A05 001
A05 001
A05 001
Cancer Chemotherapy
Bernhard Kutscher, ASTA Medica AG, Frankfurt am Main, Federal Republic of Germany
Gregory A. Curt, National Cancer Institute, Bethesda, Maryland 20205, United States
Carmen J. Allegra, National Cancer Institute, Bethesda, Maryland 20205, United States
Robert L. Fine, National Cancer Institute, Bethesda, Maryland 20205, United States
Hamza Mujagic, National Cancer Institute, Bethesda, Maryland 20205, United States
Grace Chao Yeh, National Cancer Institute, Bethesda, Maryland 20205, United States
Bruce A. Chabner, National Cancer Institute, Bethesda, Maryland 20205, United States
port mechanism that is poorly understood but duration of binding to dihydrofolate reductase
appears to play a role in the transport of drug are directly related to the length of the polyglut-
when concentrations exceed 20 µM [21], [23]. amate tail [29].
Methotrexate and reduced folates do not com-
pete for uptake by this process, and it may rep- Interaction with Dihydrofolate Reductase.
resent a means for drug entry in cells resistant to The binding of methotrexate to dihydrofolate re-
low doses of drug by virtue of defective trans- ductase has been extensively investigated by X-
port by the high-affinity mechanism. In some ray crystallographic and amino acid sequencing
models, the sensitivity of a cell to methotrex- studies [30–34]. Methotrexate binds in a stoi-
ate can be directly correlated with efficiency of chiometric fashion to a hydrophobic pocket in
drug transport, i.e., sensitive cells have a greater the target enzyme DHFR [35]. The binding affin-
capacity for drug transport and longer intracel- ity depends on multiple factors, including pH,
lular retention of drug when compared to meth- salt concentration, and NADPH concentration,
otrexate-resistant cells [24]. Because decreased and has been reported to be ca. 10 pM [36]. In
membrane transport may play a role in clinical the cell, methotrexate is a reversible inhibitor ca-
drug resistance, a number of analogs with high pable of being displaced by high concentrations
lipid solubility have been developed that can cir- of substrate. Thus, free intracellular drug in ex-
cumvent a transport deficit. Methotrexate esters, cess of the cellular dihydrofolate reductase bind-
diaminopyrimidines, and triazenates have been ing capacity is required to maintain complete
synthesized and used with success against ex- inhibition of the enzyme and thereby produce
perimental cell lines with defective methotrex- and maintain a state of reduced-folate depletion.
ate transport. If an excess of intracellular drug is not main-
tained, the intracellular reduced-folate pool re-
Intracellular Metabolism. Once inside the covers through enzymatic reduction of oxidized
cell, naturally occurring folates may be metabo- folates and cellular metabolism resumes.
lized to polyglutamates; that is, additional glu- Cellular resistance to methotrexate has been
tamyl moieties are added to the terminal gluta- most commonly associated with an increase in
mate present on the parent compound. This pro- dihydrofolate reductase activity. In general, the
cess allows for selective intracellular retention of amplified enzyme is identical to the native pro-
the polyglutamated forms and an increased affin- tein in its affinity for methotrexate; however, al-
ity for certain folate-requiring enzymes, such as tered methotrexate affinity has been reported to
thymidylate synthase, and for enzymes required correlate with sensitivity to methotrexate in a se-
for the de novo production of purine nucleotides ries of murine leukemias [36]. Increased reduc-
[25]. Like the naturally occurring folates, in- tase activity and resistance to methotrexate has
tracellular methotrexate is also polyglutamated also been demonstrated in a number of cell lines
with the addition of from one to four additional made resistant in vitro by stepwise increases
glutamyl residues [26]. This process has been in drug concentration [37], [38], and in human
demonstrated in a variety of tissues, including tumor samples from clinically resistant tumors
human breast cancer cell lines [26], normal hu- [39]. The increased enzyme levels can be cor-
man liver [27], and murine leukemia cells [28]. related to gene amplification that may take the
The polyglutamates of methotrexate are selec- form of small new pieces of chromosomal ma-
tively retained by the cells and appear to have terial, called double minutes, or of large chro-
an enhanced inhibitory potential for certain en- mosomes, referred to as homogeneously stain-
zymes [18]. The inhibitory capacity of metho- ing regions (HSRs). The former variety of am-
trexate polyglutamates for dihydrofolate reduc- plification imparts relatively unstable resistance,
tase appears to be somewhat greater than that of which requires the ongoing selective pressure of
the parent drug. The selective retention of meth- drug presence to be maintained [40], whereas the
otrexate polyglutamates may be critical for the HSRs represent a more durable form of amplifi-
delayed cytotoxicity exhibited by cells capable cation and thus resistance. Several investigators
of polyglutamate synthesis. In vitro experiments [41–43] have successfully transvected amplified
using MCF-7 human breast cancer cells have reductase genes into normal hematopoietic cells,
demonstrated that the intracellular retention and allowing greater marrow resistance to metho-
Cancer Chemotherapy 5
hydrofolate reductase for activation, is effective 5-FdUMP. In the presence of methylene tetra-
in reductase-amplified lines [44]. hydrofolate, 5-FdUMP forms a stable ternary
complex with thymidylate synthetase (TS), in-
hibiting this critical enzyme to cause “thymine-
2.2. Fluoropyrimidines less death.” As expected, cytotoxicity is pre-
vented in the presence of exogenous thymi-
5-Fluorouracil (NSC-19893) [51-21-8], 5- dine in these cells with intact salvage path-
fluoro-2,4-(1H, 3H)-pyrimidinedione, 5-FU, ways. Inhibition of TS has long been consid-
C4 H3 FN2 O2 , M r 130.08, is a fluorinated ered the principal mechanism of 5-FU cytotoxi-
pyrimidine whose structural formula resembles city, but it has also been demonstrated that 5-FU
thymine; the hydrogen in the 5-position of the can be converted to 5-FUMP, either by orotic
naturally occurring pyrimidine being replaced acid phosphoribosyl-transferase (OPRTase) in
by fluorine. The synthesis of 5-FU in 1957 [15] the presence of phosphoribosyl pyrophosphate
represents the first successful effort in the ra- (PRPP), or by stepwise conversion to the ri-
tional design of anticancer drugs [53], and was bonucleotide 5-FUR (by uridine phosphorylase)
predicated on the earlier observation that malig- followed by formation of 5-FUMP by uridine
nant cells selectively utilized uracil (and possi- kinase. This intermediate can be converted to
bly toxic uracil analogs) in vivo [16], [54]. 5-FdUMP by ribonucleotide reductase to in-
hibit TS. Alternatively, 5-FUMP can be phos-
phorylated to 5-FUTP, which may be fraudu-
lently incorporated into RNA to induce cytotox-
icity. In a number of tumor models, loss of clono-
genic capacity is directly correlated with the ex-
tent of incorporation of 5-FUTP into RNA [55],
[56]. This RNA-specific toxicity is not reversed
by thymidine. However, the precise mechanism
of RNA-induced cell kill is speculative. The
5-Fluorouracil 5-FUDR
most consistent structural effect of 5-FU expo-
sure is impaired processing of ribosomal RNA
Since the original synthesis of 5-FU [57]. This concept is not supported by current ev-
and its nucleoside 5-FUDR (NSC-27640) idence, however, as neither the synthesis nor the
[50-91-9], 2 -deoxy-5-fluorouridine, floxuri- translation of messenger RNA (mRNA) appears
dine, C9 H11 FN2 O5 , M r 246.21, much has been affected by documented 5-FUTP incorporation
learned about the mechanism of action of the into mRNA. Using human colon carcinoma
fluoropyrimidines. These drugs are useful in the cells propagated in vitro, neither quantitative
treatment of a wide range of human malignan- nor qualitative differences in the translational
cies. products (polypeptides) of 5-FUTP-containing
mRNA could be demonstrated [58]. However,
small nuclear RNA species responsible for exon
2.2.1. Mechanism of Action recognition during RNA splicing do contain sig-
nificant quantities of uridylic acid [59]. Specific
Both 5-FU and 5-FUDR are prodrugs that re- substitution of 5-FUTP into this RNA fraction
quire intracellular metabolism to their respec- may be critical for 5-FU toxicity.
tive nucleotides for cytotoxicity. The pathways As shown in Figure 1, 5-FdUMP can be fur-
for fluoropyrimidine activation are shown in Fig- ther phosphorylated to 5-FdUTP, which can be
ure 1. Each drug is enzymatically activated by incorporated into tumor cell DNA [60], [61].
different routes to FdUMP, FUMP, or FUTP, and This mechanism of drug action has been par-
each of these fluorinated nucleosides has differ- ticularly difficult to appreciate, since the fraud-
ent mechanisms of cytotoxicity. ulent base is quickly excised from DNA by
Thymidine phosphorylase converts 5-FU to the enzymes uracil-DNA glycosylase and dUTP
the deoxyribonucleotide 5-FUDR, which is then nucleotidohydrolase. Thus, when tumor cells
phosphorylated by thymidine kinase to yield are incubated at low (0.1 µM) concentrations
Cancer Chemotherapy 7
of 5-FUDR, single-strand DNA shifts to lower elevated levels of target protein represent the
molecular mass species, suggesting excision of end result of specific gene amplification (as has
the fluorinated base [62]. However, the actual been documented for methotrexate resistance)
presence of 5-FdUTP in cellular DNA cannot remains to be determined.
be detected until tumor cells are exposed to
higher drug concentrations. Apparently, the im-
portance of 5-FU incorporation into DNA varies 2.2.3. Other Fluoropyrimidines
from tumor to tumor, with human promyelocytic Attempts to develop fluoropyrimidines with im-
leukemia cells incorporating nearly 100-fold proved therapeutic indexes have resulted in the
more 5-FdUTP into DNA than mouse leukemia synthesis and clinical trial of so-called masked
cells [63]. The importance of this pathway both fluoropyrimidines. The masked fluoropyrimi-
to tumor cell cytotoxicity and therapeutic index dine 5 -deoxy-5-fluorouridine is a nontoxic pro-
remains to be elucidated. drug that is converted to 5-FU by pyrimidine
nucleoside phosphorylase. Because this enzyme
may be present to a greater degree in some tu-
2.2.2. Mechanisms of Resistance mor cells than in normal human bone marrow,
an improved therapeutic index can be demon-
Tumor cells selected for in vitro resistance may strated in vitro using breast, sarcoma, leukemia,
demonstrate a deletion of critical drug-activating and colon carcinoma cells [75]. Since 5 -deoxy-
enzymes, including uridine kinase [64], orotic 5-fluorouridine requires conversion to 5-FU and
acid phosphoribosyltransferase [65], [66], and is in itself nontoxic, tumor cells that lack phos-
uridine phosphorylase [67]. Methylene tetra- phorylase activity are resistant to the masked
hydrofolate is required for 5-FdUMP inhibition compound, while remaining cross-sensitive to
of TS, and decreased availability of intracellu- 5-FU [76].
lar folates has been involved as a mechanism of Ftorafur is a second masked fluoropyrimidine
5-FU resistance [68]. that is less myelosuppressive than 5-FU. How-
In addition, alterations in the target enzyme ever, the drug has a higher incidence of gastroin-
TS can result in drug resistance. Resistant cells testinal and nervous system toxicity, which is
have been described with altered thymidylate probably due to organ-specific localization of
synthetase having decreased affinity for FdUMP activating enzymes [77]. In early clinical stud-
[69–71]. In addition, increased specific activity ies, ftorafur has shown antitumor activity in pa-
of TS has been reported in drug-resistant fibrob- tients with 5-FU refractory colorectal and breast
lasts [72] and tumor cells [73], [74]. Whether cancer [78].
8 Cancer Chemotherapy
heavily pretreated patients with acute myeloge- the triphosphate ara-CTP, which is responsible
nous leukemia [104]. Acute dose-limiting toxi- for inhibition of DNA synthesis.
cities associated with bolus drug administration
(severe gastrointestinal symptoms, fever, life-
threatening hypotension) may be ameliorated
by administering the drug via constant intra-
venous infusion [105]. However, 5-azacytidine
is chemically unstable, undergoing ring open-
ing at the 5,6-imino double bond to form N-
formylamidinoribofuranosylguanylurea, which
further decomposes to ribofuranosylurea [106].
The halflife of this decomposition is 4 h, com- Cytosine arabinoside
plicating the task of strict dosage control of pro-
The precise mechanism by which ara-CTP
longed infusions.
inhibits DNA synthesis remains uncertain. Ara-
To circumvent the problem of aque-
CTP does inhibit both DNA polymerase α [110]
ous instability, the hydrolytically sus-
and β [111]. The former is essential for DNA
ceptible 5,6-imino bond of 5-azacytidine
synthesis, the latter for DNA repair. Ara-CTP
was reduced to produce dihydro-5-
can be directly incorporated into DNA as well,
azacytidine (NSC-264880) [62402-31-7],
and this pathway correlates strongly with cy-
4-amino-5,6-dihydro-1-β-d-ribofuranosyl-
totoxicity. The extent of drug incorporation
1,3,5-triazine-2(1H)one · monohydrochloride,
into DNA is proportional to cell kill in both
C8 H14 N4 O5 · HCl, M r 282.7 [107]. This com-
acute myelocytic and promyelocytic leukemia
pound has completed phase I trials in the 1980s
cells [112]; incorporation and cytotoxicity can
with preliminary evidence of activity in lym-
be modulated by thymidine [113]. Also ara-C-
phoma; in addition it exhibits the unusual dose-
substituted DNA is unstable under conditions of
limiting toxicity of chest pain at the maximally
alkaline elution, suggesting drug-induced strand
tolerated dose [108].
fragility and breakage [114]. Ara-CTP incor-
poration also directly blocks strand elongation
[115] and causes premature strand termination
[116]. This results in accumulation of DNA
peaks of small sizes, suggesting that preexisting
DNA may be nicked following exposure to ara-C
[117]. A further effect of ara-C exposure is inhi-
bition of DNA repair, as determined by alkaline
elution studies in L 1210 cells [118]. As might
Dihydro-5-azacytidine be expected, cells pretreated with ara-C are more
sensitive to ionizing radiation, suggesting poten-
tiation of radiation damage by inhibition of DNA
repair [119], [120–123].
2.4. Cytosine Arabinoside (Ara-C)
Ara-C (NSC-63878) [147-94-4], 4-amino-1-β-
d-arabinofuranosyl-2(1H)-pyrimidone, cytara- 2.4.1. Mechanisms of Resistance
bine, cytosine arabinoside, C9 H13 N3 O5 , M r
243.22, an antimetabolite that is a structural ana- A number of mechanisms of resistance to ara-C
log of cytidine, differs from the physiologic nu- have been described. Ara-C itself is a prodrug
cleoside by the epimeric configuration of the β- that is metabolized first to ara-CMP by deoxy-
trans-hydroxyl group at the 2 position of the cytidine kinase, then to ara-CDP by pyrimidine
sugar. The drug is transported into cells by a nucleoside monophosphate kinase, and finally
carrier-mediated process with shared affinity for to ara-CTP by nucleoside diphosphate kinase.
deoxycytidine [109]. Once ara-C has entered the Resistant cell lines lack the initial, rate-limiting
cell, cytotoxicity is dependent on formation of activating enzyme [115], [124].
10 Cancer Chemotherapy
Since the 1980s, HPLC has been used to high levels of cytidine deaminase because the
separate and semipurify deoxycytidine kinase, catabolism product, FIA-uracil, is more toxic
pyrimidine nucleoside monophosphate kinase, than the parent compound [138].
and pyrimidine nucleoside diphosphate kinase
from cell extracts [125]. In murine leukemia
cells selected for ara-C resistance, deoxycyti- 2.5. Deoxycytidine and Analogs
dine kinase was found in lower specific activity,
and similar results have been reported for other The pyrimidine antimetabolite gemcitabine (LY-
systems [126]. 188011, dFdC) [095058-81-4], 2’-deoxy-2’,2’-
In addition, expansion of the intracellular difluorocytidine, C9 H11 F2 N3 O4 is an analog of
pool of the physiologic substrate deoxycytosine deoxycytidine and a result of a program initi-
triphosphate can inhibit ara-C activation by feed- ated at Lilly Research to synthesize fluorinated
back inhibition of the initial activating enzyme d-ribose and fluorinated nucleosides [139–142].
