Você está na página 1de 19

C H A P T E R 8

GENE-DIRECTED ENZYME
PRODRUG THERAPY

Caroline J. Springer
Ion Niculescu-Duvaz
Cancer Research Campaign Centre for Cancer Therapeutics
Institute of Cancer Research
Sutton, United Kingdom

Summary 5. The Activation Process


1. Introduction 6. Augmenting the Effect
2. Background 7. Exploiting the Bystander Effect and Acquired Immunity
3. Enzyme-Prodrug Systems 8. Conclusions
4. Tailored Prodrugs for GDEPT References

Summary frequently hampered by an insufficient therapeutic index and


the emergence of drug-resistant cell subpopulations. One area
The efficacy of cancer chemotherapy is frequently ham- where progress has been made is the discovery and synthesis
pered by a low therapeutic index. Recently, strategies have of extremely cytotoxic compounds. Recently, efforts have
been developed to improve this index by selective delivery of been directed to new concepts that allow the advantages of
highly cytotoxic drugs to the tumor cells themselves. A key these highly potent drugs with methods to enhance their se-
advance in this approach has been the principle of conversion lectivity. Immunoconjugates (Frankel et al., 1996), antibody-
of a specially designed nontoxic prodrug to a cytotoxic drug directed enzyme prodrug therapy (ADEPT) (Niculescu-Du-
by an enzyme contained within the tumor cells themselves, vaz and Springer, 1995; Melton and Sherwood, 1997), and
on the tumor cell surface, or in the tumor cell microenviron- suicide gene therapy (Niculescu-Duvaz et al., 1998b; Springer
ment. Activation of prodrugs can be accomplished by and Niculescu-Duvaz, 1999) are some of the new routes un-
antibody-enzyme conjugates targeted to tumor-associated der investigation. All of these technologies are two-step treat-
antigens (ADEPT) or by enzymes expressed by exogenous ments that avoid the requirements for drugs exhibiting in-
genes in tumor cells (GDEPT). This chapter discusses the trinsic selectivity toward cancer cells, since they rely on the
state of the art in GDEPT. The focus is on the development conversion of nontoxic prodrugs to toxic drugs within tumor
of new and improved prodrugs, and includes discussion of cells themselves, on their surface, or in their close vicinity.
physicochemical and biological properties, pharmacokinetics, Gene therapy is broadly defined as a technology aimed at
and clinical trials. modifying the genetic component of cells for therapeutic
benefits. In cancer gene therapy, both malignant and nonma-
lignant cells can be targeted for a therapeutic gain (Zhang et
1. Introduction al., 1995). The possibility of rendering cancer cells more sen-
sitive to chemotherapeutics or toxins by introducing “suicide
Cancer chemotherapy encompasses a vast number of es- genes” was suggested in the late 1980s. This approach has
tablished clinical methods for the treatment of persons with two alternatives. In toxin gene therapy, genes for toxic pro-
malignant diseases. However, the efficacy of such methods is teins are transduced into cancer cells. In gene-directed

Copyright © 2002 by Academic Press.


ANTICANCER DRUG DEVELOPMENT 137 All rights of reproduction in any form reserved.
138 SPRINGER AND NICULESCU-DUVAZ

enzyme prodrug therapy (GDEPT) (Bridgewater et al., 1995) 3. Enzyme-Prodrug Systems


or virus-directed enzyme prodrug therapy (VDEPT) (Huber
et al., 1991), genes for foreign enzymes are transduced into A large number of enzyme-prodrug systems have been de-
cancer cells that can activate specific prodrugs. veloped for GDEPT recently. These are abbreviated and sum-
This chapter discusses the state of the art in GDEPT. The marized in Table 1. There are specific requirements of the en-
delivery systems constitute another important aspect of this zymes for GDEPT. It is preferable for them to have high
technology and are discussed in detail elsewhere (Roth and catalytic activity, without the need for cofactors, as these
Cristiano, 1997; Bilbao et al., 1998; Miller, 1998; Nguyen et could become rate limiting in the target cells. The enzyme
al., 1998; Robbins and Ghivizzani, 1998). A number of re- should achieve a concentration sufficient to activate the pro-
views cover the qualitative and quantitative aspects of GDEPT drug under physiologic conditions.
technologies (Roth and Cristiano, 1997; Denny and Wilson, The enzymes proposed for GDEPT can be characterized
1998; Niculescu-Duvaz et al., 1998b; Encell et al., 1999; into two major classes for convenience. The first class com-
Springer and Niculescu-Duvaz, 1999, 2000). The focus here prises enzymes of nonmammalian origin although they may
is on the development of new and improved prodrugs. have human counterparts. Examples include viral TK1 (Cul-
ver et al., 1992), bacterial CD (Mullen et al., 1992), CPG2
(Marais et al., 1996), PNP (Sorscher et al., 1994), NR (Bridge-
water et al., 1995), XGPRT (Tamiya et al., 1996), PGA
2. Background (Moore et al., 1997), Met (Miki et al., 2000), -L (Moore et
al., 1997), -Gal (Ghosh et al., 1999), yeast CD (Kievit et
GDEPT and VDEPT are two-step treatments for patients al., 1999), DAAO (Stegman et al., 1998), plant MDAE
with solid tumors. In the first step, the gene for a foreign en- (Sethna et al., 1997), and insect dNK (Johansson et al., 1999).
zyme is delivered in a vector that is targeted to the tumor for However, these enzymes all have structural requirements that
expression. In the second step, a prodrug is administered that are different with respect to their substrates in comparison to
is activated to the drug selectively by the foreign enzyme ex- the human counterparts.
pressed in the tumor. The enzyme genes should be expressed The second class comprises enzymes of human or other
exclusively, or with a relatively high ratio, in the tumor cells mammalian origin that are absent from, or are expressed at a
compared with normal tissues and blood. low concentration in, tumor cells. Examples include CE (Ko-
Current vectors for gene delivery are incapable of confer- jima et al., 1998a), TP (Patterson et al., 1995), dCK (Hapke
ring expression of the foreign enzyme in all tumor cells. et al., 1996), CPA (Hamstra et al., 2000), -Glu (Weyel et
Therefore, a bystander effect (BE) is required whereby the al., 2000), and CYP450 (Chen et al., 1996a). Their major ad-
prodrug is cleaved to an active drug that kills neighboring tu- vantage resides in the reduction in the potential for induction
mor cells that are not expressing the foreign enzyme (Free- of an immune response. Their presence in normal tissues is
man et al., 1993). likely to preclude specific activation of the prodrugs only in
The design of GDEPT systems requires prodrugs “tai- tumors unless the transduced enzymes are modified so as to
lored” for the use of the foreign enzymes selected. Preferably have different substrate specificities. Genes have been con-
the enzyme has no human homologue. An alternative is to structed to express mutated enzymes with different substrate
use a human enzyme that is normally expressed in locations requirements in comparison with the wild types (Black et al.,
inaccessible to prodrug. Treatment schedules that are clini- 1996; Smith et al., 1997; Encell et al., 1999).
cally feasible are another consideration. The enzyme gene may be engineered for expression either
It is useful to compare the efficacy of different GDEPT intracellularly or extracellularly in the recipient tumor cells
systems. We have defined two parameters for this purpose: (Marais et al., 1997). There are potential advantages to each
the “potential of activation” and the “degree of activation” of approach. With intracellular expression the prodrug must en-
each system (see Table 1) (Springer and Niculescu-Duvaz, ter the cells for activation and subsequently the active drug
2000). The first parameter represents the maximal possible must diffuse through the interstitium across the cell membrane
efficiency of a given enzyme-prodrug system in a cell system. to elicit a BE. This is not a requirement in cells where the en-
It is defined as the ratio of the IC50 of the prodrug divided by zyme is expressed tethered to the outer cell surface or secreted
the IC50 of the released drug in a control nontransduced cell because the enzyme can activate the prodrug extracellularly.
line. The degree of activation demonstrates the actual effi- A more substantial BE should theoretically be generated in
ciency of the system in the transduced cell line. It is defined the latter systems. A leak-back of the active drug into the gen-
as the IC50 of the prodrug in the nontransfected cell line di- eral circulation is a possible disadvantage of these systems.
vided by the IC50 of the prodrug in an enzyme-transduced
cell line. 1
Abbreviations are defined in Table 1.
GENE-DIRECTED ENZYME PRODRUG THERAPY 139

TABLE 1
Enzyme-Prodrug Systems

Enzyme Prodrug Systems Potential of Degree of


activation activation Clinical
Number Names and codes Origin Expression Mutation Prodrugs Released (pro)drugs fold fold trials

11 Carboxyl esterase Human, Intracellular No Irinotecan SN-38 0150–3000 17–17 1


(CE) rabbit 7-ethyl-10-[4-(1- 7-ethyl-10-hydroxy-(20S)-
piperidino)-1- camptothecin
piperidino]-
carbonyloxy-(20S)-
camptothecin
12 Carboxypeptidase Human Intracellular Yes, for secreted MTX- -peptides MTX 1000 400 —
A (CPA) and extra- or surface tethered
cellular expression and
secreted modified
substrates
13 Carboxypeptidase Pseudomonas Intracellular Yes, for extra- CMDA CMBA 421–400 111–115 1
G2 (CPG2) EC str. and extra- cellular ZD-2767 phenol-bisiodonitrogen
3.4.22.12 cellular sur- expression Self-immolative mustard; alkylating agents,
face tethered anthracycline antibiotics
14 Cytochrome P450, Human, Intracellular No Oxazaphosphorines, Alkylating agents, toxic ? 105–100 1
Human; CYP2B1, rat, rabbit ipomeanol, 2-amino- metabolites, N-acetyl-
2B6, 2C8, 2C9, anthracene (2-AA); benzoquinone imine
2C18, and 3A acetaminophen (NABQI)
Rat: CYP2B1
Rabbit: 4B1 (with
or without red-
P450)
15 Cytosine E. coli, yeast Intracellular Yes, for secreted 5-Fluorocytosine 5-Fluorouracil 1000–8000 1070–1000 1
deaminase (CD) and extra- expression (5-FC) (5-FU)
EC 3.5.4.1 (with cellular
or without uracil-
phosphoribosyl
transferase, UPRT)
16 D-Amino acid Rodhoto rula Intracellular No D-Alanine Hydrogen peroxide — — —
oxidase (DAAO) gracilis,
(yeast)
17 Deoxycytidine Human Intracellular No Cytosine arabinoside Cytosine arabinoside — — —
kinase (dCK), monophosphate
EC.2.7.1.21
18 Deoxyribonucleo- Drosophila Intracellular No Analogues of pyrimi- Analogues of pyrimidine ? ? No
tide kinase melanogaster dine and purine 2- and purine 2-deoxynu-
(DmNK) deoxynucleosides cleotide monophosphates
19 DT-Diaphorase Human, rat? Intracellular No Bioreductive agents: Reduced forms? — — No
(DT-D) EO9, etc.
10 -Galactosidase E. coli Intracellular No Self-immolative pro- Anthracycline antibiotics — — No
( -Gal) EC drugs from anthracy-
3.2.1.23 clin antibiotics
11 -Glucuronidase Human Intracellular Yes, for secreted Self-immolative Doxorubicin 235 4–5 No
( -Glu) and extra- expression HM-1826
cellular
12 -Lactamase Bacterial Extracellular, Yes Self-immolative Alkylating agents, Vinca — — —
( -L) ? secreted or (cephem prodrugs) alkaloids, anthracycline
surface antibiotics
tethered
13 Methionine- ,- Pseudomonas Intracellular No Selenomethionine Methylselenol ? 400 No
lyase (MET) putida
14 Multiple drug Tomato Intracellular No Acetylated 6-TG, 6-TG, MTX, cytotoxic — — No
activating enzyme MTX, and other purines
(MDAE) purines

