2007. 44,2811–2815. 72. TeicherBA,ChariRV:Antibodyconjugatetherapeutics: 54. RautioJ,KumpulainenH,HeimbachT,OliyaiR,OhD, challengesandpotential.ClinCancerRes,2011,17, JärvinenT,SavolainenJ:Prodrugs:designandclinical 6389–6397. applications.NatRevDrugDiscov,2008,7,255–270. 73. TeicherBA:Antibody-drugconjugatetargets.Curr 55. RautioJ,LaineK,GyntherM,SavolainenJ:Prodrugap- CancerDrugTargets,2009,9,982–1004. proachesforCNSdelivery.AAPSJ,2008,10,92–102. 74. TestaB:Prodrugs;bridgingpharmacodynamic/pharma- 56. RodriguezL,BatlleA,DiVenosaG,BattahS,DobbinP, cokineticgaps.CurrOpinChemBiol,2009,13, MacrobertAJ,CasasA:Mechanismsof5-aminolevuli- 338–344. nicacidesteruptakeinmammaliancells.BrJPharma- 75. TietzeLF,KrewerB:Antibody-directedenzymeprodrug col,2006,147,825–833. therapy:apromisingapproachforaselectivetreatment

PharmacologicalReports, 2013,65,1–14 13 ofcancerbasedonprodrugsandmonoclonalantibodies. 80. YalthoTC,OndoWG:Theuseofgabapentinenacarbil ChemBiolDrugDes,2009,74,205–211. inthetreatmentofrestlesslegssyndrome.TherAdv 76. TsumeY,VigBS,SunJ,LandowskiCP,HilfingerJM, NeurolDisord,2010,3,269–275. RamachandranC,AmidonGL:Enhancedabsorptionand 81. YuC,HuY,DuanJ,YuanW,WangC,XuH,YangXD: growthinhibitionwithaminoacidmonoesterprodrugsof Novelaptamer-nanoparticlebioconjugatesenhancesde- bytargetinghPEPT1transporters.Molecules, liveryofanticancerdrugtoMUC1-positivecancercells 2008,13,1441–1454. invitro.PLoSOne,2011,6,e24077. 77. UmapathyNS,GanapathyV,GanapathyME:Transport 82. ZhangY,ChenY,HanD,OcsoyI,TanW:Aptamers ofaminoacidestersandtheamino-acid-basedprodrug selectedbycell-SELEXforapplicationincancerstudies. valganciclovirbytheaminoacidtransporterATB0,+. Bioanalysis,2010,2,907–918. PharmRes,2004,21,1303–1310. 83. ZhangY,HongH,CaiW:Tumor-targeteddrugdelivery 78. VytlaD,Combs-BachmannRE,HusseyAM,McCarron withaptamers.CurrMedChem,2011,18,4185–4194. ST,McCarthyDS,ChambersJJ:Prodrugapproachesto reducehyperexcitationintheCNS.AdvDrugDelivRev, 2012,64,666–685. 79. WuK-M:A newclassificationofprodrugs:regulatory Received: May7,2012; intherevisedform: August2,2012; perspective.Pharmaceuticals,2009,2,77–81. accepted: August14,2012.

14 PharmacologicalReports, 2013,65,1–14