[127]. Moreover, increased dCTP pools may The difluorinated analog of cytarabine, gemc-
also compete directly with ara-CTP for DNA itabine was identified as novel antimetabolite
polymerase. Experiments have shown a relation- with a broad spectrum of antitumor activity.
ship between duration of leukemia remission Trade names: Gemicitabine monohydro-
and the ability of tumor cells obtained from pa- chloride [122111-03-9] is on the market as
tients to form and retain ara-CTP in vitro [128], Gemzar in the indication of palliative treatment
but the mechanisms underlying this relationship of locally advanced or metastatic non-small cell
remain to be established. lung cancer.
Although some workers have reported that in- Mechanism of Action. Gemcitabine shows
creased drug catabolism by cytidine deaminase good activity against human leukemic cell lines,
may underlie both de novo and acquired drug re- a number of murine solid tumors, and human
sistance in patients with leukemia [129], a def- tumor xenografts [143–146]. Gemcitabine was
inite clinical correlation between response and significantly more cytotoxic than cytarabine in
levels of this enzyme has not been established in Chinese hamster ovary cells. The major cellular
several studies [130–132]. metabolite is the 5’-triphosphate of gemcitabine.
The cytotoxicity was competitively reversed by
deoxycytidine, suggesting that the biological
2.4.2. New Analogs activity required phosphorylation by deoxycyti-
dine kinase [145].
Several ara-C analogs have been rationally de- Tumor-bearing mice were treated with ei-
signed with the goal of overcoming specific ther gemcitabine or cytarabine (20 mg/kg). DNA
mechanisms of resistance. Compounds with synthesis reached 1 % of control levels upon
lipophilic side chains are relatively resistant administration of gemcitabine. The greater
to inactivation by cytidine deaminase. Another accumulation of gemcitabine-5’-triphosphate
analog with lipophilic modifications in the side compared with cytarabine-5’-triphosphate may
chain is enocitabine (NSC-239336, BHAC) cause greater cytotoxicity and therapeutic activ-
[055726-47-1] [133], [134]. N 4 -Behenoyl-ara- ity [146].
C (BHAC) has undergone clinical trial in pa-
tients with acute leukemia [135]. Despite its
lipophilicity, the drug does not enter the cere-
brospinal fluid and concentrates in bone mar-
row and red blood cells. N 4 -Palmitoyl-ara-
C can be administered orally and appears to
be more active than the parent compound in
preclinical models [136]. Although developed
as an antiviral agent, 2 -fluoro-5-iodo-1-β-d-
Gemcitabine
arabinofuranosylcytosine (FIAC) has significant
antitumor activity [137]. Interestingly, this com- Further gemcitabine may enhance its own cy-
pound may be relatively cytotoxic for cells with totoxic effects by self-potentiation mechanisms
Cancer Chemotherapy 11
that act on, e. g., deoxycytidine monophosphate behaves more like an analog of deoxycytidine
deaminase, deoxycytidine kinase or on DNA- than adenine or deoxyadenine as indicated by
synthesis [147], [148]. reports [155] demonstrating that the presence of
fluorine in the 2-position of the adenine ring al-
ters its function as a substrate for deaminase and
2.6. 2-Halopurines and Analogs nucleoside kinases. This results in differences
in biological activity and metabolism. Halo-
Cladribine (2-CdA, RWJ-26251) 004291-
genation does not simply block deamination,
63-8, is a purine deoxyribonucleoside with re-
but also influences the enzyme that carries out
markable antileukemic activity. It represents a
the phosphorylation, as a result cytotoxicity is
significant advance over existing therapy be-
increased [156]. Fludarabine phosphate may se-
cause it is given as a single 7-day continuous
lectively inhibit the incorporation of thymidine
treatment, thus minimizing the side effects ob-
and uridine into the DNA molecule by inhibiting
served with multiple treatments. Remission rates
both ribonucleotide reductase [157] and DNA
of up to 89 % lasting for up to 25 months were ob-
polymerase [158]. The maximum tolerated dose
served in clinical trials in patients with hairy cell
(MTD) in heavily pretreated patients with ad-
leukemia [149–152]. The antimetabolite used
vanced malignancy/solid tumors on the daily
for first line treatment of hairy cell leukemia
regimen was about 15 mg/m2 . Granulocytope-
[153].
nia and thrombocytopenia were dose-limiting
Trade name: Leustatin.
[159–161].
Fludarabine phosphate (NSC-312887, 2-F-
ara-AMP) [075607-67-9] [154], 2-fluoro-9-
(5-O-phosphono-β-d-arabino-furanosyl)-9H- Pentostatin. The cytotoxic and im-
purin-6-amine, C10 H13 FN5 O7 P, M r 365.21, munosuppressant pentostatin (NSC-218321,
another cytotoxic purine antimetabolite, acts CI-825, PD-81565, YK-176, 2-deoxyco-
via inhibition of DNA synthesis. The product formycin, 2’-dCF) [063677-95-2], (R)-3-(2-
is used for treatment of patients with chronic deoxy-β-d-erythro-pentofuranosyl)-3,6,7,8-
lymphocytic leukemia. tetrahydroimidazo[4,5-d][1,3]diazepin-8-ol,
Trade names: Fludara (Berlex), Benefluor C11 H16 H4 O4 , M r 268.13, mp 220 – 225 ◦ C, can
(Schering AG). be isolated from the fermentation broth of Strep-
tomyces antibioticus NRRL 3238 [162], [163]
or Aspergillus nidulanus Y 176-2 or Emericella
[164]. The adenosine nucleoside analog pento-
statin, is the most potent inhibitor of adenosine
deaminase, which is an important and ubiquitous
cellular enzyme. The inhibition of this enzyme
leads to accumulation of dATP which inhibits
ribonucleotide reductase and thus DNA synthe-
sis. Pentostatin was launched for treatment of
Cladribine
hairy-cell leukemia refractory to α-interferon.
Trade names: Nipent (Parke-Davis, Lederle),
Coforin (Katetsuken).
Fludarabine
ply of purine precursors for nucleic acid synthe- treated with allopurinol. Furthermore, allopuri-
sis [181]. In the 1980s two new findings pertain- nol increased the plasma level of 6-MP in rab-
ing to the cytotoxicity mechanism in tumor cells bits [207]. The data suggest that inhibition of 6-
have been reported. Studies of human lymphoma MP catabolism by allopurinol may contribute to
revealed that 6-MP was a potent inhibitor of cel- a greater availability of 6-MP to tissues. Stud-
lular RNA synthesis and that 6-thioITP inhib- ies of the effect of allopurinol on the kinetics
ited both the RNA polymerase I and RNA poly- of oral and intravenous 6-MP in Rhesus mon-
merase II activities of these cells [197]. These keys and in humans demonstrated that allopuri-
data suggested that direct inhibition of the en- nol pretreatment resulted in a nearly 400 % in-
zymes mediating transcription by 6-thio-IMP crease in peak plasma concentration of 6-MP
may be one of the mechanisms for the cytotoxic in monkeys and a 500 % increase in humans,
action of 6-MP in human tumor cells. Using 6- but only when 6-MP was administered orally
TG as a cytotoxic agent resulted in severe chro- [208]. Allopurinol pretreatment had no effect on
mosome damage in wild-type CHO cells [198]. the kinetics of intravenously administered 6-MP.
Gross unilateral chromatid damage resulted, and This difference is due to the action of allopurinol
the unilateral nature of this damage was proba- on liver or intestinal xanthine oxidase and in-
bly due to malfunction of 6-TG-containing DNA hibition of first-pass metabolism of oral 6-MP.
as a replication template. This finding may explain the low and variable
plasma levels of mercaptopurine in patients with
acute lymphoblastic leukemia treated with oral
2.7.2. Mechanism of Resistance 6-MP [209]. Although these studies emphasize
the catabolic pathway for purines, all the purine
Several mechanisms of resistance to these agents and pyrimidine metabolic enzymes may be im-
have been described in experimental tumors and portant to the bioavailability and activation of
relate to the pathways of antimetabolite activa- antimetabolites.
tion and degradation. A decreased HGPRT ac- To clarify the mechanisms of resistance
tivity in tumor cells diminishes antimetabolite to thiopurines and potential drug interactions
activation, and this resistance pathway has been in tumor cells, studies have focused on the
reported by several workers [199–202]. How- thiopurine-resistant cell lines deficient in HG-
ever, the HGPRT-regulated mechanism as a ba- PRT (L 1210) and the regulation of PRPP for-
sis for drug resistance in human leukemic cells mation in thiopurine-resistant cell lines.
is relatively uncommon [203]. The resistance of The thiopurines are inactive in the base form
6-MP is related to an increase in alkaline phos- and must be converted to their respective nu-
phatase in sarcoma cells [204]. Increased alka- cleotides. This activation step requires PRPP as
line phosphatase, a membrane-bound enzyme the cofactor and HGPRT as the enzyme to cat-
that converts the active mononucleotide to 6- alyze the conversion to nucleotide. A major bio-
thioinosine and inorganic phosphate, has also chemical effect of methotrexate is the suppres-
been reported in human leukemia patients re- sion of purine biosynthesis and expansion of the
sistant to drug treatment [205], [206]. Another PRPP pool. Studies of the cytotoxic and bio-
enzyme in the degradation pathway of 6-MP that chemical interaction of methotrexate and 6-TG
needs to be considered for drug efficacy and in L 1210 mouse leukemia cells demonstrated
bioavailability is xanthine oxidase, which is re- that methotrexate can markedly enhance 6-TG
sponsible for converting 6-MP to 8-OH-6-MP activity [210]. Preexposure of cells to metho-
and subsequently to thiouric acid, which is ex- trexate resulted in a large increase in cytotoxic
creted through the urine. potency of 6-TG, whereas simultaneous expo-
To understand further the bioavailability and sure caused an antagonism of 6-TG cytotoxic ac-
pharmacokinetics of thiopurines, the effect of al- tivity. Although PRPP pools were not measured
lopurinol on 6-MP catabolism was studied. Al- quantitatively, the effect of methotrexate preex-
lopurinol, an analog of hypoxanthine, enhances posure was to increase PRPP pools, enhance the
the therapeutic efficacy of 6-MP by inhibiting activation of 6-TG to 6-TG monophosphate, and
xanthine oxidase. The urinary excretion of 6- thereby increase its incorporation into RNA.
MP metabolites is markedly reduced in patients
14 Cancer Chemotherapy
ween DNA in the double-helix structure pref- 305.20, was rationally designed and synthe-
erentially at the N-7 guanosine site. Thus, these sized as a phenylalanine derivative of nitrogen
bifunctional compounds are more effective anti- mustard with the aim of obtaining increased
tumor agents than their monofunctional analogs; specificity against melanoma tumor cells that
however, increasing the number of alkylating utilize phenylalanine or tyrosine to produce
sites on the agent beyond two does not appear melanin.
to increase antitumor activity [222]. This evi-
dence suggests that DNA cross-linking is crit-
ical for alkylator activity. Further evidence for
the importance of cross – linking to cytotoxic-
ity comes from alkaline elution studies that can
detect low levels of cross-linking in cells ex- Melphalan
posed to minimal doses of bifunctional alkylat-
ing agents [223]. Although this compound does not exhibit spe-
In contrast, monofunctional agents, such as cific antimelanoma activity, it has a broad-
procarbazine and dacarbazine, do not produce spectrum cytotoxicity in multiple myeloma,
DNA interstrand cross-links and appear to ex- breast cancer, and lymphomas.
ert toxicity by producing single-strand DNA
breaks. The increased carcinogenicity seen with
some monofunctional alkylating agents may be 3.2.1. Mechanism of Action
due to incorrect base pair substitution by DNA Melphalan is a bifunctional alkylating agent but
repair enzymes that could result in malignant differs from HN2 mainly by the presence of
transformation. an aromatic ring. This ring reduces the nucle-
ophilicity of the nitrogen atom by withdrawing
electrons, making the drug less reactive. Thus, it
3.1.2. Mechanisms of Drug Resistance can be taken orally. It retains its alkylating activ-
ity but is more selective than nitrogen mustard
Several mechanisms have been elucidated for ni- because it is less likely to form the unstable and
trogen mustard and other bifunctional alkylating highly reactive aziridine intermediate indiscrim-
agents. First, resistant cells with defective drug inately. Like nitrogen mustard, it forms DNA
transport have been described. Nitrogen mustard cross-links that are critical for cytotoxic effect.
enters cells by an active transport system that Its cellular uptake is also mediated by an active,
is physiologically utilized for choline transport. energy-dependent transport mechanism shared
Lymphoma cells resistant to nitrogen mustard with leucine and glutamine uptake [218]. Thus,
demonstrate decreased drug uptake by this spe- high concentrations of leucine or glutamine can
cific active transport site, which also decreases reduce the cytotoxicity of melphalan in a mar-
its uptake of choline [224]. Second, cytosolic in- row colony-forming unit assay [229]. Another
crease in nonprotein sulfhydryl levels [225] and transport mechanism, although less active, can
higher nonprotein-bound thiol compounds that be used by melphalan, and this mechanism is
could inactivate the drugs before they reach the shared by the neutral amino acids alanine, cys-
nucleus have been found in nitrogen mustard- teine, and serine [218].
resistant cells [226]. Third, enhanced repair of
DNA cross-linking by repair enzymes has been
demonstrated in vitro [227] and in vivo [228]. 3.2.2. Mechanism of Resistance
Decreased transport of melphalan into drug-
3.2. Melphalan resistant leukemia cells has been demonstrated
and correlated to the melphalan-resistant pheno-
Melphalan (NSC-8806) [148-82-3], 4- type. Specifically, a mutation in the higher ve-
[bis(2-chloroethyl)amino]-l-phenylalanine, l- locity transport system has been suggested that
phenylalanine mustard, C13 H18 Cl2 N2 O2 , M r results in a decreased affinity of the carrier pro-
tein for melphalan and leucine [230]. Evidence
16 Cancer Chemotherapy
for the other two mechanisms of resistance seen 3.3.2. Mechanism of Resistance
with nitrogen mustard, i.e., increased intracellu-
lar thiol compounds and increased DNA cross- Similar mechanisms of resistance occur for cy-
linking repair also, exists. clophosphamide as for nitrogen mustard. Also,
defective metabolic conversion by the hepatic
microsomal system could serve to decrease the
3.3. Cyclophosphamide bioavailability of aldophosphamide to tumor tis-
sue, but it is not known whether this is signifi-
Cyclophosphamide (NSC-26271) [50-18-0], cant.