(continues)
140 SPRINGER AND NICULESCU-DUVAZ

TABLE 1 (continued)

Enzyme Prodrug Systems Potential of Degree of


activation activation Clinical
Number Names and codes Origin Expression Mutation Prodrugs Released (pro)drugs fold fold trials

15 Nitroreductase E. coli Intracellular No CB-1954 and ana- Alkylating agents, 50,000 14–10,000 No
(NR) logues; self-immolative pyrazolidines, enediynes
16 Penicillin G ? Extracellular Yes — — — — No
amidase (PGA)
17 Purine nucleotide E. coli Intracellular No Purine nucleosides 6-Methylpurine, 2- 0025–1000 440 No
phosphorylase fluoroadenine
(PNP), EC2.4.2.1
18 Thymidine kinase, HSV or V2V Intracellular Yes, to improve Modified pyrimidine Monophosphate nucleotide 0020–1000 25
(TK), EC2.7.1.21 phosphorylation nucleosides: GCV, analogues
kinetics ACV, valacyclovir, etc;
FIAU, purine
nucleosides
19 Thymidine phos- Human Intracellular No Pyrimidine analogues, 5-Fluorodeoxyuridine 7000 165 No
phorylase (TP), 5-DFUR monophosphate
EC2.4.2.4 (5-FdRMP)
20 Xanthine-guanine E. coli Intracellular No 6-Thiopurines 6-Thiopurine nucleoside — — No
phosphoribbosyl
transferase (XGPT)

ACV, acyclovir; CB1954, 5-aziridinyl-2,4-dinitrobenzamide; CMBA, N,N-(2-chloroethyl)(2-mesyloxyethyl)aminobenzoic acid; CMDA, N,N-(2-chloroethyl)(2-


mesyloxyethyl)aminobenzoyl-L-glutamic acid; 59-DFUR, 5-deoxy-5-fluorouridine; EO9, 3-hydroxy-5-aziridinyl-l-methyl-(1H-indole-4,7-dione)-propenol; FIAU, 1-(2-deoxy-2-
fluoro- -D-arabinofuranosyl)-5-iodouracil; GCV, ganciclovir, HM-1826, N-(4- -glucuronyl-3-nitrobenzyloxycarbonyl)-doxorubicin; HSV, herpes simplex virus; MTX, methotrex-
ate; 6-TG, 6-thioguanine; VZV, varicella zoster virus; ZD-2767, 4-[bis(2-iodoethyl)aminophenyl]-oxycarbonyl-L-glutamic acid.

4. Tailored Prodrugs for GDEPT drug, without the requirement for multiple catalysis steps, since
it is possible that the needed host endogenous tumor enzymes
Prodrug design must address several issues for clinical ap- will become defective or deficient in cancer cells. Alkylating
plicability. These include ease of transport of the prodrugs agent prodrugs such as CP and CB1954 have an activation cas-
within the tumor environment. Intracellular transport is re- cade that depends on endogenous enzymes. CYP activates CP
quired if the enzyme is to be intracellularly expressed. The to an intermediate that requires 3,5-exonuclease action to gen-
prodrug should have very limited cytotoxicity whereas the erate the active metabolite (Bielicki et al., 1984; Brock, 1989).
activated drug should be as potent as possible. There should NR activates CB1954 to the 5-aziridinyl-4-hydroxylamino-2-
be effective activation of the prodrug by the expressed en- nitrobenzamide intermediate, and a further enzymatic step con-
zyme with favorable activation kinetics. Ideally, the drugs re- verts it to a powerful electrophile that can alkylate DNA (Knox
leased should kill both proliferative and quiescent cells and et al., 1988, 1992, 1993). Alkylating agent prodrugs, such as
should induce a BE. Prodrugs should be chemically stable CMDA and ZD-2767P, have an advantage since the active drug
under physiologic conditions. is released by the expressed enzyme directly after prodrug
For intracellular activation, prodrugs that are lipophilic or cleavage (Springer et al., 1990, 1995).
have active transport mechanisms are required to penetrate There is a requirement for further catalysis by mammalian
cell membranes. It appears that many prodrugs in current use, adenosine monophosphate deaminase AMP and/or GMP ki-
such as nucleoside analogues, 5-FC, CP, CMDA, and CB1954, nases with 6-methoxy purine arabinonucleoside, GCV, and
penetrate cells by passive diffusion. It is crucial that they ACV following activation by the expressed VZV- or HSV-TK
have minimal cytotoxicity, since they will be taken up by nor- (Huber et al., 1991). 5-FC is converted to 5-FU by CD. The
mal and turnover cells. Modifying the lipophilicity of pro- active metabolite is 2-deoxy-5-fluorouridine-5-monophos-
drugs is a possibility, especially when passive diffusion is in- phate (5-FdUMP) or 5-fluorouridine-5-triphosphate (5-FUTP),
volved in prodrug uptake. For systems in which the enzyme which results from 5-FU conversion by a number of mam-
is extracellularly expressed, it is ideal if the prodrug is pre- malian enzymes involving a complicated activation pattern
vented from crossing the cell membrane, in contrast with the (MacCoss and Robins, 1990). The sophisticated activation
released drug, which should be membrane permeable. pathway and the existence of salvage paths for these an-
Activation of the prodrugs is a key step. It is an advantage if timetabolites are partially responsible for their propensity to in-
the expressed enzyme can activate the prodrug directly to the duce resistance (Kinsella et al., 1997).
GENE-DIRECTED ENZYME PRODRUG THERAPY 141

The prodrugs used in GDEPT are commonly antimetabo- 1. Activation of Prodrugs by Scission Reactions
lites that require cycling cells (S phase) for cytotoxicity and This is the most common mode of activation. A wide range
are not active against quiescent cells. It has been speculated of anticancer compounds can be made less cytotoxic by con-
that resistance to GCV is not acquired but results from tumor version to esters, amides, ureides, carbamates, or glycosides.
cells that are in G0 at the time of GCV administration, ren- When suitably substituted these are often good substrates for
dering them insensitive to the action of activated GCV. This the range of enzymes used in GDEPT. For instance, sub-
hypothesis is supported by the fact that the tumors, which strates for CPG2 require an L-glutamyl residue coupled to an
grew out following GDEPT treatment, remained sensitive to aromatic nucleus by an amidic (Springer et al., 1990), urei-
GCV (Golumbek et al., 1992). dic, or carbamic bond (Springer et al., 1995; Dowell et al.,
Optimum half-lives are required from the drugs to maxi- 1996). The chemical reactivity of the benzoic, phenol or ani-
mize efficacy. The half-life should be long enough to allow the line aromatic nitrogen mustards is greatly reduced when they
BE but short enough to prevent the drug leaking out of the tu- are functionalized as esters, amides, ureas, or carbamates.
mor (Springer et al., 1995). The use of alkylating agent pro- Coupling with L-glutamic acid results in prodrugs, such as 1,
drugs has a potential advantage over purine nucleosides or 5- that are substrates for CPG2 (Scheme 1). A large number of
FC, since they are cytotoxic to noncycling as well as prodrugs have been designed and synthesized (Scheme 1).
proliferative cells. It has been shown that the NR/CB1954 sys- The degree of activation in a number of cell lines is up to
tem was effective in noncycling cells (Bridgewater et al., 1995). 400-fold (Niculescu-Duvaz et al., 1999a). Also, good in vivo
Activity against quiescent cells was also claimed for 6-MeP results were reported with this system (Marais et al., 1996;
and 2-FA in PNP-transfected cells (Secrist III et al., 1999). Stribbling et al., 2000).
A similar scission reaction occurs by CE on irinotecan (7-
A. Prodrugs ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy-(20S)-
camptothecin, 4, releasing SN-38 (7-ethyl-10-hydroxy-(20S)-
These prodrugs can be defined as pharmacologically inac- camptothecin, 5, that is approximately 1000-fold more
tive derivatives of drugs, which require chemical transforma- cytotoxic than the corresponding prodrug (Scheme 2) (Ko-
tion to release or be converted to the active drug. In a non- jima et al., 1998a; 1998b). Recently, a more active rabbit CE
self-immolative prodrug the active drug is formed directly was proposed to enhance the efficiency of the system (Wierdl
following the activation in a one-step process. The non-self- et al., 2000).
immolative prodrugs used in GDEPT can be activated by dif- The -peptides of N-{5-[N-(3,4-dihydro-2-methyl-4-
ferent types of enzymatic reactions: oxoquinazolin-6-ylmethyl]-N-methylamino}-2-thienoyl-L-glu-
1. Scission reactions (e.g., CPG2, CE, CPA, PNP, PGA, tamic acid (ZD1694) (6, R  L-Ala, L-Glu) and N-{4-[(2-
MDAE, TP, -Gal, -L, -Glu) methyl-3,4-dihydro-4-oxo-6-quinazolinyl)methyl]-N-prop-2-
2. Reductions (e.g., NR, DT-diaphorase) ynylamino}-benzoyl-L-glutamic acid (ICI 198538) (7, R 
3. Phosphorylation (e.g., HSV-TK, VZV-TK, dCK) L-Ala, L-Glu) for activation by CPA have been synthesized.

4. Hydroxylation (e.g., CYP4B1) Differentials of up to 100-fold were obtained in vitro in cell


5. Functional group substitution (NH2OH) (e.g., CD) lines with and without CPA (Springer et al., 1996) (Scheme 3).
6. Deoxyribosylation (e.g., XGPRT) Prodrugs of methotrexate (MTX), 8, have been described
7. Oxidation (e.g., DAAO) in which a blocking amino acid is conjugated to the -
carboxylic group (through an amide linkage) of the L-glu-
Most of the prodrugs used to date in clinical trials have been tamic acid residue (Smith et al., 1997; Hamstra et al., 2000).
licensed anticancer agents, as their pharmacology, pharmaco- These prodrugs cannot be internalized by the reduced folate
kinetics, and dosages are understood. cofactor (RFC) and are therefore inactive. Removal of the

SCHEME 1 X, Y  Cl, Br, I, OSO2CH3; Z  —, O, NH, CH2, S; W  CO2H, NH2, OH, CH2CO2H, SH; R1  H, 2(3)-CH3, 2(3)-F, 2(3)-Cl, 3-i-C3H7, 3-CN.
142 SPRINGER AND NICULESCU-DUVAZ

with MTX- -Phe in this system with secreted CPA in vitro


(Smith et al., 1997).
A scission reaction of a glycosidic C-N bond by PNP
released cytotoxic purines from their corresponding nucleoside
prodrugs, 10 (Scheme 4) (Sorscher et al., 1994). Of the pu-
rine nucleosides investigated, 6-methyl-9-(2-deoxy- -D-
erythropentafuranosyl)-purine, 10a, and 2-fluoro-9- -D-
arabinofuranosyladenine, 10b, proved good substrates. It
appeared that both purines were further metabolized to the
corresponding nucleoside triphosphates (Parker et al., 1998).
Both drugs were also active against quiescent cells (Secrist
SCHEME 2 Activation of irinotecan by carboxyl esterase C. III et al., 1999).

amino acid by CPA releases MTX, 9. However, since these


MTX -amino acid prodrugs are sensitive to endogenous se-
creted CPA, the enzyme was mutated to allow the design of
MTX -peptides with unnatural blocking amino acid that
are not substrates for the wild-type CPA (Scheme 3) (Smith
et al., 1997). For instance MTX- -3-cyclopentyl-Tyr prodrug
is 50,000-fold more stable to wild-type CPA (Hamstra et al.,
2000). Degrees of activation of up to 400-fold were reported

SCHEME 4 Activation of 6-MePdR and 2-FADR by PNP. (a) R  CH3;


R1  H; (b) R  NH2; R1  F.