N,N-bis(2-chloroethyl)tetrahydro-2H-1,3,2-
oxazaphosphorin-2-amine 2-oxide,
C7 H15 Cl2 N2 O2 P, M r 261.10. 3.4. Chlorambucil
Thio-TEPA
3.8. Nitrosoureas
The mechanisms of action and resistance are
similar to those of nitrogen mustard. The clinically useful nitrosoureas in-
clude carmustine (NSC-409962) [154-93-8],
N,N -bis(2-chloroethyl)-N-nitrosourea,
3.6. Ifosfamide BCNU,C5 H9 Cl2 N3 O2 , M r 214.04; lo-
mustine (NSC-79037) [13010-47-4], N-
Ifosfamide, or isofosphamide (NSC-109724) (2-chloroethyl)-N -cyclohexyl-N-nitroso-
[3778-73-2], N,3-bis(2-chloroethyl)-tetra- urea, CCNU, C9 H16 ClN3 O2 , M r 233.69;
hydro-2H-1,3,2-oxazaphosphorin-2-amine 2- methylcyclohexylchloroethylnitrosourea (NSC-
oxide, C7 H15 Cl2 N2 O2 P, M r 261.07, is an ana- 94941) [52662-76-7], 1-(2-chloroethyl)-3-
log of cyclophosphamide [231]. It has approx- (4-methylcyclohexyl)-1-nitrosourea, methyl-
imately one-third the alkylating activity of cy- CCNU, C10 H18 ClN3 O2 , M r 248; strep-
clophosphamide and requires hepatic microso- tozotocin (NSC-85998) [18883-66-4], 2-
mal conversion to its active form. Also, the rates deoxy-2{[(methylnitrosoamino)carbonyl]ami-
of conversion by metabolism are similar in both no}-d-glucopyranose, C8 H15 N3 O7 , Mr
drugs, but less biologically active alkylating 265.22; and chlorozotocin (NSC-178248)
moieties are formed in ifosfamide [232]. It has [54749-90-5], 2-({[(2-chloroethyl)nitrosoami-
shown promising results in refractory pediatric no]carbonyl}amino)-2-deoxy-d-glucose,
bone and soft tissue sarcomas, refractory tes- DCNU, C9 H16 ClN3 O7 , M r 313.69. Streptozo-
ticular tumors, and Wilms’ tumor in children tocin is a naturally occurring nitrosourea derived
[233], [235]. from Streptomyces acromogenes.
Trade names: Ifex, Holoxan (ASTA Medica)
Ifosfamide
[238]. Monofunctional CCNU and methyl- to inactivation of the DNA template, may well be
CCNU cross-link by initial carbocation for- the key mechanism of cytotoxicity [252–254].
mation and alkylation followed by loss of the Monofunctional DNA alkylations also occur fol-
chloride substituent and formation of a second lowing nitrosourea exposure and these must also
reactive carbocation. These alkylation reactions be considered cytotoxic as monofunctional alky-
appear to be the major mode of cytotoxicity for lating agents, incapable of cross-linking, retain
these agents [239], [240]. With the exception of cytotoxic activity. Monofunctional alkylations
chlorozotocin, the nitrosoureas may also form may produce single-strand DNA breaks by en-
an isocyanate compound that may play a role donuclease cleavage at apurinic sites produced
in the toxic side effects of these agents [241], by the alkylation and repair process [255].
[242], but has little importance in the antitumor
effect. In support of this view is the fact that
chlorozotocin retains its potent cytotoxic capac- 3.8.2. Mechanisms of Resistance
ity while producing little or no isocyanate com-
pound and reportedly has less marrow toxicity. The mechanisms of cellular resistance to nitro-
Experimental evidence raises additional ques- soureas remain unclear, although defective drug
tions concerning the role of isocyanates, which transport, as has been demonstrated for the clas-
may enhance the antitumor activity of these sic alkylating agents, would be unlikely given
compounds. Methylnitrosourea, which cannot the lack of need for an active membrane trans-
alkylate DNA to produce cross-linking, pro- port system. In human glioblastoma cells, in-
duces alterations of nuclear protein in a manner creased activity of a specific excision enzyme,
similar to BCNU, and isocyanates are consid- guanine-O6 -alkyltransferase, is correlated with
ered a possible explanation for these effects from in vitro resistance to nitrosoureas [256], and re-
both agents [243]. Also, sensitivity of a Walker pair mechanisms would seem a likely mecha-
tumor line made resistant to bifunctional alky- nism since mammalian cells are capable gener-
lating agents can be restored by simultaneous ally of such repair [257].
treatment with an isocyanate-producing agent
[244].
3.8.3. Analogs
3.8.1. Mechanism of Action New agents have been synthesized that con-
tain a sugar moiety similar to streptozotocin.
Because of their lipophilicity, nitrosoureas enter Analogs that contain mannose, glucose, ribose,
cells by passive diffusion as opposed to an ac- maltose, and galactose have all been produced,
tive transport mechanism common to the clas- with the maltosyl derivative being exceptionally
sic alkylating agents [245]. Once inside the cell, active in a variety of tumors tested [258], [259].
alkylating agent exposure results in pancellu- Sugar alcohols, such as mannitol, linked to nitro-
lar covalent binding of drug to proteins, nu- soureas retain their cytotoxicity but appear to
cleic acids, and to a variety of smaller intra- protect against the marrow toxicity induced by
cellular molecules. Which of these reactions is the parent nitrosourea compounds. A number of
critical for cytotoxicity remains uncertain, but derivatives with di- and tripeptides containing
the majority of evidence points to interaction di- alkyl nitrosoureas have also been introduced but
rectly with DNA as the focal point of cytotoxic- have met with only moderate preclinical success
ity [246–248]. The 7 position of guanine is par- [260].
ticularly susceptible to alkylation, and accounts There is continuous effort to obtain new nitro-
for the majority of the total alkylation of DNA soureas with higher efficiency and/or lower tox-
[249], [250]. Since the chloroethyl nitrosoureas icity and several new compounds are undergo-
are each capable of two independent alkylations ing clinical trials. In 1987 ranimustine (NSC-
[238], DNA can be cross-linked by either inter- 270561, MCNU) [058994-96-0], was marketed
strand or intrastrand processes [251]. Multiple as Cymerin (Tokyo Tanabe) [261].
studies using a variety of alkylating agents have The phosphonoalanine derivative
confirmed that DNA cross-linking, which leads fotemustine (S-10036) [092118-27-9],
Cancer Chemotherapy 19
Mitomycin
4. Anthracyclines (→ Antibiotics,
Chap. 3.2.3.) Epirubicin
Idarubicin
amycin and general toxicity lower than that of has been established that the anthracyclines can
other analogs [302, 303, 303]. The main indica- affect every DNA function, including initiation,
tions for pirarubicin are cancer of the bladder, chain elongation, DNA synthesis, DNA repair,
head and neck, and cervix. and RNA synthesis [308], [309]. Experimentally
these compounds can cause sister chromatid ex-
changes, single- and double-strand breaks, and
alkylation of DNA [310], [311]. Interestingly,
some results strongly suggest that inhibition of
DNA synthesis is not essential for cell kill. New
anthracycline analogs, such as aclacinomycin
A, which selectively inhibits preribosomal RNA
synthesis and not DNA synthesis, retain cytotox-
icity. This suggests other non-DNA-mediated
mechanisms of cytotoxicity, such as preriboso-
mal RNA synthesis [309].
Pirarubicin
Free-Radical Generation. Free-radical for-
The anthracyclines are derived from Strep- mation (highly reactive compounds with an un-
tomyces species and are structurally tetracyclic paired electron) occurs during the metabolism
chromophore antibiotics [305]. They are classi- of anthracyclines. When the microsomal en-
fied by their chromophore, otherwise known as zyme P 450 reductase or xanthine oxidase in-
aglycone, structure. The sugar most commonly teracts with and reduces the ketone oxygen in
attached to the aglycones is daunosamine, but ring B to O− , a semiquinone radical interme-
other sugars may be involved, and these are men- diate is formed. This interacts with oxygen to
tioned in Section 4.3. The basic tetracyclic agly- produce the superoxide radical with regenera-
cone structure of the anthracyclines shares many tion of the original anthracycline structure [312].
characteristics with the hydroxyanthraquinones, The superoxide radical can serve as a substrate
which are ubiquitous in nature. for superoxide dismutase to form hydrogen per-
oxide, which can interact with the superoxide
molecules to form hydroxyl radicals [313]. The
4.1. Mechanism of Action superoxide and hydroxyl radicals can interact
with and damage cells, especially the hydroxyl
DNA−RNA Binding. The exact mecha- radical, which is one of the most reactive sub-
nism of cytotoxicity by anthracyclines is un- stances known. Hydroxyl radicals can also re-
known, but they do have multiple and distinct act with purine or pyrimidine bases, amines, and
toxic effects that may kill a tumor cell in one or thiols. This free-radical formation is responsible
more ways [306]. for cardiac damage seen with chronic use of the
Initially these drugs were found to bind DNA anthracyclines in the treatment of human malig-
by intercalation between base pairs perpendicu- nancies. Evidence exists in many animal mod-
lar to the long axis of the double helix, with the els, as well as in humans, that superoxide and hy-
major binding occurring between the B and C droxyl radical formation occurs in cardiac tissue,
rings of the drugs with the bases above and be- leading to lipid peroxidation of mitochondria
low [307]. The daunosamine sugar is thought to and sarcosomes [314], [315]. Since mitochon-
bind ionically with the sugar-phosphate back- dria account for more than 40 % of cardiac mus-
bone of DNA. The binding association con- cle mass, as well as being the major source for
stant is between 105 and 106 M−1 . Intercalation ATP needed for contraction, and are coupled to
of the DNA causes a partial unwinding of the calcium release during the action potential, one
helix and thus disrupts DNA polymerases and can easily visualize how these agents mediate
transcription. However, these experiments were cardiac toxicity. However, it now seems apparent
done with DNA in vitro; DNA in vivo is orga- that cardiac tissue lacks catalase and that dox-
nized into chromatin, which is DNA wrapped orubicin destroys glutathione peroxidase [315],
around a series of histone core particles. Also, it [316]. Agents that can scavenge free-radicals are
Cancer Chemotherapy 23
that if the 4 -hydroxyl group is removed, car- compared to doxorubicin on an equimolar ba-
diac toxicity is lessened significantly [322–324]. sis mitoxantrone proved to be six to seven times
Thus, it seems that the amino sugar and/or the 4 - more potent in inhibiting the incorporation of
hydroxyl group are major determinants of car- uridine and thymidine into DNA [330]. Mitox-
diac toxicity in the anthracyclines. antrone is a cell phase nonspecific agent and
has wide spectrum of activity against several
experimental animal tumors. Cross-resistance
5. Intercalating Anthracenes and to adriamycin was not always seen [331–333].
Adriamycin-like cardiac toxicity was not found
Analogs in comparative studies using rats, dogs, rabbits,
and monkeys maybe partially due to inhibition
5.1. Mitoxantrone of free radical formation and due to the lack of
the amino sugar moiety [334], [335]. Although
Mitoxantrone hydrochloride (NSC-301739, mitoxantrone is not entirely free of cardiac tox-
DAD, CL-232315) [070476-82-3], 1,4- icity in humans, minimal nausea and vomiting
dihydroxy-5,8-bis[(2-(2-hydroxyethyl)ami- was observed [336–341].
no)ethyl]amino-9,10-anthracenedione, is a new
type of antineoplastic agent. Mitoxantrone is
active against breast cancer, acute leukemia, 5.2. Analogs
lymphoma, cervix carcinoma, and liver cell
cancer. Unlike the anthracyclines that have a Bisantrene hydrochloride (NSC-337766,
red color, the anthracenediones are deep blue. ADD, CL-216942) [071439-68-4], 9,10-
Mitoxantrone is structurally similar to adri- anthracenedicarboxaldehyde bis[(4,5-di-
amycin but without the aminosugar at C9 and hydro-1H-imidazol-2-yl] dihydrochloride,
can be synthesized starting from 1,8-dihydroxy- C22 H22 N8 ’2 HCl, M r 471.39, is an intercalating
anthrachinone [325–329]. anthracenebishydrazine cytostatic [342–344].
Trade names: Novanthrone (Lederle / Amer- The product was launched for treatment of acute
ican Cyanamide); Onkotrone (ASTA Medica non-lymphocytic leukemia [345].
AWD). Trade names: Zantrene and Cyabin (Led-
erle).
As solubility is a problem, bisantrene pro-
drugs with enhanced water solubility (e.g.,
199344, see below) were developed [346–348].
Mitoxantrone
Further intercalating agents such as am- 1940s [357]. Structurally, compounds in this
sacrine (NSC-156303/NSC-249992, m-AMSA, class share a common phenoxone ring and two
SN-11841) [051264-14-3] [349], [350], or ni- cyclic pentapeptides. The natural products dif-
tracin [4533-39-5] [351] have the acridine struc- fer among themselves in the amino acid com-
ture in common. position of the peptide chains, but only actino-
Trade names: Amsacrine is marketed as Am- mycin D is used in the clinical treatment of can-
sidine, Amecrin, and Ansidyl (Parke-Davis) cer, where it demonstrates reproducible activity
[352–354]. against gestational choriocarcinoma and Wilms’
tumor.
Actinomycin D (NSC-3053) [50-76-0],
dactinomycin, C62 H86 N12 O16 , M r 1255.47.
Actinomycin D
26 Cancer Chemotherapy
Adozelesin
CC-1065
Bizelesin
Carzelesin
Pyrindamycin A
Duocarmycin C1
[110314-48-2] [378] is the clinically farthest A series of new DNA cleaving molecules
advanced agent, while the prodrug carzelesin based on the reactive enediyne moiety
[119813-10-4] [379] and the dimer bizelesin are the anticancer antibiotics calicheam-
[129655-21-6] [380] demonstrated impressive icin [113440-58-7] [385], esperamicin
preclinical activity. [114797-28-3] [386], and dynemicin.
Further modified cyclopropylpyrroloindoles Simplified enediyne-type compounds dam-
(CPI) forming covalent adducts with DNA are age DNA upon activation by chemical or biolog-
duocarmycin A [118292-35-6] [381], [382], ical means and are extremely potent cytotoxic
pyrindamycin [118292-36-7] and FCE 24517 agents in vitro [387], [388].
[383], [384].
28 Cancer Chemotherapy
Esparamicin A1
Cellular Pharmacology. As yet it is not nism of this pleiotropic drug resistance remains
clear how vinca alkaloids cross cell membranes. to be clarified. However, tubulin is not respon-
Some data suggest an energy-dependent trans- sible for that phenomenon because there is very
port system [389], while other data suggest sim- little difference in affinity binding of colchicine
ple diffusion across membranes [390]. However, to tubulin isolated from drug-sensitive and drug-
passive diffusion is important only at drug con- resistant cells [397]. The appearance of a novel
centrations exceeding 100 µM. Transport of vin- glycoprotein on the membrane of resistant cells
cristine is completely inhibited by vinblastine, and the accelerated drug efflux – leading to im-
suggesting a common carrier. paired drug accumulation – seem, at present,
Vincristine and vinblastine exert their bio- to be the possible mechanisms responsible for
logic effect through binding to tubulin. This oc- resistance [398].
curs in interphase (late S and G2 ), producing
a transient G2 block [391]. They do not affect
DNA synthesis directly. The metabolic conse- 7.1.2. Vindesine
quences of tubulin binding include polyploidy,
nuclear fragmentation, and inhibition of cytoki- Vindesine (NSC-245467) [53643-48-4], 3-
nesis, which occurs after prolonged drug expo- (aminocarbonyl)-O4 -deacetyl-3-de(methoxy-
sure. Present studies suggest that sensitivity to carbonyl)vincaleukoblastine, deacetylvin-
these drugs increases progressively as cells ap- blastineamide, C43 H55 N5 O7 , M r 753.95 (struc-
proach mitosis and that cells at the end of the ture, Section 7.1.1), is a vinblastine metabolite.
cycle at the time of exposure are likely to ex- It possesses antitumor activity that is similar
hibit the greatest degree of mitotic disorganiza- to vincristine’s rather than that of its parent
tion [391]. compound, vinblastine. Vindesine demonstrates
Vincristine and vinblastine share a common better activity than vinblastine in some tumor
binding site on each tubulin monomer, with the models (Gardner lymphosarcoma, Ridgway os-
binding affinity (K d ) of about 1.6 × 10−6 M. teogenic sarcoma, and mammary carcinoma).
The drug concentrations necessary to produce In the past several years vindesine has attracted
50 % cell kill in vitro are ca. 4 × 10−8 M [392]. considerable attention among clinical investi-
Malignant lymphocytes appear more susceptible gators and has been intensively investigated.
than normal lymphocytes, presumably because Response rates vary, with the highest responses
of high tubulin content expressed on the surfaces observed in the highest lymphatic malignancies
of leukemic cells [393]. Other metabolic effects [399]. The drug is less neurotoxic than vin-
include inhibition of DNA and RNA and pro- cristine and appears to be active in vincristine-
tein synthesis. However, these effects are exerted resistant tumors [400], [401].
only at very high drug concentrations (1000
times greater than those achieved in vivo) and
probably are secondary phenomena. 7.1.3. Vinorelbine
metastatic ovarian cancer where standard ther- Mechanism of Action. More than two
apy has failed, paclitaxel has been marketed decades after its isolation and the elucidation
since 1993 as Taxol (Bristol-Myers Squibb) of its complex structure and cytotoxic activity
and was the best-selling anticancer agent in [433] interest in paclitaxel raised again when
1996. Taxol does not share a common binding S. Horwitz et al. [446] reported on the novel
site with other antitubulin agents and may in- mechanism of action. As spindle poison pacli-
stead bind to and stabilize polymerized tubulin taxel promotes the polymerization of tubulin
[442]. to microtubules and stabilizes them against de-
A semisynthetic analog of pacli- polymerization, whereas vinca alkaloids induce
taxel, docetaxel (NSC-628503, RP- microtubule disassembly (see Fig. 2). Thus, with
56976) [114977-28-5], (2R,3S)-N-carboxy- paclitaxel the dynamic equilibrium of assem-
3-phenylisoserine, N-tert-butyl ester, 13- bly and disassembly of microtubules is shifted
ester with 5β, 20-epoxy-1,2α-4,7β,10β,13α- in favor of the polymer, preventing cell divi-
hexahydroxytax-11-en-9-one 4-acetate 2- sion [447]. Paclitaxel binds preferentially to the
benzoate, C43 H53 NO14 , M r 807.9, is a β-tubulin subunit. This binding is reversible
derivative with an N-tert-butyloxycarbonyl- and the site is different from the binding sites
(2R,3S)phenylisoserine C-13 side chain. Do- of vinca alkaloids, colchicine and podophyllo-
cetaxel is more potent than paclitaxel in vitro toxin [448–450]. If paclitaxel is present, tubu-
[443], [444]. Impressive clinical results have lin polymerizes without exogenous GTP and
been reported for the treatment of ovarian, these stabilized, rigid microtubules cannot be
breast, and bronchial cancers with docetaxel disassembled. As a result, the dynamic orga-
[445]. The compound was launched in 1996 for nization of the cell is interrupted which leads
the treatment of locally advanced breast can- to irreversible damage in rapidly dividing cells.
cer or relapse during anthracycline therapy of Further evidence has been reported that the an-
NSCLC and breast cancer. tiproliferative activity of paclitaxel is caused by
Trade name: Taxotere (Rhône-Poulenc additional effects [451–453].
Rorer). A major impediment in the development of
taxol as a drug was its poor water solubil-
ity [454]. With the solubility enhancer Cre-
mophor EL hypersensitivity reactions, including
hypotension, urticaria, and dyspnea, occurred in
patients during rapid infusion. To cope these al-
lergic reactions, 24-hour infusions and pretreat-
ment with dexamethasone, diphenhydramine, or
cimetidine was recommended [455].
The dose-limiting toxicity of paclitaxel is
neutropenia. Several other toxic effects such as
diarrhea, nausea, and emesis are less common.
Paclitaxel
Docetaxel shares many toxic effects with pacli-
taxel such as dose-limiting neutropenia, alope-
nia, myalgias, and mucositis. In addition fluid
retention and cutanous toxicities are observed
[445], [460], [461]. Because of the low water sol-
ubility of paclitaxel and docetaxel, the synthe-
sis of more soluble taxoid prodrugs and smaller
analogs has become an interesting area of re-
search (see Section 7.5).
side chain depleted analogs such as baccatin III Their unique capability to inhibit taxol resistant
and its derivatives [456] as well as N-benzoyl- tumor cell lines [470] and their good solubility
(2R,3S)-phenylisoserine are inactive. Simplified in water are the biggest advantages as compared
side chains at C-13 (like acetic, crotonic, or to paclitaxel. Their in vivo activity is similar to
phenylacetic acid) possess reduced activity. that of paclitaxel [469].
3’-Cyclohexyl-3’-dephenylpaclitaxel has a
similar cytotoxicity to paclitaxel. Further mod-
ifications in the aromatic 3’-phenyl group gave
compounds that were equipotent with paclitaxel.
The compound with a 3’-(p-methoxyphenyl) has
a slightly increased activity. Compounds with
different substituents in the N-benzoyl part were
similar to paclitaxel if these substituents were R = H, Epothilone A
aromatic, aliphatic substituents reduced cyto- R = CH3 , Epothilone B
toxicity.
As the 2’-hydroxyl group is essential for max-
imal biological activity [457] esterification leads
to a total loss of activity in the microtubule assay, 8. Heavy-Metal Complexes
whereas cytotoxicity remains unchanged. Thus
various amino acid esters were produced as pro- 8.1. cis-Platinum
drugs.
Notable loss of cytotoxicity was observed cis-Platinum (NSC-119875) [15663-27-1],
with A-ring modified analogs and oxidation at cis-diamminedichloroplatinum, cisplatin,
C-10 or C-7. 7-Acetylpaclitaxel and the C-7 PtCl2 (NH3 )2 , M r 300.05, was the first heavy-
epimer were similar in their ability to inhibit cell metal compound to be introduced into clinical
proliferation. All derivatives without the intact cancer chemotherapy. The discovery of this
oxetane moiety are inactive [458]. Further, all unique agent was predicated on the serendipi-
C-4 modified analogs were devoid of activity tous observation that bacterial growth was in-
underscoring the vital importance of these func- hibited when culture medium was subjected to
tional groups [459]. an alternating current using platinum electrodes
[471]. Moreover, the spent medium itself devel-
oped bacteriocidal characteristics, even in the
7.5. Epothilone A and B absence of electrical current. Detailed analysis
confirmed that, of the several platinum species
The high cytotoxicity and good stabiliza- produced by electrolysis, it was the cis isomer of
tion of microtubule raised interest in the PtCl2 (NH3 )2 that had antibacterial activity. In
natural products epothilone A [152044-53-6] 1969, Rosenberg and co-workers reported that
and B [152044-54-7] originally isolated from cis-platinum also had potent antitumor activ-
myxobacteria Sorangium cellulosum [462]. ity in murine tumors [472], and phase I clinical
Since Bollag et al. [463] reported on the mech- studies with the drug were initiated two years
anism of action, which resembles that of pacli- later [473], [474]. Cis-platinum, in combination
taxel numerous reports were published on total with other agents, has led to highly active and
synthesis [464–468] and biology of epothilones. often curative regimens in patients with testic-
34 Cancer Chemotherapy
ular, ovarian, and head and neck cancer [475], hemolytic anemia, hypomagnesemic tetany, al-
[476]. lergic reactions, and hepatotoxicity. Nephrotox-
icity was dose limiting in early clinical trials
and appeared to be due to tubular reabsorption
8.1.1. Mechanism of Action of active platinum species causing proximal and
distal tubule necrosis [483–485]. Tubular dam-
As with the alkylating agents, one of the ma- age has been reported to cause defective reab-
jor factors mediating the cytotoxicity of cis- sorption of magnesium, resulting in hypomagne-
platinum is probably the formation of cross-links semia (which may result in tetany) [486], [487],
between opposing strands of DNA (interstrand as well as inappropriate renal loss of calcium,
cross-links), linkage within a single strand of potassium, and phosphorus [488]. The acute re-
DNA (intrastrand cross-links), or the formation nal toxicities of cis-platinum appear to be sec-
of linkages dependent on the hydrolysis of cis- ondary to activation of the renin – angiotensin
platinum in solution. While the covalent stabil- system, resulting in reduced renal blood flow and
ity of Pt−NH3 bonds is high, both chlorides glomerular filtration [484], [489]. Thus, this tox-
are good leaving groups and can be displaced icity can be mitigated by high-volume diuresis
by water or hydroxyl ions to form positively [490]. In 1984 it has been demonstrated that even
charged, aquated platinum species that avidly re- high doses of cis-platinum are well tolerated
act with nucleophilic sites on macromolecules, when administered with high-volume chloresis,
especially the N 7 position of guanine and the which not only dilutes urinary platinum levels
N 3 position of cytosine [477]. Formation of the but also prevents leaving of the chloride groups
active intermediate is inhibited in the presence to form the toxic aquated molecule [491]. When
of Cl− ; in plasma the Cl− concentration is suffi- administered by this regimen, the limiting side
cient to inhibit aquation of the drug, which has an effects of cis-platinum become neurotoxicity
in vitro plasma half-life of several hours [478]. and myelosuppression, while renal function is
However, this process occurs rapidly in the in- remarkably spared.
tracellular milieu, where Cl− concentrations are
low.
The cis configuration is central to cytotox- 8.1.2. Mechanisms of Resistance
icity; the trans isomer is devoid of antitumor
effects. This fact suggests that of the reactions Although bacterial resistance to cis-platinum ap-
caused by the aquated platinum species, forma- pears to be due to increased efficiency of DNA
tion of intrastrand cross-links is most important repair [492], the mechanisms of tumor cell re-
(intrastrand cross-links cannot be formed by the sistance are less clear. Alkaline elution stud-
inactive trans compound). ies have demonstrated a direct relation between
As a result of nucleophilic attack on macro- DNA cross-linking and tumor cell resistance
molecules, cis-platinum causes changes in the [493–495], and a cis-platinum-resistant murine
structural conformation of DNA [479] as well as leukemia line has been described in which cross-
intra- and interstrand cross-links. These changes links are formed at a reduced rate [496]. How-
inhibit RNA transcription from the DNA tem- ever, it remains uncertain whether this resis-
plate [480] and, probably more important, di- tance is secondary to accumulation, impaired
rectly inhibit DNA synthesis itself [481]. In ad- activation, or altered DNA repair processes. As
dition, cis-platinum can react with tumor cell with the alkylating agents in general, high lev-
membrane to cause presentation of new anti- els of metallothionein have been reported in cis-
genic determinants [482]. Whether this mech- platinum-resistant cells [497]. This sulfhydryl-
anism is related to tumor cell recognition and rich protein is known to protect from metal tox-
immune response remains to be determined. icity by specifically binding to platinum, cad-
As a heavy-metal-based compound, cis- mium, and other heavy metals [498].
platinum has unique clinical toxicities, including Because the cellular mechanisms of resis-
renal tubular damage, severe nausea and vomit- tance to cis-platinum are so poorly character-
ing, high-tone hearing loss, and peripheral neu- ized, new platinum analogs in clinical trial have
ropathy. Less common are myelosuppression,
Cancer Chemotherapy 35
been selected in an attempt to reduce host toxic- 418 [514], nedaplatin (NSC-375101D, 254-
ity with the aim of improving therapeutic index S), [095734-82-0], cis-diamine(glycolato-
[499]. O1 ,O2 )platinum, C2 H8 N2 O3 Pt, M r 303.19
[515], and oxaliplatin (OHP) [61825-94-3],
[Sp-4-2-(1R-trans)](1,2-cyclohexanediamine-
8.2. Carboplatin N,N’)[ethanedioxato(2− )-O,O’]-platinum,
C8 H14 N2 O4 Pt [516] were clinically investi-
Carboplatin (NSC-241240, CBDCA) gated in depth. Nedaplatin entered the market
[839805-03-3], cis-diammine[1,1-cyclo- in 1997 for the treatment of head and neck,
butanedicarboxylato-(2)-O,O’]platinum(II), small-cell lung, non-small cell lung, esophageal,
C6 H12 N2 O4 Pt, M r 371.3, is a second genera- bladder, testicular, ovarian, and uterine cervical
tion cisplatin analog without significant nephro- cancers [517–520].
toxicity or neurotoxicity and with less emetic Trade name: Agupla (Shionogi).
potential than the parent compound [500–503]. Further studies of additional “third-
Clinical trials show activity against several tu- generation” compounds are centering
mor types. Carboplatin is especially effective on the elimination of toxicity, en-
in treatment of ovarian and small-cell lung can- hanced therapeutic activity, non-cross-
cer [504], [505]. The minimal emetogenic dose resistance and selective drug delivery
of cis-platinum in dogs is 9 mg/m2 , whereas [521]. Ormaplatin (NSC-363812, U-77233)
for carboplatin the dose is 624 mg/m2 . In addi- [62816-98-2] [±(trans)]-tetrachloro(1,2-cyclo-
tion, this compound retains antitumor activity in hexanediamine-N,N’]platinum, C6 H14 Cl4 N2 Pt
cis-platinum-restistant murine leukemia [506], and Lobaplatin (D-19466) [135558-11-1] are
demonstrating consistent antitumor activity in representatives of a series of platinum com-
patients with ovarian cancer in the absence of plexes in clinical development [522–524].
either ototoxicity, nephrotoxicity, or neuropathy
[507], [508]. Carboplatin entered the market in
1989 and is the leading platinum complex can-
cer drug with sales of $ 373 × 106 worldwide
in 1996. Carboplatin is indicated for the treat- Nedaplatin Lobaplatin
ment of ovarian cancer and sales have benefited
from the drug’s use in combination with Taxol
[509–512].
Trade names: Paraplatin (Bristol-Myers
Squibb), Carboplat (BMS).
Iproplatin
Panomifene
9.1.3. Analogs
Faslodex
Idoxifene
38 Cancer Chemotherapy
Fadrazole Letrozole
Atamestane
Cancer Chemotherapy 39
9.3. Antiandrogens
9.3.1. Flutamide
Prostate cancer is primarily a disease of the el-
derly and the second most common cancer in Flutamide (Sch-13521, NK-601, FTA)
men in the United States. Ever since Huggins [013311-84-7], 2-methyl-N-[4-nitro-3(trifluor-
and Hodges demonstrated the partial androgen methyl)phenyl]-propanamide, C11 H11 F3 N2 O3
dependence of most prostatic tumors more than was discovered in the early 1970s. Unlike the
50 years ago, androgen deprivation has become steroids it can easily be synthesized [576], [577]
40 Cancer Chemotherapy
and is devoid hormonal activities. Total sales of Trade name: Anandron (Roussel-Uclaf).
the product were $ 271 × 106 in 1996.
Trade names: Eulexin and Drogenil Mechanism of Action. Nilutamide compet-
(Schering-Plough). itively inhibits binding of androgens to the cy-
tosolic androgen receptor. Administration over
Mechanisms of Action. In a comparative 7 days to immature, castrated male rats, nilu-
study in castrated rats flutamide was shown to tamide inhibited the increase of prostate weights
be equipotent to cyproterone acetate as an an- induced by testosterone in a dose-dependent
tiandrogen [578]. Several groups had observed manner [588]. In rat pituitary cells nilutamide
that flutamide was a more potent antiandrogen in reverses the inhibition of LHRH-induced LH re-
vivo than in vitro and suggested the involvement lease elicited by dihydrotestosterone. It is prob-
of an active metabolite [579], [580]. The major ably because of these effects that nilutamide
metabolite was identified as 2-hydroxyflutamide is recommended for use only in surgically or
analog (Sch-16423) [52806-53-8] and high lev- medically castrated males [589]. Nilutamide has
els of 2-hydroxyflutamide in the plasma led demonstrated an antiandrogenic action in sev-
to the conclusion that this was the active eral animal tumor models [590].
form of flutamide [581]. Although the plasma Single dose kinetics of nilutamide in volun-
levels of testosterone increased on flutamide teers (100 mg) indicate a half-life of 43 ± 3 h,
treatment, the levels of testosterone and di- compared to 5.2 h for flutamide (200 mg) [591].
hydrotestosterone in androgen target tissue were Clinical trials with nilutamide have concen-
reduced [582]. Flutamide exerts its antiandro- trated on combination therapy with surgical or
genic action by blocking the binding of andro- medical castration. The side effects observed in
gens to the cytosolic androgen receptor and / or patients include hot flushes, nausea, vomiting,
inhibiting the nuclear binding of androgens in and visual problems [592].
the target tissue [581].