A similar mechanism was suggested for activation of the


TP/5-DFUR system, although there was no demonstration
that human TP could cleave the gycosidic bond of 5-DFUR
to yield 5-FU (Patterson et al., 1995; Evrard et al., 1999).
A system based on the use of -Glu and a glycosylated
derivative of doxorubicin was developed. This system has
been improved by using a secreted form of the human lyso-
somal enzyme that converts the doxorubicin prodrug (HMR
1826) to the active drug in the tumor interstitium. The effect
is mediated through a BE (Weyel et al., 2000).
Recently, a system based on a gene encoding the glycosi-
dase linamarase was developed using the nontoxic cyanogenic
glycoside substrate linamarin (Izquierdo and Corts, 1999).
The system released the highly diffusible cyanhydric acid,
which had a large BE on neighboring cells.

2. Activation of Prodrugs by Reduction (NR, DT-d)


Reduction of the prodrug CB-1954, 13, by rat DT-d (kcat 
SCHEME 3 Activation of MTX- -peptides by wild type or mutated CPA. 4.1 min1) at the 4-nitro group to the hydroxylamine 14 con-
R  L-Ala, L-Glu; R1  Glu, Asp, 2-carboxy-Phe, 3-carboxy-Phe, 2-iodo-
Phe, 2-cyclopentyl-Phe, 2-cyclohexyl-Phe, 3-cyclobutyl-Phe (8a), 3-
verted the prodrug to a bis-alkylating agent capable of cross-
cyclopentyl-Phe, 2-cyclopentyl-Tyr, 3-cyclobutyl-Tyr, 3-cyclopentyl-Tyr linking DNA (Scheme 5). Human DT-d can also perform this
(8b), 3-carboxy-Tyr, naphtyl-Ala. reduction, but at a slower rate (kcat  0.64 min1) (Boland et
GENE-DIRECTED ENZYME PRODRUG THERAPY 143

SCHEME 6 Activation of EO9 by DT-diaphorase.

oxynucleosides) are converted to mono-, di-, and triphos-


phate analogues by a range of enzymes. The triphosphates
are incorporated into DNA or RNA. Therefore, modified
purines or pyrimidines can act as fraudulent substrates in-
hibiting DNA or RNA synthesis, as long as they behave as
substrates for the mammalian kinases, DNA or RNA poly-
merases. Some modified nucleosides, such as GCV, 20, ACV,
24, FIAU, 25, and Ara-M, 26, are converted to their corre-
sponding monophosphates by endogenous monophosphate
kinases with low efficiency. However, viral monophosphate
kinases such as herpes simplex virus (HSV)-TK or varicella
zoster virus (VZV)-TK are very effective in achieving this
conversion. These nucleosides are used as prodrugs with the
SCHEME 5 Activation of nitroderivatives of alkylating agents by NR (E.
coli) and DT (rat). X  Cl, Br, I; R  H, CH2CHOHCH2OH. HSV-TK or VZV-TK enzymes (Scheme 7). Degrees of acti-
vation up to 1000 fold were achieved with the HSV-TK/GCV
and up to 600 fold with the VZV-TK/araM system. (Averett
al., 1991). Escherichia coli NR was able to reduce either the et al., 1991; Huber et al., 1991; Rubsam et al., 1998).
2- or the 4-nitro groups in CB-1954 more efficiently (kcat  This approach has led to promising results in vivo and
360 min1) (Anlezark et al., 1995; Friedlos et al., 1997). De- more than 30 clinical trials have been initiated with HSV-
grees of activation were reported to be up to 10,000-fold in TK/GCV mainly in glioblastoma. Prodrugs with increased
rodent cell lines, whereas an activation of 670-fold was de- lipophilicity compared to GCV (R  elaidic acid ester) im-
scribed in human cell lines (Friedlos et al., 1998). proved the effect in vitro (Balzarini et al., 1998). The in-
A similar reduction occurs on aromatic nitrogen mustard creased cytotoxicity of this compound is explained by the
analogues, such as 16 or 17 (kcat  1580 min1) with E. coli prolonged intracellular retention of the GCV anabolites in
NR (Anlezark et al., 1995). Here the activation mechanism is transduced tumor cells. Some clinical advantages were
different since the prodrugs already possess a difunctional claimed for valaciclovir, the oral form of GCV (Hasenburg et
alkylating moiety. The reduction may be fully activating the al., 1999). It was considered that the kinetics were not opti-
aromatic nitrogen mustards, 16 or 17, by electrons released mal for these substrates. The enzyme was mutated, leading to
from the newly formed amino or hydroxylamino groups. The a TK that compared to wild-type kinase renders the cell 43-
bisiodo- or bisbromo analogues (16, X  I, Br, R  CONH2; fold more sensitive to GCV and 20-fold more sensitive to
17, X  I, Br, R  CONH2) proved to be superior to CB- ACV. The enhanced killing correlates to altered substrate
1954 from a range of derivatives. Degrees of activation up to specificity (the Km of the mutant is 10 mM, 5-fold lower than
2532-fold were reported in vitro for these compounds (Fried- the Km of the wild-type enzyme) (Black et al., 1996).
los et al., 1997) (Scheme 5). dCK is another enzyme with the ability to activate pro-
The human DT-d (NQO1) has also been suggested for use drugs by phosphorylation. The human enzyme is involved in
in GDEPT with bioreductive drugs such as EO9, 19, mito- the salvage pathway of deoxyribonucleotide synthesis. The
mycin C, streptonigrine, and diaziquinone (Warrington et al., monophophorylation step, catalyzed by dCK, is the rate-
1998) (Scheme 6). limiting step in the conversion of deoxyribonucleosides to
deoxyribonucleotides triphosphates. A number of modified nu-
3. Activation of Prodrugs by Phosphorylation (Tks) cleosides with anticancer activity, ([1-( -D-arabinofuranosyl)-
Phosphorylation is a common reaction in the metabolic cytosine, ara-C, 29; 2-chloro-2-deoxyadenosine, CdA, 32;
pathway of purines and pyrimidines. Nucleosides (or de- 2-fluoro-9-( -D-arabinofuranosyl)-adenine, FaraA, 33; and
144 SPRINGER AND NICULESCU-DUVAZ

SCHEME 7 Activation of GCV, ACV, FIAU, and Ara-M by viral TK. R  valine ester; elaidic acid ester.

2,2-difluorodeoxycytosine, dFdC or gemcitabine, 34) are


activated by this mechanism (Scheme 8).
Since tumors express low levels of dCK endogenously, it
has been suggested that dCK gene transfer may increase their
sensitivity to such modified nucleosides. Ara-C/dCK was pro-
posed for GDEPT (Hapke et al., 1996; Manome et al., 1996).

4. Activation of Prodrugs by C-hydroxylation


(Cytochrome P450)
Rabbit cytochrome P450 (CYP4B1) is capable of activating
ipomeanol, 35, efficiently to cytotoxic metabolites (Scheme 9)
(Blaise Smith et al., 1995). This combination was recently pro-
posed for GDEPT since the human CYP4B1 isoenzyme pos-
sesses only 1% of the rabbit enzyme activity toward ipomeanol
(Rainov et al., 1998). CYP4B1/2-aminoanthracene (2-AA) is
a similar combination with a degree of activation up to 20-fold
in glioma cells expressing CYP4B1 (Rainov et al., 1998).
The human CYP1A2 was shown to be the main isoform re-
sponsible for the oxidation of acetaminophen, 36, to acetylim-
inoquinone, 37 (NABQI) (Scheme 9). Recently, this system
was proposed as a candidate for GDEPT because of the high
cytotoxicity of the released NABQI (Tatcher et al., 2000). A SCHEME 8 Activation of analogues of purine and pyridimine nucleotides
BE effect was reported in ovarian- or colon-carcinoma- by dCK.
GENE-DIRECTED ENZYME PRODRUG THERAPY 145

et al., 1994). One problem is the low sensitivity for 5-FU of


many malignant cells expressing CD due to the absence of
uracylphosphoribosyltransferase (UPRT) that catalyzes the
conversion of 5-FU to 5-FdUMP in mammalian cells. A hy-
brid gene for CD and UPRT was transfected into tumor cells
to overcome this drawback. The enhanced conversion of 5-FC
to 5-FdUMP also has a positive effect on the uptake of the ex-
tracellular prodrug. We have calculated that a degree of acti-
vation of 70- to 1000-fold was obtained in a number of trans-
fected cell lines (Springer and Niculescu-Duvaz, 2000).
Recently, it was found that CD isolated from yeast has ad-
vantages over the bacterial enzyme (Kievit et al., 1999). This
GDEPT system was studied extensively. It was found to have
a better BE than HSV-TK/GCV and is currently in clinical tri-
als. A phase I clinical trial of suicide therapy with CD/5-FC
used with the c-erbB-2 promoter in breast cancer has been de-
SCHEME 9 Activation of ipomeanol by rabbit CYP4B1 and of aceta-
minophen by CYP1A2. scribed. The approach proved to be safe and resulted in tar-
geted expression in up to 90% of cases (Pandha et al., 1999).
derived cells transfected with CYP1A2, following aceta- 6. Activation of Prodrugs by Deoxyribosylation of
minophen treatment. Purine Bases (XGPRT)
5. Activation of Prodrugs by Functional Group The bacterial enzyme XGPRT, in contrast to its human
Substitution (CD) counterpart HPRT, can convert xanthine to xanthine
monophosphate effectively. XGPRT is also able to transfer
CD is an enzyme isolated from E. coli that can convert the the deoxyribose phosphate moiety to 6-mercaptopurine (6-
nontoxic antifungal 5-FC, 38, to the anticancer drug 5-FU, 39. MP), which is further activated to its highly cytotoxic triphos-
The latter is converted enzymatically to 5-fluorodeoxyuridine phate analogue (Mulligan and Berg, 1981). 6-Thioguanine
monophosphate (5-FdUMP), 41, a thymidylate synthase in- (6-TG), 44, can also be used since it is metabolized to 6-
hibitor, or the triphosphate analogue, 42, that is incorporated thioxanthine (6-TX), 45, by endogenous enzymes (Scheme
as a fraudulent base in DNA or RNA (Scheme 10) (Huber 11). Degrees of activation of 10- to 20-fold were obtained for
both prodrugs (Tamiya et al., 1996).