In patients improvements were seen in pain
relief, prostatic enlargement and induration, re-
duction of metastases, and increase in body
weight and phosphatase. Most common side ef-
fects of flutamide therapy are gynecomastia and
breast tenderness [584]. More recent trials use a Nilutamide
combination of flutamide with LHRH agonists
(see Section 9.4). This is the concept of maxi-
mal androgen withdrawal in which the LHRH 9.3.3. Bicalutamide
agonist wipes out androgens of testicular origin
and the antiandrogen blocks the action of andro- Bicalutamide (ICI-176334) [090357-06-5],
gens of adrenal origin at the androgen receptor (±)-4-[3-(4-fluorophenylsulfonyl)-2-hydroxy-
[585–587]. The therapeutic benefits seem to be 2-methylpropionamido]-2-(trifluoromethyl)benzo-
greatest in patients with minimal disease at the nitrile, C18 H14 F4 N2 O4 S, M r 430.37 was dis-
start of treatment. covered at ICI / Zeneca and selected from more
than 1000 compounds as having the desired
properties of a pure, nonsteroidal, peripher-
ally selective antiandrogen [593], [594]. The
product was launched for the treatment of ad-
vanced prostate cancer in combination with
Flutamide LHRH analogs or surgical castration.
Trade name: Casodex (Zeneca).
binds some 50 times less effectively than di- LHRH agonists (so-called superagonists) are
hydrotestosterone and about 100 times less ef- used in a depot form, which bring about desen-
fectively than R-1881 to the prostate androgen sitization of the pituitary receptors and thus in-
receptor, Its affinity for the prostate receptor terrupt the signal cascade. This results in a bio-
is about four-fold higher, that for the pituitary chemical “castration”, which opens up new ther-
receptor ten times higher than that of hydrox- apeutic possibilities for hormone-dependent dis-
yflutamide [595]. In vivo studies revealed that eases such as prostate cancer, breast cancer, and
bicalutamide is about five times as potent as endometriosis. Although superagonists are gen-
flutamide after oral application. Bicalutamide erally well tolerated, they have the disadvantage
did not cause a significant elevation in LH or that hormone secretion (estrogen, testosterone)
testosterone at any of the doses tested, whereas is initially stimulated before the depletion of re-
flutamide elicited increases. Half-life of bicalu- ceptors or down regulation can take place, and
tamide in prostate cancer patients who received thus the illness temporarily worsens [605]. This
10, 30 or 50 mg/d bicalutamide was around 6 d. has led to the development of the third concept:
The compound was well tolerated in all doses use of LHRH antagonists [606] (see Fig. 3). In
[596–598]. the late 1980s about 5000 LHRH analogs had
Nilutamide and bicalutamide offer advan- been synthesized worldwide and tested in vitro
tages over flutamide because of their long half- or in vivo [607–609]. Whilst LHRH agonists
lifes and sustained serum levels on once-daily have been on the market for about ten years,
dosing. The latter is essential to prevent andro- the LHRH antagonists that have been developed
gen stimulation. farthest are still in clinical testing [610–614].
Figure 3. Antitumor activity of LHRH agonists and antagonists in femaleFSH = follicle stimulating hormone;
ACTH = adrenocorticotropic hormone
Table 1. Structure of LHRH agonists on the market (given are only those amino acid residues that are different in LHRH).
1 2 3 4 5 6 7 8 9 10
LHRH Glp[a] His Trp Ser Tyr Gly Leu Arg Pro Gly-NH2
Buserelin (Hoechst) d-Ser(tBu) Gly-NHEt
Nafarelin d-(2)Nal
Leuprorelin (Abbott, d-Leu Gly-NHEt
Takeda)
Goserelin (Zeneca) d-Ser(tBu) Azagly-NH2
Histrelin (Ortho) d-His(Bzl) Gly-NHEt
Triptorelin (Ferring) d-Trp
highest turnover. The Lupron line of products is responding C-terminal heptapeptide, and then
the leading anticancer hormone drug with world- the N-terminal dipeptide is condensed with this
wide sales in 1996 of $ 810 × 106 followed by to form the complete sequence. Control of the
Zoladex with sales of $ 563 × 106 . physicochemical process parameters, such as
Annual production is less than 100 kg for concentrations, precipitation, separations, reac-
buserelin and significantly over 100 kg for tion temperature profiles, and purification tech-
the market leader, leuprorelin. At this or- niques, is important for successful scale-up of
der of magnitude, substances are only pro- peptide synthesis to a technical scale [617].
duced by classical organic preparative syn-
thesis (fragment condensation in solution).
Hoechst, for example, synthesizes the non- 9.4.1.1. Leuprorelin Acetate
apeptide buserelin from the units pyrog- Because leuprorelin inhibits the synthesis of
lutamic acid-histidine (Glp-His), tryptophan- androgen and estrogen, the drug blocks the
serine-tyrosine (Trp-Ser-Tyr), and d-Serine- growth of hormone-dependent tumors by shut-
tert-butyl ether-leucine-arginine-proline-NHEt ting down testosterone production. Leuprore-
(D-Ser(tBu)-Leu-Arg-Pro-NHEt); the tri- and lin is indicated for the treatment of advanced
tetrapeptide units are coupled to form the cor- prostate cancer, as an alternative to castration
Cancer Chemotherapy 43
for the treatment of endometriosis, and for the extremities in animal experiments, due in part
presurgical management of patients with anemia to massive histamine release by mast cell de-
caused by benign fibroid tumors. granulation. Cyanosis and respiratory impair-
Trade name: Lupron (Abbott, Takeda). ment were also observed [620]. The cause of
these intolerable side effects is thought to be the
combination of d-arginine at position 6 with the
9.4.1.2. Goserelin three aromatic amino acids at the N-terminus of
the sequence [621]. For the desired biological
As certain prostate tumors grow in response to antagonist potency, a d-configured basic amino
testosterone, goserelin prevents the production acid is necessary at position 6. A. V. Schally
of testosterone in testes and is therefore indi- et al. achieved the breakthrough to highly ac-
cated for the treatment of prostate cancer, ad- tive antagonists free of side effects with the
vanced breast cancer, and endometriosis. derivatives SB-75 (cetrorelix) [120287-85-6]
Trade name: Zoladex (Zeneca). and SB-88 [120287-93-6], by incorporating hy-
drophilic, nonbasic amino acids with side-chain
carbamoyl functions at position 6 (d-Citrulline6 ,
9.4.2. LHRH Antagonists d-homocitrueline) [621], [622].
Today, e.g., cetrorelix is manufactured ex-
Common to the intrinsic activity of all superag- clusively by classical fragment condensation on
onists is the initial temporary stimulation of go- a kilogram scale. Two synthesis strategies pro-
nadotropin release. Soon after the use of highly ceed via either the N-terminal tripeptide (d-
active agonists became an established therapy, Nal1 -d-Cpa2 -d-Pal3 ) or the C-terminal tripep-
a search began for corresponding antagonists, tide (Arg8 -Pro9 -d-Ala10 ) and the complemen-
which do not bring about an initial hormone tary heptapeptide (Ser4 -d-Ala10 ) or (Nal1 -Leu7 )
release, to avoid this therapeutically counter- to the protected decapeptide with tert-butyl side
productive effect. A final big hurdle for clini- chain protection. The deprotection with hy-
cal use of highly active antagonists was the in- drochloric acid is followed by final purification
herent anaphylactic potential of these peptides. by preparative HPLC. The C- and N-terminal
Starting from the sequence of native LHRH, functionalization of the acetylated decapeptide
the individual positions of the peptide chain amide, necessary for biological activity and to
were examined in rapid succession for their avoid rapid enzymatic degradation, is introduced
contribution to biological activity. Particular at- at the level of the terminal tripeptide. Function-
tention was paid to side effects. The most ef- alization is achieved by acetylation of the free
fective early improvements in antagonistic ac- α-amino group of naphthylalanine1 with acetyl-
tivity were achieved by using d-phenylalanine hydroxysuccinimide, or amminolysis of the re-
(d-Phe) instead of histidine at position 2, by sulting alanine10 methyl ester in alcoholic am-
d-amino acids at position 6 instead of Gly, monia [623].
and the exchange of C-terminal glycine for d- Folkers et al. were able to improve active
alanine (d-Ala10 ). Further stepwise optimiza- antagonists successively by consistent and sys-
tion led to the sequence scheme now usual tematic modifications both at the relevant se-
for all modern antagonists of d-Nal1 -d-Cpa2 -d- quence positions and the side chain substituents;
Pal3 (Nal = 2-naphthylalanine, Cpa = Phe(4-Cl), the most successful were complex substitutions
Pal = 3-pyridylalanine) as hydrophobic cluster, at the positions 5, 6 and, in part, 8 [624–628].
a d-configured aromatic or aliphatic, yet hy- An important contribution was made by Rivier
drophilic, aminocarboxylic acid at position 6, et al., who synthesized the decapeptide “aza-
and the C-terminal hydrophilic sequence Xxx8 - line” with novel modifications at positions 5
Pro9 -d-Ala10 where Xxx is either arginine or and 6, where aminotriazole-substituted p-ami-
isopropyllysine. Excellent documentation of the nophenylalanine or lysine are positioned [629–
stepwise optimization can be found in [618], 631]. Azaline B [134457-28-6] is probably one
[619]. of the most active antagonists presently available
Antagonists of the second generation caused worldwide.
formation of temporary edemas of the face and
44 Cancer Chemotherapy
Deghenghi published a highly active de- bioavailability), yet have a high binding affin-
capeptide sequence with minimal histamine re- ity. Ideally, such substances should be able to be
lease and good water solubility. This structure, administered orally, be sufficiently stable in the
known as antarelix [151277-78-5], differs from organism, and possess favorable pharmacolog-
SB-75 (cetrorelix) in that it has homocitrulline6 ical parameters comparable to peptide antago-
instead of citrulline6 and isopropylysine8 in- nists. Clinical investigations of the influence of
stead of arginine8 [633]. By using Lys(iPr)8 , the antimycotic ketoconazole on prostate can-
residual potential for histamine release can be cer and testosterone suppression indicate that it
further reduced. Organon is developing the an- may have a LHRH-mimetic effect [648], [649].
tagonist ganirelix (RS26306) [124904-93-4] un- Further investigations by Abbott showed weak
der license from Syntex; this is a decapep- antagonistic activity both in vitro and in vivo for
tide with novel alkyl-modified d- and l- ketoconazole and its modified analogs; however,
homoarginine units at positions 6 and 8 [634]. the biological activity of such derivatives is most
With ramorelix (HOE 013) [127932-90-5], probably not mediated mainly by LHRH an-
Hoechst (HMR) has a peptide antagonist with tagonism [650], [651]. Complex, highly substi-
a sugar – amino acid unit (O-α-l-rhamnosyl- tuted nitrogen heterocycles seem to have LHRH
d-serine6 ), that has improved water solubility antagonistic potential [652]. In patent litera-
[632]. Schering has also synthesized peptide an- ture benzodiazepines, benzodiazepinones, het-
tagonists, using nonproteinogenic amino acids, erocyclic benzo-substituted alkylamines, and
such as ε-dialkylated lysine or benzodiazepine thienopyridine carboxylic acid derivatives are
aminocarboxylic acids [635], [636]. Abbott’s A- described as LHRH receptor antagonists with
76154 [136989-30-5] is an octapeptide antago- receptor-binding inhibition at submicromolar
nist with LHRH receptor affinity of a similar or- concentrations, suitable amongst other uses as
der of magnitude to that of active decapeptides antitumor agents for hormone-dependent tumors
such as A-75998 [135215-95-1] or “Nal-Glu” [647], [653]. A lead structure from the latter sub-
[103733-02-4] [644]. stance class is in extensive pharmacological tri-
als [654–656].
9.4.2.2. Peptidomimetics
exerts substantial antitumor activity in appro- treated with oral capsules were terminated early
priate models. Because of its special pharma- due to gastrointestinal intolerance [682].
cology – high activity against mammary carci-
noma and low toxicity to normal tissue – it is
an ideal candidate for topical treatment of cu- 11. Economic Aspects
tanous breast cancer metastases. The substance
was launched in 1993 as a topical formulation The world cancer drug market is expected to
for palliative treatment of refractory skin metas- reach $ 14 × 109 by 2000 [10], [14]. Total sales
tases of breast cancer [676]. of the top 100 anticancer drugs generated a
Trade name: Miltex (ASTA Medica). turnover of $ 4.66 × 109 in 1996. Of the worlds
A variety of derivatives were synthesized and eight top-selling anticancer drugs, four – the
characterized, some of which appeared to be prostate cancer-drugs flutamide (Eulexin), bica-
considerably less toxic to the gastrointestinal lutamide (Casodex), leuprorelin (Lupron) and
tract [677]. One of them, perifosine (D-21266) goserelin (Zoladex) – are merely palliative,
[157716-52-4] is currently in clinical trials. yet have combined annual sales of $ 1.7 × 109 ,
Mechanism of Action. Miltefosine shows whereas sales of the breast cancer drugs tamox-
highly selective antineoplastic activity in ifen (Nolvadex) and paclitaxel (Taxol) are ap-
DMBA- and N-methyl-N-nitrosourea (MNU)- proaching $ 500 × 106 and $ 850 × 106 , respec-
mammary carcinoma of the rat. The activity tively.
is not due to an antiestrogenic effect in these Taxol, BMS’s drug for treating ovarian and
estrogen-dependent tumor models [678]. The breast cancers, was the biggest selling anticancer
treatment was tolerated and no overt toxic symp- agent in 1996, with sales of $ 813 × 106 .
toms were observed. The mechanism of action
of miltefosine is most probably different from
that of other chemotherapeutic agents [679]. 12. References
Incorporation of miltefosine and metabolites
into biological membranes was demonstrated, 1. I. B. Weinstein, Cancer Res. (Suppl.) 51
affecting the interactions of receptor proteins (1991) 5080.
or membrane associated enzymes involved in 2. R. Doll, Eur. J. Cancer 26 (1990) 5007.
3. C. LaVecchina, F. Levi, F. Lucchini, S.
growth control and cellular signalling [680].
Garattini, Anti-Cancer-Drugs 2 (1991) 215.
The reduction in tumor mass was accompanied 4. D. Kessel, In Vivo 8 (1994) 829.
by morphological changes compatible with the 5. T. Tsurno, A. Toninda, Anti-Cancer-Drugs 6
induction of differentiation [675], [681]. Fur- (1995) 213.
thermore, the decrease of tumors was mainly 6. G. Eisenbrand, S. Lauch-Birkel, W. C. Tang,
due to cell loss by apoptosis. Synthesis (1996) 1246.
7. G. B. Elion, Cancer Res. 45 (1985) 2943.
8. Scrip 1997, July 15, p. 20.
9. Chem. Marketing Report, July 14, 1997, p. 19.
10. Med. Ad-News, May 1996, p. 56.
11. G. M. Cragg, D. J. Newman, K. M. Snader, J.
Nat. Prod. 60 (1997) 52 – 60.
Ilmofosine 12. K. Nasmyth, Science 274 (1996) 1643 – 1645.
13. G. Dratta, H. Pagano, Annu. Rep. Med. Chem.
31 (1996) 241 – 248.
14. Pharma Business, July/August 1997, p. 62.
15. S. A. Rosenberg: “Principles of Surgical
Oncology,” in V. T. DeVita, S. Hellman, S. A.