SCHEME 11 Activation of 6-TG and 6-TX by XGPRT.

7. Production of H2O2 by D-Amino Acids Oxidation


H2O2 is a relatively stable, membrane-permeable mole-
cule that is cytotoxic to quiescent as well as proliferating
cells. Intracellularly generated H2O2 can be reduced to hy-
droxyl radicals via transition-metal-catalyzed Haber–Weiss
chemistry (Halliwell and Gutteridge, 1989). The radicals re-
act with DNA, lipids, and protein, resulting in cell death.
SCHEME 10 Activation of 5-FC by CD. H2O2 was generated selectively in tumor cells by transfection
146 SPRINGER AND NICULESCU-DUVAZ

of the DAAO gene from R. gracilis and selected amino acids nects the drug to that part of the molecule that is recognized by
(Scheme 12). the enzyme (Niculescu-Duvaz et al., 1999a). A self immolative
prodrug designed for activation by CPG2 is described in Scheme
13. As shown, the L-glutamic acid and the active drug are sep-
arated by a 4-hydroxy- or 4-amino-substituted benzylic spacer.
Activation of these prodrugs involves two steps. First, CPG2
cleaves the oxycarbonyl or carbamoyl L-glutamyl bond. This is
SCHEME 12 Production of H2O2 by oxidation of D-amino-acids with
DAAO.
followed by the spontaneous decomposition of the carbonic or
carbamic acid thus formed, with loss of CO2. Second, the self-
immolative intermediate, 48, is fragmented by 1,6 elimination,
The stereoselectivity for D-amino acids appears to be ab- releases a carbonic or carbamic acid, and generates an active
solute, since L-amino acids are neither substrates nor inhibitors drug, 51, and a quinoneimine, 50, with loss of CO2.
of the enzyme (Pollegioni et al., 1992). The mammalian
DAAO enzyme possesses very low activity (KM 6.5 mM,
for D-Ala) and it oxidizes achiral glycine, but this amino acid
is a poor substrate for DAAO from R. gracilis. The IC50 of
D-Ala decreased from more than 100 mM to 2.4 mM in
DAAO-transfected cells, and a further decrease was obtained
when L-buthionine-(S,R)-sulfoximine (BSO, a glutathione
transferase inhibitor) was added to the cells. This DAAO/D-
Ala combination was suggested as a candidate for GDEPT in
the management of the brain tumors (Stegman et al., 1998).

B. Self-Immolative Prodrugs
Many of the enzymes used in GDEPT impose rigid struc-
tural requirements for their prodrug substrates. This limits the
anticancer agents that can be used in the design of the pro-
drugs. To overcome this “self-immolative prodrugs” have
been synthesized. A self-immolative prodrug can be defined
as a compound that generates an unstable intermediate after
activation, which then extrudes the active drug in a series of
subsequent steps. The activation process is generally enzy-
matic in nature and is distinct from the extrusion step. The
extrusion of the active drug relies on a supplementary spon-
SCHEME 13 Activation of self-immolative drugs by CPG2. X, Y  F, Cl,
taneous fragmentation. The mechanism involved can be a Br, I, OSO2CH3; Z  O, NH; R1  H, OH.
1,6-, 1,4 elimination (Wakselman, 1983; Greenwald et al.,
1999) or a cyclization reaction (Sykes et al., 1999). The site
of activation is usually separated from the site of extrusion. Aromatic or aliphatic nitrogen mustards, 53, and anthracy-
The potential advantages of these types of prodrugs is that cline antibiotics, 54, with an amino or hydroxy group were
the range of drugs that can be converted to prodrugs is greatly acylated resulting in deactivated molecules. The anthracy-
extended and is unrestricted by the structural substrate re- clines, such as doxorubicin and daunorubicin, are antitumor
quirements for a given enzyme. There is also the possibility of drugs with a wide spectrum of activity in human tumors. Their
altering the lipophilicity of the prodrugs with minimal effect therapeutic efficacy is often limited by toxic side effects, mainly
on the activation kinetics and improving the kinetics of activa- cardiotoxicity and myelosuppression. Several “non-self-
tion by modifying electronic or steric features of the active immolative” and “self-immolative” anthracycline prodrugs were
drug. A number of self-immolative prodrugs have been de- prepared for activation by different enzymes to overcome these
signed and synthesized based on a range of activation processes. toxicities mainly by derivatization of the amine functionality of
the daunosamine (Andrianomenjanahary et al., 1992; Gesson
1. Prodrugs Designed to Be Activated by an et al., 1994; Farquhar et al., 1998). Unfortunately, CPG2 was
Enzymatic Scission Reaction Followed by a unable to activate any of the reported prodrugs. Furthermore,
1,6 or 1,4 Elimination direct addition of L-glutamyl residues to doxorubicin or
The 1,6-elimination strategy requires the use of a benzylic daunorubicin did not generate a molecule that was a substrate
spacer in the construction of the prodrugs, 47. The spacer con- for CPG2. Therefore, prodrugs 47 (R  54, R1  OH and
GENE-DIRECTED ENZYME PRODRUG THERAPY 147

R  54, R1  H) derived from doxorubicin and daunorubicin


were based on self-immolative linker strategies with the aim of
reducing their intrinsic toxicity (Scheme 14). These prodrugs
were activated by CPG2, releasing the active drugs by a 1,6-
elimination mechanism (Niculescu-Duvaz et al., 1999b).
Self-immolative prodrugs of nitrogen mustards provided
SCHEME 15 Activation of cephem self-immolative prodrugs by -lacta-
degrees of activation of 12- to 14-fold and a potential of ac- mase (1, 4-elimination). R  alkyl, aryl, heterocycle, etc.; R1  alkylating
tivation of 20- to 33-fold for compounds 47 (R  53; Z  agent, platinum compound, Vinca alkaloid, anthracycline antibiotics.
NH, O; X  Y  Cl) (Niculescu-Duvaz et al., 1998a). For
the doxorubicin prodrug 47 (R  54, R1  OH), the degree density in the  system and stabilizing the positive charge on
of activation was 11-fold and the potential of activation 21- the benzyl carbon. This triggers a 1,6-elimination process
fold (Niculescu-Duvaz et al., 1999b). Another approach, with the extrusion of the active drug (Scheme 16). A prodrug
based on an enzymatic scission reaction catalyzed by E. coli 64 (R  67) containing an enedyine residue has been syn-
-Gal, was described. 4( -D-Galactopyranosyl)benzyl- thesized by this method (Hay et al., 1999b).
N,N,N,N-tetrakis-(2-chloroethyl)phosphorodiamidate was
synthesized. The prodrug, 55, releases by a two-step frag-
mentation process, the alkylating antitumor agent N,N,N,N-
tetrakis-(2-chloroethyl)phosphorodiamidic acid, 59, when in-
cubated with -Gal (Ghosh et al., 1999) (Scheme 14).

SCHEME 16 Activation of 4-nitrobenzyloxycarbonyl self-immolative


prodrugs with NR.
SCHEME 14 Activation of self-immolative alkylating prodrugs by -G.

In an effort to extend the range of substrates for NR, the


Recently, secreted -lactamase (Moore et al., 1997) was 2-nitroimidazole moiety was considered as a possible sub-
also proposed for GDEPT. There are a number of self- strate. It can fragment after reduction of the nitro group to
immolative prodrugs, 61, capable of being activated by this release the drug. A carbamate prodrug from 5-amino-1-
enzyme (Jungheim and Shepherd, 1994). Activation is the re- (chloromethyl)-3-(5,6,7-trimethoxyindol-2-ylcarbonyl)-1,2-
sult of an enzymatic scission reaction followed by a 1,4-elim- dihydro-3H-benz[e] indole 69 (R  68) has been synthesized
ination process (Scheme 15). and was activated under hypoxic conditions (Hay et al.,
1999a) (Scheme 17). Degrees of activation of 21- to 135-fold
2. Prodrugs Designed to Be Activated by an Enzymatic and 21-fold were reported for compounds 64 (R  67) and
Reduction Followed by a 1,6 Elimination 69 (R  68), respectively (Hay et al., 1999a, 1999b).
Self-immolative prodrugs, including derivatives of aniline
nitrogen mustard, enediyne, and amino-seco-cyclopropylindolyl, 3. Prodrugs Designed to Be Activated by an Enzymatic
were designed as substrates for E. coli NR. Despite different Reduction Followed by Cyclization
substrate specificities, they were all reduced to the corre- A different type of prodrug was prepared based on the re-
sponding 4-hydroxylamino derivatives, increasing electron ductive release of the amines from 2-(2,6-dinitrophenylamino)-
148 SPRINGER AND NICULESCU-DUVAZ

Human CYT2B6 and CYT3A4 are the most important isoen-


zymes in liver activation of CP and IF. Rat CYT2B1 is also
highly effective. Cytochrome P450 genes (CYT2B1, 2B6, 2C8,
2C9, 2C18 3A) were transfected in tumor cells and investigated
in conjunction with oxazaphosphorines (Patterson et al., 1999;
Waxman et al., 1999). These studies have also shown that ther-
apeutic benefit is possible despite the contribution of hepatic
metabolism to CP and IF activation. Degrees of activation in the
range of 10- to 100-fold were reported. An improvement in ac-
SCHEME 17 Activation of 2-nitro-imidazolylcarbamates self-immolative tivation was noticed when a double suicide gene therapy sys-
prodrugs by NR. tem containing a hybrid gene of CYP4B1 and P450-reductase
were transfected to malignant cells (Chen et al., 1997).

propanamide, via hydroxylaminoamide cyclization-extrusion


reaction (Sykes et al., 1999). This process is greatly accelerated
by an H-bonding “conformational lock” between the anilino
5. The Activation Process
NH group and the adjacent 2-(6)-nitro group (Scheme 18).
A. Kinetic Parameters
Prodrug examples containing aniline nitrogen mustard, 72
(R  75) or 5-amino-1-(chloromethyl)-3-(5,6,7-trimethoxyindol- The concentration of the drug and the rate at which it is re-
2-ylcarbonyl)-1,2-dihydro-3H-benz[e]indole, 72 (R  68), have leased at the activation site depends on the kinetic parameters
been synthesized but were not activated efficiently by NR (Sykes of the enzyme-prodrug system. The Km is an expression of the
et al., 1999). amount of substrate required to reach half of maximum ve-
locity, Vmax. The turnover number, kcat, or the Vmax supplies
4. Prodrugs Designed to Be Activated by an Enzymatic additional information about the reaction rate, since both are
Hydroxylation Followed by a 1,4 Elimination terms to express the rate of drug release. It is impossible to
Cyclophosphamide (76, R  CH2CH2Cl, R1  H) and compare enzyme-prodrug systems solely on this basis since
ifosfamide (76, R  H, R2  CH2CH2Cl) undergo sponta- these data are not often recorded. It can be hypothesized that
neous decomposition after hydroxylation by the cytochrome low Km and high Vmax (or kcat) are associated with more ef-
P450 enzyme via the open-form aldophosphamide, 78 fective systems (Springer and Niculescu-Duvaz, 2000). This
(Scheme 19). Aldophosphamide releases a phosphoramidic hypothesis is exemplified when yeast CD is compared with
mustard, 79, after the 1,4 elimination of acrolein, 80. This bacterial CD (Kievit et al., 1999). The yeast enzyme, which
class of prodrug was one of the first types of self-immolative proved more effective than its bacterial counterpart, has a
prodrug to be synthesized in cancer chemotherapy, and the lower Km and a higher Vmax. An alternative correlation that re-
mechanism of self-immolation is well known (Brock, 1989). sulted in the same conclusion was reported when the activity

SCHEME 18 Activation of self-immolative 2-(2, 6-dinitro-phenylamino)propionamide by NR.