Miltefosine
Rosenberg (eds.): Cancer: Principles and
In clinical trials mild dryness and flaking of Practice of Oncology, J. B. Lippincott Co.,
the skin were noted, but no systemic toxicity Philadelphia 1982, pp. 93 – 102.
was reported after topical administration. Sev- 16. S. Farber, L. K. Diamond, R. D. Mercer, R. F.
eral phase I and II studies in patients that were Sylvester, V. A. Wolff, N. Engl. J. Med. 238
(1948) 787 – 793.
Cancer Chemotherapy 47
65. P. Reyes, T. C. Hall, Biochem. Pharmacol. 18 91. L. H. Van der Ploeg, R. A. Flavill, Cell 19
(1969) 1587 – 1590. (1980) 947 – 958.
66. D. K. Kasbeker, D. M. Greenberg, Cancer Res. 92. O. Niwa, T. Sugarhar, Proc. Natl. Acad. Sci.
23 (1963) 818 – 825. USA 78 (1981) 6290 – 6294.
67. P. Reichard, O. Skold, G. Klein, Nature 93. S. Friedman, Molec. Pharmacol. 19 (1980)
(London) 183 (1959) 939 – 941. 314 – 320.
68. J. A. Houghton, S. J. Masada, J. O. Phillips et 94. D. V. Santi, C. E. Garrett, P. J. Barr, Cell 25
al., Cancer Res. 42 (1982) 144 – 149. (1983) 9 – 10.
69. C. Heidelberger, G. Kaldos, K. L. Mukherjee 95. P. G. Constantinides, P. A. Jones, W. Geness,
et al., Cancer Res. 20 (1960) 903 – 909. Nature (London) 267 (1977) 364 – 366.
70. M. M. Lastieboff, B. Kedzioska, W. Rode, 96. P. A. Jones, S. M. Taylor, Cell 20 (1980)
Biochem. Pharmacol. 32 (1983) 2259 – 2267. 85 – 93.
71. A. R. Bapat, C. Zarow, P. V. Dannenberg, J. 97. T. J. Ley, J. DeSimone, N. P. Anagnov et al., N.
Biol. Chem. 258 (1983) 4130 – 4136. Engl. J. Med. 307 (1982) 1469 – 1475.
72. C. C. Rossana, L. G. Rao, L. F. Johnson, Mol. 98. J. Vesely, A. Chiak, Pharmacol. Therap. 2
Cell. Biol. 2 (1982) 1118 – 1125. (1978) 813 – 840.
99. T. Lee, M. R. Kovan, Biochem. Pharmacol. 25
73. F. Baskin, S. C. Carlin, P. Kraus et al., Molec.
(1976) 1737 – 1742.
Pharmacol. 11 (1975) 105 – 117.
100. A. Cihak, H. Vessela, F. Sorm, Biochem.
74. D. G. Priest, B. E. Ledford, M. T. Doig,
Biophys. Acta 166 (1968) 277 – 279.
Biochem. Pharmacol. 29 (1980) 1549 – 1553.
101. A. Cihak, J. Vesely, Biochem. Pharmacol. 21
75. R. D. Armstrong, E. Cadman, Cancer Res. 43
(1972) 3257 – 3265.
(1983) 2525 – 2528. 102. J. Vesely, A. Cihak, F. Sorm, Int. J. Cancer 2
76. J. L. Au, Y. M. Rustum, J. Minonda, B. I. (1967) 639 – 646.
Sriva-stava, Biochem. Pharmacol. 32 (1983) 103. J. Doskacil, F. Sorm, FEBS Lett. 2 (1974)
541 – 545. 30 – 32.
77. Y. M. El Sayed, W. Sadee, Cancer Res. 43 104. D. D. Von Hoff, M. Slavik, F. M. Muggia, Ann.
(1983) 4039 – 4044. Intern. Med. 85 (1976) 237 – 245.
78. E. J. Ansfield, G. J. Kallas, J. P. Simpson, J. 105. Z. H. Israili, W. R. Vogler, E. S. Mingidi et al.,
Clin. Oncol. 1 (1983) 107 – 110. Cancer Res. 36 (1971) 1453 – 2461.
79. F. Sorm, A. Piskala, A. Cihak et al., 106. J. A. Beisler, J. Mol. Chem. 21 (1978)
Experientia 20 (1964) 202 – 203. 204 – 208.
80. A. Piskala, F. Sorm, Collect. Czech. Chem. 107. J. A. Beisler, M. M. Abbasi, J. S. Driscoll,
Commun. 29 (1964) 2060 – 2075. Cancer Treat. Rep. 60 (1976) 1671 – 1674.
81. P. G. W. Plagemann, M. Behrens, D. Abraham, 108. G. A. Curt, J. A. Kelley, R. L. Fine et al., Proc.
Cancer Res. 38 (1978) 2458 – 2466. Am. Assoc. Clin. Oncol. 3 (1984) 37.
82. J. C. Drake, R. G. Stoller, B. A. Chabner, 109. P. G. W. Plagemann, R. Marz, R. M. Wolhvete,
Biochem. Pharmacol. 26 (1977) 64 – 66. Cancer Res. 38 (1978) 978 – 989.
83. T. Lee, M. Karon, R. L. Momparler, Cancer 110. A. Iwagaki, T. Nakamura, G. Wakisaka,
Res. 34 (1974) 2481 – 2488. Cancer Res. 29 (1969) 2169 – 2176.
84. A. Cihak, Collect. Czech. Chem. Commun. 39 111. M. Y. Chu, G. A. Fisher, Biochem. Pharmacol.
(1974) 3782 – 3792. 11 (1962) 423 – 430.
85. J. Doskel, V. Paces, F. Sorm, Biochim. 112. P. P. Major, E. M. Egan, G. P. Beardsley, M. D.
Biophys. Acta 145 (1967) 771 – 779. Minden et al., Proc. Natl. Acad. Sci. USA 78
86. L. H. Li, E. J. Olin, T. J. Fraser et al., Cancer (1983) 3235 – 3239.
Res. 30 (1970) 2770 – 2775. 113. P. P. Major, L. Sargent, E. M. Egan, D. W.
87. B. F. Vanyosbin, S. G. Tkacheug, A. W. Kufe, Biochem. Pharmacol. 30 (1981)
Belozinsky, Nature (London) 225 (1976) 2221 – 2224.
948 – 949. 114. P. P. Major, E. M. Egan, D. Herrick, D. W.
88. J. L. Mandell, P. Chambon, Nucleic Acids Res. Kufe, Biochem. Pharmacol. 31 (1982)
7 (1979) 2081 – 2103. 861 – 866.
89. J. D. McGhee, G. D. Gineler, Nature (London) 115. M. Y. Chu, G. A. Fisher, Biochem. Pharmacol.
280 (1979) 419 – 420. 14 (1965) 333 – 341.
116. M. R. Atkinson, M. Deutscher, A. Kornberg,
90. C. Scheir, T. Maniatis, Proc. Natl. Acad. Sci.
A. Russel et al., Biochemistry 8 (1969)
USA 77 (1980) 6634 – 6638.
4897 – 4904.
Cancer Chemotherapy 49
117. P. A. Diskwel, F. Wanka, Biochim. Biophys. 141. Eli Lilly, EP 184365, 1986 (G. B. Grindey,
Acta 520 (1978) 461 – 471. L. W. Hertel).
118. R. J. Frann, C. M. Egan, D. W. Kufe, Leuk. 142. Eli Lilly, EP 306190, 1989T. S. Chou, P.
Res. 7 (1983) 243 – 249. Heath). L. E. Patterson).
119. J. F. Ward, E. I. Jones, W. F. Blakely, Cancer 143. S. Chubb et al., Proc. Am. Assoc. Cancer Res.
Res. 44 (1984) 59 – 63. 28 (1987) Abst. 1282.
120. S. R. Bähring-Kuhlmey, Drugs of Today 13 144. D. Y. Bouffard, L. F. Momparler, R. L.
(1977) 475. Momparler, Eur. J. Pharmacol. 183 (1990) 2,
121. A. A. Claesen et al., Tetrahedron Lett. (1966) Abst. 032.
3499. 145. V. Heinemann, L. W. Hertel, G. B. Grindley,
122. P. Major et al., Proc. Natl. Acad. Sci. USA 78 W. Phukett, Cancer Res. 48 (1988) 14,
(1981) 3235. 4024 – 4031.
123. R. Preston, Treat. Carcinogen Mutagen 1 146. M. N. Serradel, J. Castañer, Drugs of the
(1980) 147. Future 15 (1990) 8, 794 – 797.
124. P. Drahovsky, W. Kreis, Biochem. Pharmacol. 147. V. Heinemann et al., Proc. Am. Assoc. Cancer
19 (1970) 940 – 944. Res. 30 (1989) Abst 2204.
125. W. Kreis, J. Graham, L. A. Damin, Biochem. 148. H. H. Hansen, Ann. Oncol. 120 (1994)
Pharmacol. 31 (1982) 3831 – 3837. 519 – 528.
126. J. Balzarini, E. De Cleireg, Mol. Pharmacol. 149. E. Beutler, Lancet 340 (1992) 8825, 952.
150. H. M. Bryson, E. M. Sorkin, Drugs 46 (1993)
23 (1983) 175 – 181.
5, 872.
127. A. W. Harris, E. C. Reynolds, L. R. Finch,
151. E. H. Estey et al., Blood 79 (1992) 4, 882.
Cancer Res. 39 (1979) 538 – 541.
152. S. P. Smith, Hosp. Formul. 28 (1993) 7, 621.
128. Y. Rustum, H. Priesler, Cancer Res. 39 (1979) 153. C. P. Robinson, Drugs Today 29 (1993) 6, 379.
42 – 49. 154. US Dept. Of Health US 4357324, 1982 (J. A.
129. C. D. Stuart, P. J. Burke, Nat. New Biol. 233 Montgomery, A. T. Shortnacy).
(1971) 109 – 110. 155. R. W. Brockman, Y. C. Cheng, F. M. Schabel,
130. P. Chang, P. H. Wiernik, S. D. Reich et al.: J. M. Montgomery, Cancer Res. 40 (1980)
“Prediction of Response to Cytosine 3610 – 3615.
Arabinoside and Daunorubicin in Acute 156. J. A. Montgomery, Cancer Res. 42 (1982)
Nonlymphocytic Leukemia,” in F. Mandelli 3911 – 3917.
(ed.): Therapy of Acute Leukemias, 157. W. C. Tseng et al., Mol. Pharmacol. 21 (1982)
Lombardo-Editore, Rome 1977, pp. 148 – 159. 474 – 477.
131. J. F. Smyth, A. B. Robins, C. L. Leese, Eur. J. 158. S. J. Hopkins, Drugs of the Future 10 (1985)
Cancer 12 (1976) 567 – 573. 1, 20.
132. M. N. H. Tattersall, U. K. Ganeshaguru, A. V. 159. Annu. Drug Data Rep. (1992) 633 – 634.
Hoffbrand, Br. J. Haematol. 27 (1974) 39 – 46. 160. J. S. Whelan et al., Brit. J. Cancer 64 (1991)
133. M. Aoshima et al., Cancer Res. 37 (1977) 1, 120.
1481. 161. H. S. Hochster et al., J. Clin. Oncol. 10 (1992)
134. F. Cabaninas, Drugs of the Future 5 (1980) 1, 28.
12, 603. 162. H. W. Dion et al., Ann. NY Acad. Sci. 284
135. T. Ueda, T. Nakamura, S. Ando et al., Cancer (1977) 21 – 29.
Res. 43 (1983) 3412 – 3416. 163. Parke Davis, US 3923785, 1975 (A. Ryder et
136. K. Hoie, T. Tsuro, K. Naganuma, S. al.).
Tsukagoshi et al., Cancer Res. 44 (1984) 164. P. W. Woo et al., J. Heterocycl. Chem. 11
172 – 177. (1974) 4, 641 – 643.
137. C. W. Young, R. Schneider, B. Leyland-Jones 165. J. F. Smith et al., Cancer Chemother.
et al., Cancer Res. 43 (1983) 5006 – 5009. Pharmacol. 1 (1978) 49 – 51.
138. Y. C. Cheng, R. S. Tan, J. L. Ruth, G. 166. J. F. Smith et al., Proc. Am. Assoc. Cancer Res.
Dutschman, Biochem. Pharmacol. 32 (1983) 20 (1979).
726 – 729. 167. G. A. Le Page et al., Cancer Res. 36 (1976)
139. L. W. Hertel, J. S. Kroin, J. W. Misner, J. M. 1481 – 1485.
Tustin, J. Org. Chem. 53 (1988) 11, 168. M. N. Seradel, J. Castañer, Drugs of the Future
2406 – 2409. 6 (1981) 419 – 420.
169. M. Blick et al., Am. J. Hematol. 33 (1990) 3,
140. Eli Lilly, EP 122707, 1984 (L. W. Hertel).
205 – 209.
50 Cancer Chemotherapy
170. S. Bernhard et al., Med. Pediatr. Oncol. 19 195. A. R. P. Paterson, D. M. Tidd: “6-Thiopurines,”
(1991) 4, 276. in A. C. Sartorelli, D. G. Johns (eds.):
171. A. D. Ho et al., J. Natl. Canver Inst. 82 (1990) Handbook of Experimental Pharmacology,
17, 1416 – 1420. vol. 38, Springer-Verlag, Berlin 1975, Part 2,
172. E. H. Kraut et al., J. Clin. Oncol. 7 (1989) 2, pp. 384 – 403.
168 – 172. 196. D. L. Hill, L. L. Bennett Jr., Biochemistry 8
173. R. S. Witte et al., Invest. New Drug 10 (1992) (1969) 122.
1,49. 197. R. T. Kavahata, L. F. Chuang, C. A. Holmberg,
174. B. J. Kane, J. G. Kuhn, M. K. Roush, Ann. B. I. Osburn et al., Cancer Res. 43 (1983)
Pharmacother. 26 (1992) 939 – 947. 3655 – 3659.
175. G. B. Elion, E. Burgi, G. H. Hitchings, J. Am. 198. J. Maybaum, H. G. Mandel, Cancer Res. 43
Chem. Soc. 74 (1952) 411. (1983) 3852 – 3856.
176. G. B. Elion, G. H. Hitchings, J. Am. Chem. 199. J. S. Lazo, K. M. Huang, A. C. Sartorelli,
Soc. 77 (1955) 1676. Cancer Res. 37 (1977) 4250.
177. J. H. Burchenal, M. L. Murphy, R. R. Ellison et 200. C. K. Carrico, A. C. Sartorelli, Cancer Res. 37
al., Blood 8 (1953) 965. (1977) 1868.
178. G. H. Hitchings, G. B. Elion, Ann. N.Y. Acad. 201. K. Kim, W. J. Blechman, V. G. H. Riddle, A. B.
Sci. 60 (1954) 195. Pardee, Cancer Res. 41 (1981) 4529.
179. R. W. Brockman, C. S. Debavadi, P. Stutts, 202. J. D. Strobel-Stevens, S. M. El Dareer, M. W.
D. J. Hutchison, J. Biol. Chem. 236 (1961) Trader, D. L. Hill, Biochem. Pharmacol. 31
1471 – 1479. (1982) 3133.
180. J. D. Davidson, Cancer Res. 20 (1960) 203. M. Rosman, H. E. Williams, Cancer Res. 33
225 – 232. (1973) 1202.
181. G. B. Elion, Fed. Proc. 26 (1967) 898 – 904. 204. M. K. Wolpert, S. P. Damle, J. E. Brown et al.,
182. J. F. Henderson, M. K. Y. Khoo, J. Biol. Chem. Cancer Res. 31 (1971) 1620.
240 (1965) 2349 – 2357. 205. M. Rosman, M. L. Lee, W. A. Creasey et al.,
183. F. F. Snyder, J. F. Henderson, Can. J. Biochem. Cancer Res. 34 (1974) 1952.
51 (1973) 943 – 948. 206. E. M. Scholar, P. Calabresi, Biochem.