GENE-DIRECTED ENZYME PRODRUG THERAPY 149

SCHEME 19 Activation of CP and IF by CYP4B1 via aldophosphamide followed by 1,4-elimination. CP, R1  H; R2  CH2CH2Cl; IF, R1 
CH2CH2Cl; R2  H.

of CP (higher kcat, lower IC50) was compared with that of IP volved directly or indirectly influence this process. It is there-
(lower kcat, higher IC50) (Chang et al., 1993). fore useful to define the degree of activation that reflects the
It is not yet known if a slow, constant release of the active efficiency of the system in a transfected cell line (as defined
drug is preferable to a rapid release. One possibility is that here in Section 2). Theoretically, it will be lower than or at
for drugs acting against both quiescent and proliferating cells, best equal to the potential of activation. This was found to be
the former is likely to be the appropriate choice. By contrast, the case for all the systems analyzed in Table 1.
for drugs acting only against proliferating cells the second al- One of the drawbacks of these parameters is that although
ternative may be preferable. they give us an image of the behavior of a GDEPT system in
vitro, they may not reflect accurately the situation in vivo. A
number of additional factors, such as pharmacokinetics,
B. Potential of Activation
biodistribution, and immunologic effects, serve to complicate
It is clear that enzyme-prodrug systems should be de- the overall picture. However, on a rational basis, these para-
signed with as large as possible activation potential. How- meters are useful for comparing different GDEPT systems
ever, not all systems can be described in this way since the and should also be helpful in designing new systems.
cytotoxicities of the released drugs are sometimes unavail- There are means to increase the efficacy of a GDEPT sys-
able. Recently, a new mechanism for the cytotoxicity of GCV tem. One is to increase the available concentration of the pro-
was proposed based on the ability of ganciclovir nucleotide drug. An improved uptake of the prodrug is important for the
triphosphate (GCVTP) to be incorporated into DNA com- efficacy of the enzyme expressed intracellularly in GDEPT
bined with the intrinsic cytotoxicity for ganciclovir nucleotide systems. The concomitant expression of E. coli CD and uracil
monophosphate (GCVMP), 21 (Rubsam et al., 1998). There- phosphoribosyltransferase (UPRT) significantly improved the
fore, the potential of activation of this system (as defined here cytotoxicity of 5-FC. It was shown that the combination of the
in Section 2) calculated solely on the basis of IC50 of GCVTP, two genes facilitated the uptake of 5-FC by direct channeling
22, appears to be inaccurate. Another interesting mechanism of 5-FU (the product of CD activation) to 5-fluorouridine
is for CP activated by CYT P450. A maximum differential of monophosphate by the second enzyme in the cascade, UPRT
20- to 25-fold is theoretically achievable based on the IC50 (Tiraby et al., 1996).
values of CP and its corresponding phosphoramide mustard.
However, the degree of activation was found to be greater
than 100-fold (Springer and Niculescu-Duvaz, 2000), which 6. Augmenting the Effect
suggests that the CP phosphoramide mustard is not the final
active metabolite. One way of increasing the efficiency of a GDEPT system
is to mutate the enzyme for surface-tethered or external ex-
pression. The potential advantages of extracellular expression
C. Degree of Activation
are twofold. It should give an improved BE since the drug
The activation process is not always easy to understand. will be generated in the tumor interstitium rather than inside
The physicochemical, electronic, and steric parameters in- the cell. Also, the prodrug need not enter the cell to became
150 SPRINGER AND NICULESCU-DUVAZ

activated, and therefore prodrugs that are excluded from cells demonstrated that double-suicide gene therapy (HSV-
can be exploited. TKCD), but not transfer of either gene individually, allowed
CPG2 was the first enzyme mutated for expression teth- the elimination of human carcinoma cell lines in vitro and in
ered to the outer cell membrane (Marais et al., 1997). Muta- vivo (Uckert et al., 1998). Generally, such strategies have led
tion of three glycosylation sites (Asn222, Asn264, and to therapeutic improvements in vivo (Rogulski et al., 1997).
Asn272) to the conserved amino acid Gln was required
to produce a protein stCPG2(Asn222Gln, Asn264Gln,
Asn272Gln) capable of activating CMDA, 1(R  H, Z  —, 7. Exploiting the Bystander Effect and
X  Cl, and Y  mesyl). The mutated enzyme exhibits the Acquired Immunity
same substrate requirements compared to the wild-type (Km 
7 M for MTX), with a somewhat lower but still effective A crucial component of GDEPT is that the released drug
turnover (kcat  3500 min1). The system based on external should be capable of inducing a BE. Most of the systems re-
expressed enzyme showed advantages in term of BE (Marais ported exhibit a significant BE, at least in vivo. The mecha-
et al., 1997). nisms of the BE have been intensively investigated and include
Enzyme such as secreted -Glu (Weyel et al., 2000) has the generation of active metabolites, gap junction transport,
recently been described. Abstracts but not publications illus- and export of cytotoxic metabolites able to kill nontransfected
trating other secreted enzymes as -L (Moore et al., 1997), neighboring cells as well as immune responses.
CD (Rehemtulla et al., 1997), and PGA (Moore et al., 1997) It is difficult to compare the BE from different systems
have been published. due to the different conditions employed. A quantitative ex-
CPA is an enzyme that was expressed in a secretory form pression of the BE was proposed using the NR/CB1954 sys-
to improve its BE. The secretory CPAST3 was constructed by tem in a range of human tumor cell types. The IC50 values of
introducing a decapeptide (GLSARNRQKR) between the non-NR-expressing cells were measured in the presence of
prodomain and the mature domain of rat CPA (Smith et al., differing proportions of NR-expressing cells. The shift in
1997). The expression of this mutated enzyme in cancer cells IC50 was used to calculate a value for the BE, termed the
is responsible for their sensitization to MTX- -substituted transmission efficiency (TE), which is the decrease in IC50
prodrugs, 6. An externally expressed CPADAF, tethered to the due to the BE as the percentage of the maximum decrease
cell, was also reported (Hamstra et al., 2000). possible. The percentage of NR-expressing cells for which
A problem with the CPA/MTX- -substituted prodrugs sys- the TE was 50% (TE50) is a single datum point for the BE.
tem is that the prodrugs are not stable in vivo as they are hy- The TE50 in the cell lines ranged from 0.3% to approxi-
drolyzed by endogenous CPA. New bulky phenylalanine and mately 2% (Friedlos et al., 1998).
tyrosine (substituted in position 2 or 3 with t-butyl, The toxic nucleoside phosphates resulting from the acti-
cyclopropyl-, cyclobutyl-, or cyclopropyl moieties) of MTX vation of purine (GCV, ACV, etc.) or pyrimidine (Ara-C, 5-
(i.e., MTX- -3-cyclobutylphenylalanine, 8a, or MTX- -3- FDUR, etc.) nucleosides are nondiffusible across cell mem-
cyclopentyltyrosine, 8b) were synthesized. These compounds branes and require a cell-to-cell contact for a BE (Freeman et
are not substrates for the wild-type enzyme and therefore are al., 1993; Manome et al., 1996). By contrast, for diffusible
stable in vivo. A mutant of the hCPA1 was produced in which drugs like aldophosphamide (or phosphoramide mustard), 6-
Thr268 was changed with Gly (hCPA1-T268G) to make more MeP, 2-FA, and CMDB, no such requirement is necessary
room for the substrate (Smith et al., 1997). This mutant can hy- (Huber et al., 1994; Bridgewater et al., 1995; Chen and Wax-
drolyze the bulky prodrugs 8a and 8b with good kinetics (kcat/Km man 1995).
are 1.8 and 0.16 M1s1 for 8a and 8b, respectively). The permeable toxic metabolites formed following pro-
Using similar techniques, mutants of HSV-TK were ob- drug activation are released by efflux from dead and dying
tained with improved kinetic parameters for GCV and ACV genetically modified cells. This mechanism is postulated for
(Black et al., 1996). Another way to enhance GDEPT effi- the metabolites resulting from 5-FC, CP, IP, CMDA, 6-MeP,
ciency is by using two or more suicide genes expressed sep- linamarin, and H2O2 generated by oxidation of D-amino acids.
arately or as a fusion gene. “Double-suicide gene therapy” The BE requires different mechanisms with purine and pyrim-
has been reported wherein a combination of suicide genes is idine nucleosides since the toxic phosphorylated metabolites
introduced simultaneously to augment the effects. The ratio- cannot diffuse across cell membranes. The HSV-TK/GCV
nale is that the released active drugs act by different mecha- system that releases nonpermeable metabolites requires cell-
nisms leading to a synergistic cell kill (Aghi et al., 1999; to-cell contact for the BE. However, the mechanism is com-
Blackburn et al., 1999). plex. The transfer of cytotoxic GCV metabolites from HSV-
Resistant cell populations are less likely to occur when TK-transfected cells to wild-type tumor cells via gap junctions
drugs with different mechanisms of action are used. It was has been demonstrated as a BE mechanism (Touraine et al.,
GENE-DIRECTED ENZYME PRODRUG THERAPY 151

1998a). When gap junction function was evaluated with a dye be induced either by stimulation of the host immune system
transfer technique, tumor cells resistant to the BE did not or by the use of additional cytokine gene therapy. Long-
show dye transfer from cell to cell, whereas BE-sensitive tu- lasting immunity in immunocompetent animals has developed
mor cells did. It was suggested that enhancement of the HSV- as a response to HSV-TK transduction followed by GCV treat-
TK/GCV BE could be achieved by pharmacologic manipula- ment (Pavlovic et al., 1996). These studies suggest that an in-
tion of the gap junctions in vivo. Dieldrin, 81 (Scheme 20), a tact immune system is important for long-term tumor sup-
drug known to decrease gap junction communications, di- pression with HSV-TK in vivo (Gagandep et al., 1996).
minished the dye transfer and also inhibited the BE. Api- Additional cytokine genes have been examined. Mice
genin, 82, a flavonoid, and lovastatin, 83, an HMG-CoA re- treated with both HSV-TK and IL-2 genes developed effec-
ductase inhibitor, were shown to up-regulate gap junction tive systemic antitumoral immunity against tumorigenic
function and dye transfer in tumors expressing gap junctions rechallenges. In an attempt to enhance and prolong the im-
and to enhance the BE in vivo (Touraine et al., 1998a, 1998b). munity, a third vector containing the mouse granulocyte-
macrophage colony-stimulating factor (mGM-CSF) gene was
employed. The animals treated simultaneously with HSV-
TKIL-2mGM-CSF vectors followed by administration of
GCV developed long-term antitumor immunity and survived
for more than 4 months without recurrence (Chen et al.,
1995, 1996b)

8. Conclusions

To date GDEPT systems have shown efficacy in vivo.


There is still room for improvement. The enzyme can be mu-
tated to obtain greater efficiency of activation for a given pro-
drug. Alternatively, the active site may be mutated to provide
greater specificity to the prodrugs as substrates for the trans-
fected genes. The synthesis of new prodrugs, such as self-
immolative prodrugs, prodrugs releasing more cytotoxic drugs,
drugs able to optimize the BE effect, or prodrugs taking ad-
SCHEME 20 Compounds influencing gap junction communications. vantage of carrier in order to cross cell membranes, should be
also considered. The design of optimized enzyme-prodrug
systems and the use of cocktails of prodrugs releasing drugs
There are data that appear to contradict the gap junction with different mechanisms of action, which could be activated
hypothesis and suggest that other mechanisms may be in- by the same enzyme, could also lead to improvements.
volved. Under different experimental conditions it was found Additional strategies are being aimed at improving the
that the BE depended on the concentration of the enzyme, the therapy for solid tumors. For example, combining GDEPT
number of cells expressing HSV-TK, and the overall conflu- with radiotherapy or immunotherapy has been suggested.
ence of the cells, and that the BE did not correlate with gap Such an approach may involve either a sequential treatment
junction functional communications as determined by the Lu- schedule (GDEPT/radiation therapy or GDEPT/immunother-
cifer yellow assay (Boucher et al., 1998; Imaizumi et al., apy) or the transfection of suicide gene(s) together with genes
1998). capable of increasing the sensitivity of the tumors to radia-
When the enzyme is expressed extracellularly the drug is tion or enhancing the potential of the host immune system
released outside the cells and an improved BE would be ex- with cytokine genes. Ultimately, delivery and targeting are
pected. The released drugs should be highly diffusible, and the crux of efficacy in GDEPT.
prodrugs releasing permeable metabolites are still favored.
Elicitation of an immune response has been described as
a positive factor in GDEPT. Although data are available that Acknowledgments
show that the BE occurs in immunocompromised animals,
other reports show that the BE is mediated through the re- This work was funded by the Cancer Research Campaign
lease of cytokines in vivo (Pavlovic et al., 1996; Ramesh et (grants SP2330/0201 and SP2330/0102) and the Institute of
al., 1996; Hall et al., 1998). Beneficial immune effects may Cancer Research.
152 SPRINGER AND NICULESCU-DUVAZ

References 450 gene transfer: Development of a combined chemotherapy/cancer


gene therapy strategy. Cancer Res. 55, 581–589.
Aghi, M., Chou, T. C., Suling, K., Breakefield, X. O., and Chiocca, I. A. Chen, L., Waxman, D. J., Chen, D., and Kufe, D. C. (1996a). Sensitization
(1999). Multimodal cancer treatment mediated by a replicating oncolytic of human breast cancer cells to cyclophosphamide and ifosfamide by
virus that delivers the oxazaphosphorine/rat cytochrome P450 2B1 and transfer of a liver cytochrome P-450 gene. Cancer Res. 56, 1331–1340.
ganciclovir/herpes simplex virus thymidine kinase gene therapies. Can- Chen, S. H., Kosai, K., Xu, B., Pham-Nguyen, K., Contant, C., Finegold,
cer Res. 59, 3861–3865. M. J., and Woo, S. L. C. (1996b). Combination suicide and cytokine gene
Andrianomenjanahary, S., Dong, X., Florent, J. C., Gaudel, G., Gesson, J. P., therapy for hepatic metastases of colon carcinoma: Sustained antitumor
Jacquesy, J. C., Koch, M., Michel, S., Mondon, M., Monneret, C., Petit, immunity prolongs animal survival. Cancer Res. 56, 3758–3762.
P., Renoux, B., and Tillequin, F. (1992). Synthesis of novel targeted pro- Chen, L., Yu, L. J., and Waxman, D. J. (1997). Potentiation of cytochrome
drugs of anthracyclines potentially activated by a monoclonal antibody P450/cyclophosphamide-based cancer gene therapy by coexpression of
galactosidase conjugate (Part 1). Bioorg. Med. Chem. Lett. 2(9), the P450 reductase gene. Cancer Res. 57, 4830–4837.
1093–1096. Chen, S. H., Li Chen, X. L., Wang, Y., Kosai, K., Finegold, M. J., Rich,
Anlezark, G. M., Melton, R. G., Sherwood, R. F., Wilson, W. R., Denny, S. S., and Woo, S. L. C. (1995). Combination gene therapy for liver
W. A., Palmer, B. D., Knox, R. G., Friedlos, F., and Williams, A. (1995). metastasis of colon carcinoma in vivo. Proc. Natl. Acad. Sci. USA 92,
Bioactivation of dinitrobenzamide mustards by an E. coli B nitroreduc- 2577–2581.
tase. Biochem. Pharmacol. 50, 609–618. Culver, K. W., Ram, Z., Wallbridge, S., Oldfield, H. E. H., and Blaese,
Averett, D. R., Kozalka, G. A., Fyfe, J. A., Roberts, G. B., Purifoy, D. J. M., M. R. (1992). In vivo gene transfer with retroviral vector-producer cells
and Krenitsky, T. A. (1991). “6-methoxypurine arabinoside as a selective for treatment of experimental brain tumors. Science 256, 1550–1552.
and potent inhibitor of varicella-zoster virus.” Antimicrob. Agents Denny, W. A., and Wilson, W. R. (1998). The design of selectively-activated
Chemother. 35(5), 851–857. anti-cancer prodrugs for use in antibody-directed and gene-directed en-
Balzarini, G., Degreve, B., Andrei, G., Neyst, J., Sandvold, M., Myhren, F., zyme prodrugs therapies. J. Pharm. Pharmacol. 50(4), 387–394.
and de Clerq, E. (1998). Superior cytostatic activity of the ganciclovir Dowell, R. I., Springer, C. J., Davies, D. H., Hadley, E. M., Burke, P. J.,
elaidic acid ester due to the prolonged intracellular retention of ganci- Boyle, F. T., Melton, R. G., Connors, T. A., Blakey, D. C., and Mauger,
clovir anabolites in herpes simplex virus type 1 thymidine kinase gene- A. B. (1996). New mustard prodrug for antibody-directed enzyme pro-
transfected tumor cells. Gene Ther. 5, 419–426. drug therapy: Alternative to amide link. J. Med. Chem. 39, 1100–1105.
Bielicki, L., Voelcker, G., and Hohorst, H. J. (1984). Activated cyclophos- Encell, L. P., Landis, D. M., and Loeb, L. A. (1999). Improving enzymes for
phamide: an enzyme-mechanism-based suicide inactivator of DNA poly- gene therapy. Nat. Biotechnol. 17, 143–147.
merase/3, 5 exonuclease. J. Cancer Res. Clin. Oncol. 107, 195–198. Evrard, A., Cuq, P., Ciccolini, J., Vian, L., and Cano, J.-P. (1999). Increased
Bilbao, G., Contreras, J. L., Gómez-Navarro, J., and Curiel, D. T. (1998). Im- cytotoxicity and bystander effect of 5-fluorouracil and 5-deoxy-5-
proving adenoviral vectors for cancer gene therapy. Tumor Targeting 3, fluorouridine in human colorectal cancer cells transfected with thymidine
59–79. phosphorylase. Br. J. Cancer 80(11), 1726–1733.
Black, M. E., Newcomb, T. G., Wilson, H. M., and Loeb, L. A. (1996). Cre- Farquhar, D., Cherif, A., Bakina, E., and Nelson, G. A. (1998). Intensely po-
ation of drug-specific herpes simplex virus type 1 thymidine kinase mu- tent doxorubicin analogues: Structure-activity relationships. J. Med.
tants for gene therapy. Proc. Natl. Acad. Sci. USA 93, 3525–3529. Chem. 41, 965–972.
Blackburn, R. V., Galoforo, S., Corry, P. M., and Lee, Y. J. (1999). Aden- Frankel, A. E., FitzGerald, D., Siegall, C., and Press, O. W. (1996). Advances
oviral transduction of a cytosine deaminase/thymidine kinase fusion gene in immunotoxin biology and therapy: a summary of the Fourth Interna-
into prostate carcinoma cells enhances prodrug and radiation sensitivity. tional Symposium on Immunotoxins. Cancer Res. 56, 926–932.
Int. J. Cancer 82, 293–297. Freeman, S. M., Abboud, C. N., Whartenby, K. A., Packman, C. H., Koeplin,
Blaise Smith, P., Tiano, H. F., Nesnow, S., Boyd, M. R., Philpot, R. M., and D. S., Moolten, F. S., and Abraham, G. N. (1993). The “bystander ef-
Langenbach, R. (1995). 4-Ipomeanol and 2-aminoanthracene cytotoxic- fect”: Tumor regression when a fraction of the tumor mass is genetically
ity in C3H/10T1/2 cells expressing rabbit cytochrome P450 4B1. modified. Cancer Res. 53, 5274–5283.
Biochem. Pharmacol. 50, 1567–1575. Friedlos, F., Court, S., Ford, M., Denny, W. A., and Springer, C. J. (1998).
Boland, M. P., Knox, R. J., and Roberts, J. J. (1991). The differences in ki- “Gene-directed enzyme prodrug therapy: quantitative bystander cytotox-
netics of rat and human DT-diaphorase result in in a differential sensi- icity and DNA damage induced by CB1954 in cells expressing bacterial
tivity of cells lines to CB 1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide. nitroreductase.” Gene Ther. 5(1), 105–112.
Biochem. Pharmacol. 41, 867–875. Friedlos, F., Denny, W. A., Palmer, B. D., and Springer, C. J. (1997). Mus-
Boucher, P. D., Ruch, R. J., and Shewach, D. S. (1998). Differential tard prodrugs for activation by Escherichia coli nitroreductase in gene-
ganciclovir-mediated cytotoxicity and bystander killing in human colon directed enzyme prodrug therapy. J. Med. Chem. 40(8), 1270–1275.
carcinoma cell lines expressing herpes simplex virus thymidine kinase. Gagandep, S., Brew, R., Green, B., Christmas, S. E., Klatzmann, D., Poston
Human Gene Ther. 9(6), 801–814. G. J., and Kinsella, A. R. (1996). Prodrug-activated gene therapy: In-
Bridgewater, G., Springer, C. J., Knox, R., Minton, N., Michael, P., and volvement of an immunological component in the “bystander effect”.
Collins, M. (1995). Expression of the bacterial nitroreductase enzyme in Gene Ther. 3(2), 83–88.
mammalian cells renders them selectively sensitive to killing by the pro- Gesson, J. P., Jacquesy, J. C., Mondon, M., Petit, P., Renoux, B., Andri-
drug CB1954. Eur. J. Cancer 31A(13/14), 2362–2370. anomenjanahary, S., Dufat-Trinh Van, H., Koch, M., Michel, S., Tillequin,
Brock, N. (1989). Oxazaphosphorine cytostatics: past-present-future. Cancer F., Florent, J. C., Monneret, C., Bosslet, K., Czech, J., and Hoffmann, D.
Res. 49, 1. (1994). Prodrugs of anthracyclines for chemotherapy via enzyme-mono-
Chang, T. K. H., Weber, G. F., Crespi, C. L., and Waxman, D. J. (1993). Dif- clonal antibody conjugates. Anticancer Drug Des. 9, 409–423.
ferential activation of cyclophosphamide and ifosphamide by cy- Ghosh, A. K., Khan, S., and Farquhar, D. (1999). “A -galactosidase phos-
tochromes P450 2B and 3A in human liver microsomes. Cancer Res. 53, phoramide mustard prodrug for use in conjunction with gene-directed
5629–5637. enzyme prodrug therapy.” Chem. Communications 2527–2528.
Chen, L., and Waxman, D. J. (1995). Intratumoral activation and enhanced Golumbek, P. T., Hamzeh, F. M., Jaffee, E. M., Levitsky, H., Lietman, P. S.,
chemotherapeutic effect of oxazaphosphorines following cytochrome P- and Pardoll, D. M. (1992). Herpes simplex-1 virus thymidine kinase
GENE-DIRECTED ENZYME PRODRUG THERAPY 153