184. P. C. L. Wong, J. F. Henderson, Biochem. J. Pharmacol. 28 (1979) 445.
129 (1972) 1085 – 1094. 207. L. Tterlikkis, J. L. Day, D. A. Brown, E. C.
185. T. Fields, L. Brox, Can. J. Biochem. 52 (1974) Schroeder, Cancer Res. 43 (1983)
441 – 446. 1675 – 1679.
186. C. D. Green, D. W. Martin Jr., Proc. Natl. 208. S. Zimm, J. M. Collins, D. O’Neill, B. A.
Acad. Sci. USA 70 (1973) 3698 – 3702. Chabner et al., Clin. Pharmacol. Therap. 34
187. G. B. Elion, S. Callahan, W. Rundles, G. H. (1983) 810 – 817.
Hitchings, Cancer Res. 23 (1963) 209. S. Zimm, J. M. Collins, R. Riccardi, D.
1207 – 1217. O’Neill et al., N. Engl. J. Med. 308 (1983)
188. G. H. Hitchings, Cancer Res. 23 (1963) 1005 – 1009.
210. R. D. Armstrong, R. Vera, P. Snyder, E.
1218 – 1225.
Cadman, Biochem. Biophys. Res. Commun.
189. T. Nakamura, Blood & Vessel (Jpn.) 2 (1971)
109 (1982) 595.
237 – 248.
211. M. Inomata, F. Fukuoka, A. Hoshi, K. Kuretani
190. T. Higuchi, T. Nakamura, G. Wakisaka,
et al., J. Pharmacobiodyn. 4 (1981) 928.
Blood &Vessel (Jpn.) 3 (1972) 313 – 318.
212. D. M. Tidd, I. Gibson, P. D. G. Dean, Cancer
191. P. W. Allan, L. L. Bennett Jr., Biochem.
Res. 42 (1982) 3769.
Pharmacol. 20 (1971) 847 – 852.
213. M. Inaba, M. Fukui, N. Yoshida, S. Tsukagoshi
192. A. Hampton, J. Biol. Chem. 238 (1963)
et al., Cancer Res. 42 (1982) 1103.
3068 – 3074.
214. C. P. J. Adair, H. J. Bragg, Ann. Surg. 93
193. R. J. McCollister, W. R. Gilbert, D. M. Ashton,
(1931) 190.
J. B. Wyngaarden, J. Biol. Chem. 239 (1964)
215. L. P. Jacobson, C. L. Spurr, E. S. O. Barron et
1560 – 1563.
al., J. Am. Med. Assoc. 132 (1946) 126.
194. R. P. Miech, R. E. Parks Jr., J. H. Anderson Jr., 216. M. Colvin: “The Alkylating Agents,” in B. A.
A. C. Sartorelli, Biochem. Pharmacol. 16 Chabner (ed.): Pharmacologic Principles of
(1967) 2222 – 2227. Cancer Treatment, W. B. Saunders,
Philadelphia 1982, pp. 276 – 308.
Cancer Chemotherapy 51
217. N. O. Goldstein, R. J. Rutman, Cancer Res. 24 243. J. M. Dornish, I. Smith-Kielland, FEBS Lett.
(1964) 1363. 139 (1981) 41.
218. R. W. Ruddon, J. M. Johnson, Mol. 244. K. D. Tew, A. L. Wang, Molec. Pharmacol. 21
Pharmacol. 4 (1968) 258. (1982) 729.
219. G. P. Wheeler, J. A. Alexander, Cancer Res. 245. A. Begleiter, H. Y. P. Lam, G. J. Goldenberg,
29 (1969) 98. Cancer Res. 37 (1977) 1022.
220. P. Brookes, P. D. Lawley, Biochem. J. 80 246. N. G. Goldstein, R. J. Rutman, Cancer Res. 24
(1961) 486. (1964) 1363.
221. C. C. Price, G. M. Gaucher, P. Koneru et al., 247. R. W. Ruddon, J. M. Johnson, Molec.
Biochim. Biophys. Acta 166 (1968) 327. Pharmacol. 4 (1968) 258.
222. A. Loveless, W. C. J. Ross, Nature (London) 248. J. J. Roberts, T. P. Brent, A. R. Crathorn, Eur. J.
166 (1950) 1113. Cancer 7 (1971) 515.
223. K. W. Kohn, L. C. Erickson, R. A. G. Ewig et 249. W. P. Tong, D. B. Ludlam, Biochim. Biophys.
al., Biochemistry 15 (1976) 4629. Acta 608 (1980) 174.
224. R. J. Rutman, E. H. C. Chun, F. A. Lewis, 250. C. C. Price, G. M. Gaucher, P. Koneru et al.,
Biochem. Biophys. Res. Commun. 32 (1968) Biochim. Biophys. Acta 166 (1968) 327.
251. P. Brookes, P. D. Lawley, Biochem. J. 80
650.
(1961) 486.
225. I. Hirono, Nature (London) 186 (1960) 1059.
252. C. B. Thomas, R. Osieka, K. W. Kohn, Cancer
226. G. Calcutt, T. A. Connors, Biochem.
Res. 38 (1978) 2448.
Pharmacol. 12 (1963) 839. 253. C. C. Erickson, M. O. Bradley, S. M. Ducore et
227. R. A. G. Ewig, K. W. Kohn, Cancer Res. 37 al., Proc. Natl. Acad. Sci. USA 77 (1980) 467.
(1977) 2114. 254. R. A. G. Ewig, K. W. Kohn, Cancer Res. 38
228. L. C. Ericson, G. Laurent, N. A. Sharkey et al., (1978) 3197.
Nature (London) 288 (1980) 727 – 729. 255. W. G. Verly, Y. Paquette, Cancer J. Biochem.
229. D. T. Vistica, J. N. Toal, M. Rabinowitz, 50 (1972) 217.
Biochem. Pharmacol. 27 (1978) 2865. 256. L. C. Ericson, G. Laurent, N. A. Sharkey et al.,
230. W. R. Redwood, M. Colvin, Cancer Res. 40 Nature (London) 288 (1980) 727 – 729.
(1980) 1144. 257. A. R. Crathorne, J. J. Roberts, Nature (London)
231. W. P. Brade, J. Engel, Drugs Today 20 (1984) 211 (1966) 150.
491 – 496. 258. K. Tsujihara, M. Ozeki, T. Morikawa, M.
232. P. J. Creaven, L. M. Allen, D. A. Alford et al., Kawamori et al., J. Med. Chem. 25 (1982) 441.
Clin. Pharmacol. Therap. 16 (1974) 77. 259. Y. Akaike, Y. Arai, H. Takuchi, H. Satoh,
233. T. A. Conners: “Alkylating Agents, Gann 73 (1982) 480.
Nitrosourea, and Alkyltriazines,” in H. M. 260. W. J. Zeller, M. Berger, G. Eisenbrand, W.
Pinedo, B. A. Chabner (eds.): Cancer Tang et al., Arzneim.-Forsch. 32 (1982) 484.
Chemotherapy Annual 5, Elsevier, Amsterdam 261. J. R. Prous, Drug News Perspect 1 (1988) 1,
1983, pp. 30 – 65. 35.
234. A. Mittelman et al., J. Urol. (Baltimore) 115 262. ADIR, FR 2536075, 1984 (C. Cudennec, G.
(1976) 409. Lavielle).
235. W. A. Skinner, H. F. Gram, M. O. Green et al., 263. M. N. Serradel, J. Castañer, Drugs of the
J. Med. Pharmaceut. Chem. 2 (1960) 299. Future 14 (1989) 11, 1042 – 1046.
236. K. A. Hyde, E. Acton, W. A. Skinner et al., J. 264. S. Filippeschi et al., Anticancer Res. 14
Med. Pharmaceut. Chem. 5 (1962) 1. (1988) 11, 1351 – 1354.
237. F. M. Schabel, T. P. Johnston, G. S. McGaleb et 265. D. Khayat et al., Cancer Res. 47 (1987)
al., Cancer Res. 23 (1963) 226. 6782 – 6785.
238. K. W. Kohn, Cancer Res. 37 (1977) 1450. 266. J. A. Boutin et al., Eur. Cancer Oncol. (1989).
239. R. J. Weinkam, D. F. Deen, Cancer Res. 42 267. P. Deloffre, C. A. Cudennec, G. Lavielle, J. P.
(1982) 1008. Bizzari, 15th Int. Cong. Chemother., Istanbul
240. J. Mendel, R. Thust, H. Schwartz, Arch. 1987, Abst. 755.
268. J. Jacquillat et al., Proc. Am. Assoc. Cancer
Geschwulstforsch. 52 (1982) 371.
Res. 30 (1989) Abst. 1088.
241. B. J. Bowdon, J. Grimsley, H. H. Lloyd,
269. P. Zeller, H. Gutmann, B. Hegedus et al.,
Cancer Res. 34 (1974) 194.
Experientia 19 (1963) 129.
242. L. C. Panasci, D. Green, R. Nagourney et al.,
270. V. T. DeVita, A. Serpick, P. Carbone, Ann.
Cancer Res. 37 (1977) 2615.
Intern. Med. 73 (1970) 542.
52 Cancer Chemotherapy
271. J. Gutterman, A. Huang, P. Hochstein, Proc. 300. Microbiochem. Res. Found., US 4303785, (H.
Soc. Exp. Biol. Med. 130 (1979) 797. Naganawa, T. Takenchi, H. Umezawa).
272. R. J. Weinkam, D. A. Shiba, Life Sci. 22 301. H. Umezawa et al., J. Antibiot. 32 (1979)
(1978) 937. 1082 – 1085.
273. D. L. Dunn, R. A. Lubet, R. A. Prough, Cancer 302. M. N. Serradel, J. Castañer, Drugs of the
Res. 39 (1979) 4555. Future 8 (1983) 7, 610 – 611.
274. D. E. Schwartz, W. Bollag, P. Obrecht, 303. T. Nishimura et al., J. Antibiot. 33 (1980)
Arzneim.- Forsch. 17 (1967) 1389. 737 – 743.
275. D. Reed, F. Dost, Proc. Am. Assoc. Cancer 304. D. Dantchev et al., J. Antibiot. 32 (1979)
Res. 7 (1965) 57. 1085 – 1086.
276. M. Baggiolini, B. Dewald, H. Aebi, Biochem. 305. H. Brockmann, Fortschr. Chem. Org. Naturst.
Pharmacol. 18 (1969) 2187. 50 (1973) 121.
277. C. Pueyo, Mutat. Res. 67 (1979) 189. 306. C. E. Myers: “Anthracyclines,” in B. A.
278. E. Therman, Cancer Res. 32 (1972) 1111. Chabner (ed.): Pharmacologic Principles of
279. W. Kreis, Proc. Am. Assoc. Cancer Res. 7 Cancer Treatment, W. B. Saunders,
(1966) 39. Philadelphia 1982, pp. 416 –434.
280. A. Sartorelli, S. Tsunamura, Proc. Am. Assoc. 307. R. Phillips, A. DiMarco, F. Zunino, Eur. J.
Cancer Res. 6 (1965) 55. Biochem. 85 (1978) 487.
281. H. Lam, A. Begleiter, W. Stein et al., Biochem. 308. R. B. Painter, Cancer Res. 38 (1978) 4445.
Pharmacol. 27 (1978) 1883. 309. G. Daskal, C. Woodard, S. T. Crooke et al.,
282. J. L. Skibba, D. D. Beal, G. Ramirez et al., Cancer Res. 38 (1978) 467.
Cancer Res. 30 (1970) 147. 310. W. E. Ross, L. A. Zwelling, K. W. Kohn, Int. J.
283. P. Farina, A. Gescher, J. A. Hickman, J. K. Radiat. Oncol., Biol. Phys. 5 (1979) 1221.
Horton et al., Biochem. Pharmacol. 31 (1982) 311. B. Sinha, R. H. Sik, Biochem. Pharmacol. 29
1887. (1980) 1867.
284. T. L. Loo, G. E. Housholder, A. H. Gerulath et 312. K. Handa, S. Sato, Gann 66 (1975) 43.
al., Cancer Treat. Rep. 60 (1976) 149. 313. N. R. Bachur, S. L. Gordon, M. V. Gee, Cancer
285. D. D. Beal, J. L. Skibba, K. K. Whitnable et al., Res. 38 (1977) 1745.
Cancer Res. 36 (1976) 2827. 314. R. Ogura, H. Toyama, T. Shimada et al., J.
286. A. H. Gerulath, T. L. Loo, Biochem. Appl. Biochem. 1 (1979) 325.
Pharmacol. 21 (1972) 2335. 315. J. H. Doroshow, G. Y. Locker, C. E. Myers, J.
287. D. W. Kaiser, I. T. Thurston, J. R. Dudley et al., Clin. Invest. 65 (1980) 128.
J. Am. Chem. Soc. 73 (1951) 2984. 316. N. W. Revis, N. Marusic, J. Mol. Cell. Cardiol.
288. J. F. Worzalla, B. D. Kaima, B. M. Johnson et 10 (1978) 945.
al., Cancer Res. 34 (1974) 2669. 317. T. R. Tritton, S. A. Murphree, A. C. Sartorelli,
289. J. F. Worzalla, B. D. Kaima, B. M. Johnson et Biochem. Biophys. Res. Commun. 84 (1978)
al., Cancer Res. 33 (1973) 2810. 802.
290. B. C. V. Mitchley, S. A. Clarke, T. A. Connors 318. T. R. Tritton, G. Yeh, Science 217 (1982) 248.
et al., Cancer Treat. Rep. 61 (1977) 3. 319. E. Goormaghtigh, R. Brasseur, J. M.
291. M. Ames, G. Powis, J. S. Kovach et al., Cancer Ruysschaert, Biochem. Biophys. Res.
Res. 39 (1979) 5016. Commun. 104 (1982) 314.
292. L. M. Lake, E. E. Grunden, B. M. Johnson, 320. C. E. Myers, C. Simone, L. Gianni et al., Proc.
Cancer Res. 35 (1975) 2858. Am. Assoc. Cancer Res. 22 (1981) 112.
293. A. J. Cumber, W. C. J. Ross, Chem. Biol. 321. T. Oki, Jpn. J. Antibiot. 30 (1977) 570.
Interact. 17 (1977) 349. 322. R. K. Y. Zee-Cheng, C. C. Cheng, J. Med.
294. V. Iver, W. Szybalski, Science (Washington) Chem. 21 (1978) 291.
145 (1964) 55 – 58. 323. L. H. Patterson, B. M. Gandecka, J. R. Brown,
295. L. Littlefield et al., Mut. Res. 81 (1981) 377. Biochem. Biophys. Res. Commun. 110 (1983)
296. J. Mac Donald et al., Ann. Intern. Med. 93 399.
(1980) 533. 324. E. D. Kharasch, R. F. Novak, Biochem.
297. Drugs Today 20 (1984) 489; 21 (1985) 420. Biophys. Res. Commun. 108 (1982) 1346.
298. Drugs of the Future 8 (1983) 402; 9 (1984) 325. S. S. Legha, Drugs Today 20 (1984) 12,
371; 10 (1985) 420. 629 – 638.
299. F. Arcamone et al., J. Med. Chem. 18 (1975)
703.
Cancer Chemotherapy 53
326. Drugs of the Future 5 (1980) 234; 6 (1981) 352. Drugs of the Future 5 (1980) 277; 8 (1983)
316; 7 (1982) 357; 8 (1983) 64; 9 (1984) 535.