gene is unable to completely eliminatelive, nonimmunogenic tumour cell (aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954) to 5-(aziridin-1-yl)-4-
vaccines. J. Immunother. 12, 224–230. hydroxylamino-2-nitrobenzamide is a form of NAD(P)H dehydrogenase
Greenwald, R. B., Pendri, A., Conover, C. D., Zhao, H., Choe, Y. H., Mar- (quinone) (EC 1.6.99.2). Biochem. Pharmacol. 44, 2297–2301.
tinez, A., Shum, K., and Guan, S. (1999). Drug delivery system employ- Knox, R. J., Friedlos, F., Biggs, P. J., Fliter, W. D., Gaskell, M., Goddard, P.,
ing 1,4- or 1,6-elimination: Poly(ethylene glycol) prodrugs of amine- Davies, L., and Jarman, M. (1993). Identification, synthesis and proper-
containing compounds. J. Med. Chem. 42, 3657–3667. ties of 5-(aziridin-1-yl)-2-nitro-4-nitrosobenzamide, a novel DNA cross-
Hall, S. J., Sanford, M. A., Atkinson, G., and Chen, S.-H. (1998). Induction linking agent derived from CB 1954. Biochem. Pharmacol. 46, 797–803.
of potent antitumor natural killer cell activity by herpes simplex virus- Knox, R. J., Friedlos, F., Sherwood, R. F., Melton, R. G., and Anlezark,
thymidine kinase and ganciclovir therapy in an orthotopic mouse model G. M. (1992). The bioactivation of 5-(aziridin-1-yl)-2,4-dinitrobenzamide
of prostate cancer. Cancer Res. 58, 3221–3225. (CB 1954). II. A comparison of an Escherichia coli nitroreductase and
Halliwell, B., and Gutteridge, J. M. (1989). “Free Radicals in Biology and Walker DT-diaphorase. Biochem. Pharmacol. 44, 2297–2301.
Medicine.” Clarendon Press, Oxford. Kojima, A., Hackett, N. R., and Crystal, R. G. (1998a). Reversal of CPT-11
Hamstra, D. A., Page, M., Maybaum, J., and Rehemtulla, A. (2000). Ex- resistance of lung cancer cells by adenovirus-mediated gene transfer of
pression of endogenously activated secreted or cell surface carboxypep- the human carboxylesterase cDNA. Cancer Res. 58, 4368–4374.
tidase A sensitizes tumor cells to methotrexate-a-peptide prodrugs. Can- Kojima, A., Hackett, N. R., Ohwada, A., and Crystal, R. G. (1998b). In vivo
cer Res. 60, 657–665. human carboxylesterase cDNA gene transfer to activate the prodrug
Hapke, D. M., Stegmann, A. P. A., and Mitchell, B. S. (1996). Retroviral CPT-11 for local treatment of solid tumors. J. Clin. Invest. 101,
transfer of deoxycytidine kinase into tumor cell lines enhances nucleo- 1789–1796.
side toxicity. Cancer Res. 56, 2343–2347. MacCoss, M., and Robins, M. J. (1990). Anticancer pyrimidines, pyrimidine
Hasenburg, A., Tong, X. W., Rojas-Martinez, A., Nyberg-Hoffman, C., nucleosides and prodrugs. In “Chemistry of Antitumour Agents”
Kieback, C. C., Kaplan, A. L., Kaufman, R. H., Ramzy, I., Aguilar- D. E. V. Wilman, ed., pp. 261–298. Blackie & Son Ltd., Chapman &
Cordova, E., and Kieback, D. G. (1999). Thymidine kinase (TK) gene Hall, London, New York.
therapy of solid tumors: Valacyclovir facilitates outpatients treatment. Manome, Y., Wen, P. Y., Dong, Y., Tanaka, T., Mitchell, B. S., Kufe, D. W.,
Anticancer Res. 19, 2163–2166. and Fine, H. A. (1996). Viral vector transduction of the human deoxycy-
Hay, M. P., Sykes, B. M., Denny, W. A., and Wilson, W. R. (1999a). A 2- tidine kinase cDNA sensitizes glioma cells to the cytotoxic effects of cy-
nitroimidazole carbamate prodrug of 5-amino-1-(chloromethyl)-3-[(5,6,7- tosine arabinoside in vitro and in vivo. Nat. Med. 2(5), 567–573.
trimethoxyindol-2-yl)carbonyl]-1,2-dihydro-3H-benz[e]indole (amino- Marais, R., Spooner, R. A., Light, Y., Martin, J., and Springer, C. J. (1996).
seco-CBI-TMI) for use with ADEPT and GDEPT. Bioorg. Med. Chem. Gene-directed enzyme prodrug therapy with a mustard prodrug/car-
Lett. 9, 2237–2242. boxypeptidase G2 combination. Cancer Res. 56, 4735–4742.
Hay, M. P., Wilson, W. R., and Denny, W. R. (1999b). Nitobenzyl carbamate Marais, R., Spooner, R. A., Stribbling, S. M., Light , Y., Martin, J., and
prodrugs of enediynes for nitroreductase gene-directed enzyme prodrug Springer, C. J. (1997). A cell surface tethered enzyme improves efficiency
therapy. Bioorg. Med. Chem. Lett. 9, 3417–3422. in gene-directed enzyme prodrug therapy. Nat. Biotechnol. 15, 1373–1377.
Huber, B. A., Richards, C. A., and Krenitsky, T. A. (1991). Retroviral- Melton, R. G., and Sherwood, R. F. (1997). Antibody-enzyme conjugates for
mediated gene therapy for the treatment of hepatocellular carcinoma: An cancer therapy. J. Natl. Cancer Instit. 88, 153–160.
innovative approach for cancer therapy. Proc. Natl. Acad. Sci., USA 88, Miki, K., Xu., M., An, Z., Wang, X., Yang, M., Al-Refaie, W., Sun, X., Bara-
8039–8043. nov, E., Tan, Y., Chishima, T., Shimada, H., Moossa, A. R., and Hoffman,
Huber, B. E., Austin, E. A., Richards, C. A., Davis, S. T., and Good, S. S. R. M. (2000). Survival efficacy of the combination of the methioninase
(1994). Metabolism of 5-fluorocytidine to 5-fluorouracil in human col- gene and methioninase in a lung cancer orthotopic model. Cancer Gene
orectal tumor cells transduced with the cytosine deaminase gene: Signif- Ther. 7(2), 332–338.
icant antitumor effects when only a small percentage of tumor cells ex- Miller, A. D. (1998). Cationic liposomes for gene therapy. Angew. Chem. Int.
press cytosine deaminase. Proc. Natl. Acad. Sci. USA 91, 8302–8306. Ed. 37, 1768–1785.
Imaizumi, K., Hasegawa, Y., Kawabe, T., Emi, N., Saito, H., Naruse, K., and Moore, J., Ohmstede, C., Dickerson, S., Chu, L., Sethna, P., Davis, S., and
Shimokata, K. (1998). Bystander tumoricidal effect and gap junctional Dev, I. (1997). Gene therapy utilizing enzymes capable of extracellular
communication in lung cancer cells. Am. J. Respir. Cell Mol. Biol. 18, prodrug conversion of multiple prodrugs. Proc. Am. Assoc. Cancer Res.
205–212. 38, 379 (abstract 2544).
Izquierdo, M., and Corts, M. L. (1999). Suicide gene therapy system for the Mullen, C. A., Kilstrup, M., and Blaese, R. M. (1992). Transfer of the bac-
treatment of brain tumours. WO 9947653 Boehringer Ingelheim Interna- terial gene for cytosine deaminase to mammalian cells confers lethal sen-
tional, 39 pp. sitivity to 5-fluorocytosine: A negative selection system. Proc. Natl.
Johansson, M., Van Rompey, A. R., Degreves, B., Balzarini, J., and Karls- Acad. Sci. USA 89, 33–37.
son, A. (1999). Cloning and characterization of the multisubstrate de- Mulligan, R. C., and Berg, P. (1981). Selection for animal cells that express
oxyribonucleoside kinase of Drosophila melanogaster. J. Biol. Chem. the Escherichia coli gene coding for xanthine-guanine phosphoribosyl-
274(34), 23814–23819. transferase. Proc. Natl. Acad. Sci. USA 78(4), 2072–2076.
Jungheim, L. N., and Shepherd, T. T. (1994). Design of antitumor prodrugs: Nguyen, J. T., Wu, P., Clouse, M. E., Hlatky, L., and Terwilliger, E. F.
substrates for antibody targeted enzymes. Chem. Rev. 94, 1553–1566. (1998). Adeno-associated virus-mediated delivery of antiangiogenic fac-
Kievit, E., Bershad, E., Ng, E., Sethna, P., Dev, I., Lawrence, T. S., and Re- tors as an antitumor stratergy. Cancer Res. 58, 5673–5677.
hemtulla, A. (1999). Superiority of yeast over bacterial cytosine deami- Niculescu-Duvaz, D., Niculescu-Duvaz, I., Friedlos, F., Martin, J., Spooner,
nase for enzyme/prodrug gene therapy in colon cancer. Cancer Res. 59, R., Davies, L., Marais, R., and Springer, C. J. (1998). Self-immolative
1417–1421. nitrogen mustard prodrugs for suicide gene therapy. J. Med. Chem.
Kinsella, A. R., Smith, D., and Pickard, M. (1997). Resistance to chemother- 41(26), 5297–5309.
apeutic antimetabolites: a function of salvage pathway involvement and Niculescu-Duvaz, I., Friedlos, F., Niculescu-Duvaz, D., Davies, L., and
cellular response to DNA damage. Br. J. Cancer 75(7), 935–945. Springer, C. J. (1999a). “Prodrugs for antibody- and gene-directed en-
Knox, R. J., Boland, M. P., Friedlos, F., Coles, B., Southan, C., and Roberts, zyme prodrug therapies (ADEPT and GDEPT).” Anticancer Drug De-
J. J. (1988). The nitroreductase enzyme in Walker cells that activates 5- sign 14, 517–538.
154 SPRINGER AND NICULESCU-DUVAZ

Niculescu-Duvaz, I., Niculescu-Duvaz, D., Friedlos, F., Spooner, R., Martin, Smith, G. K., Banks, S., Blumenkopf, T. A., Cory, M., Humphreys, J.,
J., Marais, R., and Springer, C. J. (1999b). Self-immolative anthracycline Laethem, A. M., Miller, G., Moxham, C. P., Mullin, R., Ray, P. H., Wal-
prodrugs for suicide gene therapy. J. Med. Chem. 42(13), 2485–2489. ton, L. M., and Wolfe III, L. A. (1997). Toward antibody-directed en-
Niculescu-Duvaz, I., Spooner, R., Marais, R., and Springer, C. J. (1998). zyme prodrug therapy with the T268G mutant of human carboxypepti-
Gene-directed enzyme prodrug therapy. Bioconjug. Chem. 9(1), 4–22. dase A1 and novel in vivo stable prodrugs of methotrexate. J. Biol. Chem.
Niculescu-Duvaz, I., and Springer, C. J. (1995). Antibody-directed enzyme 272, 15804–15816.
prodrug therapy (ADEPT): A targeting strategy in cancer chemotherapy. Sorscher, E. J., Peng, S., Bebock, Z., Allan, P. W., Bennett Jr., L. L., and
Curr. Med. Chem. 2, 687–706. Parker, W. B. (1994). Tumor cell bystander killing in colonic carcinoma
Pandha, H. S., Martin, L. A., Rigg, A., Hurst, H. C., Stamp, G. W. H., Sikora, utilizing the Escherichia coli deo-D gene to generate toxic purines. Gene
K., and Lemoine, N. R. (1999). Genetic prodrug activation therapy for Ther. 1, 233–238.
breast cancer: A phase I clinical trial of erbB-2-directed suicide gene ex- Springer, C. J., Antoniw, P., Bagshawe, K. D., Searle, F., Bisset, G. M. F.,
pression. J. Clin. Oncol. 17, 2180–2189. and Jarman, M. (1990). Novel prodrugs which are activated to cytotoxic
Parker, W. B., Allan, P. W., Shaddix, S. C., Rose, L. M., Speegle, H. F., alkylating agents by carboxypeptidase G2. J. Med. Chem. 33, 677–681.
Gillepsie, G. Y., and Bennett, L. L. (1998). Metabolism and metabolic Springer, C. J., Bavetsias, V., Jackman, A. L., Boyle, T. F., Marshall, D., Ped-
actions of 6-methylpurine and 2-fluoroadenine in human cells. Biochem. ley, R. B., and Bisset, G. M. F. (1996). Prodrugs of thymidylate synthase
Pharmacol. 55, 1673–1681. inhibitors: potential for antibody-directed enzyme prodrug therapy
Patterson, A. V., Zhang, H., Moghaddam, A., Bicknell, R., Talbot, D. C., (ADEPT). Anticancer Drug Des. 11, 625.
Stratford, I. J., and Harris, A. L. (1995). Increased sensitivity to the pro- Springer, C. J., Dowell, R., Burke, P. J., Hadley, E., Davies, D. H., Blakey,
drug 5-deoxy-5-fluorouridine and modulation of 5-fluoro-2-deoxyuri- D. C., Melton, R. G., and Niculescu-Duvaz, I. (1995). Optimization of
dine sensitivity in MCF-7 cells transfected with thymidine phosphory- alkylating agent prodrug derived from phenol and aniline mustards: A
lase. Br. J. Cancer 72, 669–675. new clinical candidates prodrug (ZD2767) for antibody-directed prodrug
Patterson, L. H., McKeown, S. R., Robson, T., Gallagher, R., Raleigh, S. M.,
therapy (ADEPT). J. Med. Chem. 38(26), 5051–5065.
and Orr, S. (1999). Antitumour prodrug development using cythocrome
Springer, C. J., and Niculescu-Duvaz, I. (1999). Patent property of prodrug
P450 (CYP) mediated activation. Anticancer Drug Design 14, 473–486.
involving gene therapy (1996–1999). Exp. Opin. Ther. Patents. 9(10),
Pavlovic, J., Nawrath, M., Tu, R., Heinicke, T., and Moelling, K. (1996).
1381–1388.
Anti-tumor immunity is involved in the thymidine kinase-mediated killing
Springer, C. J., and Niculescu-Duvaz, I. (2000). Prodrug-activating systems
of tumors induced by activated Ki-ras (G12V). Gene Ther. 3, 635–643.
in suicide gene therapy. J. Clin. Invest. 105(9), 1161–1167.
Pollegioni, L., Falbo, B., and Pilone, M. S. (1992). Specificity and kinetics
Stegman, L. D., Zheng, H., Neal, E. R., Ben-Yoseph, O., Pollegioni, L., Pil-
of Rodotorula gracilis D-amino-acid oxidase. Biochim. Biophys. Acta
lone, M. S., and Ross, B. D. (1998). Induction of cytotoxic oxidative
120, 11–16.
stress by D-alanine in brain tumor cells expressing Rhodotorula gracilis
Rainov, N. G., Dobberstein, K.-U., Sena-Estevez, M., Herlinger, U., Kramm,
D-amino acid oxidase: A cancer gene therapy strategy. Human Gene
C. M., Philpot, R. M., Hilton, J., Chiocca, E. A., and Breakefield, X. O.
(1998). New prodrug activation gene therapy for cancer using cytochrome Ther. 9, 185–193.
P450 4B1 and 2-aminoanthracene/4-ipomeanol. Human Gene Ther. 9, Stribbling, S. M., Friedlos, F., Martin, J., Davies, L., Spooner, R. A., Marais,
1261–1273. R., and Springer, C. J. (2000). Regressions of established breast cancer
Ramesh, R., Marrogi, A. J., Munshi, A., Abboud, C. N., and Freeman, S. M. xenografts by carboxypeptidase G2 suicide gene therapy and the pro-
(1996). In vivo analysis of the “bystander effect”: A cytokine cascade. drug CMDA are due to a bystander effect. Human Gene Ther. 11,
Exp. Hematol. 24, 829–838. 285–292.
Rehemtulla, A., Chang, E., Davis, M. A., and Lawrence, T. S. (1997). Ex- Sykes, B. M., Atwell, G. J., Hogg, A., Wilson, W. R., O’Connor, C. J., and
tracellular targeting of cytosine deaminase results in prolonged produc- Denny, W. R. (1999). N-substituted 2-(2,6-dinitrophenylamino)-
tion of 5-fluorouracil compared to intracellular cytosine deaminase. Proc. propanamides: Novel prodrugs that release a primary amine via nitrore-
Am. Assoc. Cancer Res. 38, 381 (abstract 2555). duction and intramolecular cyclization. J. Med. Chem. 42, 346–355.
Robbins, P. D., and Ghivizzani, S. C. (1998). Viral vectors for gene therapy. Tamiya, T., Ono, Y., Wei, M. X., Mroz, P. J., Moolten, F. L., and Chiocca,
Pharmacol Ther. 80(1), 35–47. E. A. (1996). Escherichia coli gpt gene sensitizes rat glioma cells to
Rogulski, K. R., Zhang, K., Kolozsvary, A., Kim, J. H., and Freitag, S. O. killing by 6-thioxanthine or 6-thioguanine. Cancer Gene Ther. 3(3),
(1997). Pronounced antitumor effect and tumor radiosensitization of 155–162.
double suicide gene therapy. Clin. Cancer Res. 3, 2081–2088. Tatcher, N. J., Edwards, R. J., Lemoine, N. R., Doehmer, J., and Davies,
Roth, J. A., and Cristiano, R. G. (1997). Gene therapy for cancer: what have D. S. (2000). The potential of acetaminophen as a prodrug in gene-
we done and where are we going? J. Natl. Cancer Instit. 89, 21–30. directed enzyme prodrug therapy. Cancer Gene Ther. 7(4), 521–525.
Rubsam, L. Z., Davidson, L., and Shewach, D. S. (1998). Superior Tiraby, G., Reynes, J.-P., Tiraby, M., Cazaux, C., and Drocourt, D. (1996).
cytotoxicity with gancyclovir compared with acyclovir and 1- -D- Suicide genes and combinations of pyrimidine nucleoside and nucle-
arabinofuranosylthymine in herpes simplex virus-thymidine kinase- obase analogues with suicide genes for gene therapy. WO 9616183
expressing-cells: a novel paradigm for cell killing. Cancer Res. 58, 3873–3882. Cayla, France, 47 pp.
Secrist III, J. A., Parker, W. B., Allan, P. W., Bennett, L. L., Waud, W. R., Touraine, R. L., Ishii-Morita, H., Ramsey, W. J., and Blaese, R. M. (1998a).
Truss, J. W., Fowler, A. T., Montgomery, J. A., Ealick, S. E., Gillepsie, The bystander effect in the HSVtk/ganciclovir system and its relation to
G. Y., Gadi, V. K., and Sorscher, E. J. (1999). Gene therapy of cancer: gap junctional communication. Gene Ther. 5, 1705–1711.
Activation of nucleoside prodrugs with E. coli purine nucleoside phos- Touraine, R. L., Vahanian, N., Ramsey, W. J., and Blaese, R. M. (1998b). En-
phorylase. Nucleosides Nucleotides 18(4, 5), 745–757. hancement of the herpes simplex virus thymidine kinase/ganciclovir by-
Sethna, P., Talarico, T., Merrill, B., Moore, J., Davis, S., Dikerson, S., and stander effect and its antitumor efficacy in vivo by pharmacologic ma-
Dev, I. (1997). Cloning of the gene of a multiple drug activation enzyme nipulation of gap junctions. Human Gene Ther. 9(16), 2385–2391.
from tomato for viral-directed enzyme prodrug therapy (VDEPT) for Uckert, W., Kammertons, T., Haack, K., Qin, Z., Gebert, J., Schendel, D. J.,
cancer. Pro. Am. Assoc. Cancer Res. 38, 381 (abstract 2554). and Blankenstein, T. (1998). Double suicide gene (cytosine deaminase
GENE-DIRECTED ENZYME PRODRUG THERAPY 155

and herpes simplex virus thymidine kinase) but not single gene transfer P450-based cancer gene therapy: Recent advances and future prospects.
allows reliable elimination of tumor cells in vivo. Human Gene Ther. 9, Drugs Metab. Rev. 31(2), 503–522.
855–865. Weyel, D., Sedlacek, H.-H., Muller, R., and Brusselbach, S. (2000). Secreted
Wakselman, M. (1983). 1,4- and 1,6-elimination from hydroxy- and amino- human -glucuronidase: a novel tool for gene-directed enzyme prodrug
substituted benzyl systems: Chemical and biochemical application. Nou- therapy. Gene Ther. 7, 224–231.
veau J. Chimie 7, 439–447. Wierdl, M., Morton, C. L., and Potter, P. M. (2000). Development of aden-
Warrington, K. H., Teschendorf, C., Cao, L., Muzyczka, N., and Siemann, ovirus expressing a secreted rabbit liver carboxylesterase for VDEPT
D. W. (1998). Developing VDEPT for DT-diaphorase (NQO1) using an with CPT-11. Proc. Am. Assoc. Cancer Res. 41, 671 (abstract 4266).
AAV vector plasmid. Int. J. Radiat. Oncol. Biol. Phys. 42(4), 909–912. Zhang, W. W., Fujiwara, T., Grimm, E. A., and Roth, J. A. (1995). Advances
Waxman, D. J., Chen, L., Hecht, J. E., and Jounaidi, Y. (1999). Cytochrome in cancer gene theraphy. Adv. Pharmacol. 12, 289–341.

Você também pode gostar