380; 10 (1985) 429. 353. L. Steinherz et al., Cancer Treat. Rep. 66
327. American Cyanamid, USP 4197249, 1980 (1982) 483.
(F. E. Durr, K. C. Murdock). 354. J. Kolwas, Drugs Today 15 (1979) 200.
328. R. K.-Y. Zee-Cheng, C. C. Cheng, J. 355. G. A. Curt, N. J. Clendeninn, B. A. Chabner,
Med.-Chem. 22 (1979) 1024. Cancer Treat. Rep. 68 (1984) 87 – 99.
329. K. C. Murdock et al., J. Med. Chem. 22 (1979) 356. T. Tsuruo, H. Lida, M. Nojiri, Cancer Res. 43
9, 1024 – 1030. (1983) 2905.
330. F. I. Durr, R. E. Wallace, R. V. Citarella, 357. S. Waksman, H. B. Woodruff, Proc. Soc. Exp.
Cancer Treat Rev. 10 (1983) 3 – 11. Biol. Med. 45 (1940) 609 – 611.
331. R. E. Wallace, K. C. Murdock, R. B. Angier, 358. H. M. Sobell, S. C. Jam, T. D. Sakore et al.,
F. E. Durr, Cancer Res. 39 (1979) 1570 – 1574. Nat. New Biol. 231 (1971) 200.
359. F. Takusagawa et al.: 12th Congress of the
332. R. K. Johnson et al., Cancer Treat. Rep. 63
IUCR Associated Meeting on Molecular
(1979) 425 – 439.
Structure and Biologic Activity, Buffalo, New
333. D. D. Von Hoff, C. A. Coltuan, B. Forseth,
York, August 26 – 28, 1981, p. 55.
Cancer Res. 41 (1981) 1853 – 1855.
360. E. Reich, R. M. Franklin, A. J. Shatkin et al.,
334. B. M. Henderson et al., Cancer Treat. Rep. 66
Proc. Natl. Acad. Sci. USA 48 (1982) 1238.
(1982) 1139 – 1143. 361. H. S. Schwartz, E. Godoyien, R. Y. Ambaye,
335. B. M. Sparano, G. Gordon, C. Hall, M. J. Cancer Res. 287 (1968) 192.
Iatropoulos, J. F. Noble, Cancer Treat. Rep. 66 362. M. N. Goldstein, K. Hamm, E. Amrod, Science
(1982) 1145 – 1158. 151 (1966) 1555 – 1556.
336. K. C. Anderson et al., Cancer Treat. Rep. 67 363. H. Umezawa, K. Meada, T. Takeuchi et al., J.
(1983) 435 – 438. Antibiot. Ser. A. 19 (1966) 200 – 206.
337. F. C. Schell et al., Cancer Treat. Rep. 66 364. B. T. Sikic, Z. H. Siddik, T. E. Gram, Cancer
(1982) 1641 – 1643. Treat. Rep. 64 (1980) 659 – 667.
338. C. B. Pratt et al., Cancer Treat. Rep. 67 (1983) 365. T. Takita, Y. Muraoka, H. Umezawa:
85 – 88. “Bleomycin and Peplomycin,” in H. M.
339. D. V. Unverferth et al., Cancer Treat. Rep. 67 Pinedo, B. A. Chabner (eds.): Cancer
(1983) 343 – 350. Chemotherapy Annual 6, Elsevier, Amsterdam
340. M. S. Aapro, D. S. Alberts, Invest. New Drugs 1984, pp. 85 – 90.
2 (1984) 329 – 330. 366. A. Hagiwara et al., Anti-Cancer Drug Res. 2
341. S. A. Taylor, B. T. Tranum, D. D. Von Hoff, (1988) 319 – 324.
J. J. Constanzi, Invest. New Drugs 3 (1985) 367. S. Saito, Y. Umezawa, T. Yoshioka et al., J.
67 – 69. Antibiot. (Tokyo) 36 (1983) 92 – 95.
342. J. A. Elliott et al., Anti-Cancer Drug Res. 3 368. J. Fugimito, H. Higashi, G. Kosaki, Cancer
(1989) 4, 271 – 282. Res. 36 (1976) 2248 – 2251.
343. R. F. Marschke et al., Med. Pediat. Oncol. 16 369. M. Chien, A. P. Grollman, S. B. Horwitz,
(1988) 4, 269 – 270. Biochemistry 16 (1977) 3641 – 3645.
344. K. Hillier, Drugs of the Future 6 (1981) 12, 370. W. J. Caspay, C. Niziak, D. A. Lanzo et al.,
762; 7 (1982) 896. Mol. Pharmacol. 16 (1979) 256 – 260.
345. K. C. Murdock et al., J. Med. Chem. 25 (1982) 371. S. Biables, J. R. Warr, Genet. Res. 34 (1979)
505. 269 – 279.
346. American Cyanamid, EP 338372, 1989 (V. J. 372. M. Mayaki, T. Ono, S. Hori et al., Cancer Res.
Lee, K. C. Murdock) 35 (1975) 2015 – 2018.
347. American Cyanamid, US 4900838, 1990 373. S. Akiyama, M. Kuwano, J. Cell Physiol. 107
(K. C. Murdock) (1981) 147 – 153.
374. S. Akiyama, K. Ikezaki, H. Kunamochi et al.,
348. K. C. Murdock et al., J. Med. Chem. 36 (1993)
Biochem. Biophys. Res. Commun. 101 (1981)
15, 2098.
55 – 60.
349. US Dept. Of Health, USP 25157, 1981 (H.
375. W. E. G. Mueller, M. Geisort, R. K. Zahn et al.,
Dubicki, J. L. Parsons, F. W. Starks).
Eur. J. Cancer Clin. Oncol. 19 (1983)
350. B. F. Cain et al., J. Med. Chem. 18 (1975)
665 – 669.
1110.
376. P. G. Sorensen, M. Dorth, H. H. Hansen, Eur.
351. Annu. Drug Data Report (1981) 177.
J. Cancer Clin. Oncol. 19 (1983) 319 – 323.
54 Cancer Chemotherapy
377. R. K. Johnson, R. P. Hertzberg, Annu. Rep. 405. P. Mangeney et al., J. Org. Chem. 44 (1979)
Med. Chem. 25 (1990) 129. 3765 – 3768.
378. P. A. Aristoff et al., Invest. New Drugs 7 406. P. Maral, C. Bourut, E. Chenn, G. Mathe,
(1989) 364. Cancer Chemother. Pharmacol. 5 (1981) 3,
379. J. P. Mc Govren et al., Invest. New Drugs 7 197 – 199.
(1989) 448. 407. R. Maral, C. Bourut, E. Chenn, G. Mathe,
380. M. A. Mitchell, P. D. Johnson, M. G. Williams, Cancer Lett. 22 (1984) 49 – 54.
P. A. Aristoff, J. Am. Chem. Soc. 11 (1989) 408. G. Mathe, P. Reizenstein, Cancer Lett. 27
6428. (1985) 285 – 293.
381. K. Goni et al., Jpn. J. Cancer Res. 83 (1992) 409. M. Besenval et al., Semin. Oncol. 16 (1989) 2,
113. 37.
382. D. L. Boger, M. S. S. Palanki, J. Am. Chem. 410. A. Depierre et al., Semin. Oncol. 16 (1989) 2,
Soc. 114 (1992) 9318. 26.
383. M. Fontana et al., Anti-Cancer Drug Res. 7 411. J. B. Sorensen, Drugs 44 (1992) 60.
(1992) 131. 412. P. B. Schiff, A. S. Kende, S. B. Horwitz,
384. D. Volpe et al., Invest. New Drugs 10 (1992) Biochem. Biophys. Res. Commun. 85 (1978)
255. 737 – 740.
385. N. Zein, M. Poncin, R. Nilakantan, G. A. 413. R. A. Bender, B. A. Chabner: “Tubulin
Ellestad, Science 244 (1989) 697. Binding Agents: Epipodophyllotoxin,” in B. A.
386. B. H. Long et al., Proc. Natl. Acad. Sci. USA Chabner (ed.): Pharmacologic Principles of
86 (1989) 2. Cancer Treatment, W. B. Saunders,
387. K. C. Nicolaou, Science 256 (1992) 1172. Philadelphia 1982, pp. 263 – 266.
388. K. C. Nicolaou, W. M. Dai, J. Am. Chem. Soc. 414. J. D. Loike, S. B. Horwitz, Biochemistry 15
114 (1992) 8908. (1976) 5435 – 5438.
389. W. A. Bleyer, S. A. Frisby, V. T. Oliverio, 415. D. K. Kalwinsky, A. T. Look, J. Ducore, A.
Biochem. Pharmacol. 24 (1979) 633 – 639. Fridland, Cancer Res. 43 (1983) 1592 – 1596.
390. R. A. Bender, W. D. Kornreich, Proc. Am. 416. A. J. Wozniak, W. E. Ross, Cancer Res. 43
Assoc. Cancer Res. 22 (1981) 227. (1983) 130 – 135.
391. H. Mujagic, S. S. Chen, R. Geist et al., Cancer 417. A. J. Wozniak, B. S. Glisson, K. R. Hande,
Res. 43 (1983) 3591 – 3597. W. E. Ross, Cancer Res. 44 (1984) 626 – 629.
392. D. V. Jackson, R. A. Bender, Cancer Res. 39 418. B. H. Long, A. Minocha, Proc. Am. Assoc.
(1979) 4341 – 4349. Cancer Res. 24 (1983) 321.
393. R. Schrek, Am. J. Clin. Path. 62 (1974) 1 – 7. 419. M. E. Wall, M. C. Wani, C. E. Cook, K. H.
394. R. A. Bender, W. A. Bleyer, S. A. Frisby et al., Palmer, J. Am. Chem. Soc. 88 (1966) 3888.
Cancer Res. 35 (1975) 1305 – 1308. 420. J.-C. Cai, C. R. Hutchinson, Chem. Heterocyl.
395. R. F. Zager, S. A. Frisby, V. T. Oliverio, Cancer Compd. 25 (1983) 753.
Res. 33 (1973) 1670 – 1676. 421. Yakult Honsha, EP 137145, 1985 (T. Miyasaka
396. I. S. Johnson, H. F. Wright, G. H. Svoboda et et al.).
al., Cancer Res. 20 (1980) 1016 – 1022. 422. Smith Kline Beecham Corp., EP 321122, (J. C.
397. G. A. Curt, B. D. Bailey, H. Mujagic et al.,
Boehm, S. M. Hecht, K. G. Holden, R. K.
Proc. Am. Assoc. Cancer Res. 25 (1984) 337.
Johnson, W. D. Kingsbury).
398. W. T. Beck, M. C. Cirtain, J. L. Lefko, Cancer
423. W. D. Kingsbury et al., J. Med. Chem. 34
Treat. Rep. 67 (1983) 875 – 879.
(1991) 98 – 107.
399. R. B. Sklaroff, D. Straus, C. Young, Cancer
424. N. Osheroff, Pharmacol. Ther. 41 (1989)
Treat. Rep. 63 (1979) 793 – 794.
223 – 41.
400. G. Mathe, J. L. Misset, F. deVassal et al.,
425. L. Liu, J. Wang, Proc. Natl. Acad. Sci. USA 84
Cancer Treat. Rep. 62 (1978) 805.
(1987) 7024 – 7027.
401. M. Bayssas, J. Gouveia, F. deVassal et al.,
426. A. J. Ryan, S. Squires, H. L. Strutt, R. T.
Cancer Res. 74 (1980) 91 – 99.
Johnson, Nucl. Acid Res. 19 (1991) 12,
402. R. Z. Andriamialisoa, N. Langlois, Y. Langlois,
3295 – 3300.
P. Potier, Tetrahedron 36 (1980) 3053 – 3060.
427. M. N. Serradel, J. Castañer, R. M. Castañer,
403. M. N. Serradel, J. Castañer, Drugs of the
Drugs of the Future 12 (1987) 3, 207.
Future 11 (1986) 7, 575 – 577.
428. C. H. Spiridonis, Drugs of the Future 20
404. M. R. Paintrand, I. Pignot, J. Electron.
(1995) 5, 483 – 489.
Microsc. 32 (1983) 2, 115 – 124.
Cancer Chemotherapy 55
534. S. Kallio et al., Cancer Chemother. 563. P. V. Plourde et al., Breast Cancer Res. Treat.
Pharmacol. 17 (1986) 103 – 108. 30 (1994) 1, 103 – 111.
535. L. Kangas et al., Cancer Chemother. 564. K. Schieweck, A. S. Bhatnagar, A. Matter,
Pharmacol. 17 (1986) 109 – 113. Cancer Res. 48 (1988) 834.
536. V. C. Jordan, B. Gosden, Mol. Cell. 565. K. Schieweck et al., Proc. Am. Assoc. Cancer
Endocrinol. 27 (1982) 27, 921. Res. 29 (1988) Abst. 968.
537. S. P. Robinson et al., Eur. J. Cancer Clin. 566. M. Dowsett et al., Breast Cancer Res. Treat.
Oncol. 24 (1988) 12, 1817. 27 (1993) 1 – 2, Abst. 87.
538. R. Valavaara et al., Eur. J. Cancer Clin. Oncol. 567. P. V. Plourde et al., Proc. Am. Soc. Clin. Oncol.
24 (1988) 4, 785. 12 (1993) Abst. 165.
539. S. R. Ebbs et al., Lancet II (1987) 621. 568. C. Huggins, C. V. Hodges, Cancer Res. 1
540. R. F. Kauffmann et al., J. Pharmacol. Exp. (1941) 293.
Ther. 280 (1997) 146 – 153. 569. J. A. Smith, J. Urol. 137 (1987) 1 – 10.
541. M. A. Ferreira, M. M. Caramona, L. M. 570. J. Trachtenberg, J. Urol. 132 (1984) 61.
Celeste, Proc. Br. Pharmacol. Soc. (1995) 571. G. H. Rasmusson et al., J. Med. Chem. 27
Abs. 267 P (1984) 1690 – 1701.
542. M. Dukes, Pharm. J. 257 (1996) 176. 572. M. R. Robinson, B. S. Thomas, Brit. Med. J. 4
543. A. Hoshi, Drugs of the Future 16 (1991) 3, (1971) 391 – 394.
217. 573. F. Neumann, J. Steroid Biochem. 19 (1983)
544. Annu. Drug Data Report 15 (1993) 4, 378. 391 – 402.
545. A. Brodie, J. Steroid Biochem. Mol. Biol. 40 574. B. J. Furr, Clin. Oncol. 2 (1988) 581 – 590.
(1991) 1 – 3, 255. 575. H. Tucker, Drugs of the Future 15 (1990) 3,
546. S. A. Wells et al., Cancer Res. Suppl. 42 255 – 265.
(1982) 3454. 576. Drugs Today 20 (1984) 296.
547. R. D. Burnett, D. N. Kirk, J. Chem. Soc. Perkin 577. Drugs of the Future 1 (1976) 108; 8 (1983)
Trans. I (1973) 1830. 270.
548. R. C. Coombes, Eur. J. Cancer 28A (1992) 578. R. O. Neri, E. A. Peets, J. Steroid Biochem. 6
12, 1941. (1975) 815 – 819.
549. D. Cunningham et al., Brit. J. Cancer 55 579. W. I. P. Mainwaring, F. R. Mangan, P. A.
(1987) 331. Feherty, M. Freifeld, Med. Cell. Endocrinol. 1
550. J. H. Davis et al., Brit. J. Cancer 66 (1992) 1, (1974) 113 – 128.
139. 580. S. Liao, D. K. Howell, T. M. Chang,
551. M. Dowsett et al., Cancer Res. 49 (1989) Endocrinology 94 (1974) 1205 – 1209.
1306. 581. B. Katchen, S. Buxbaum, J. Clin. Endocrinol.
552. M. Dowsett et al., Eur. J. Cancer 28 (1992) Metab. 41 (1975) 373 – 379.
2 – 3, 415. 582. J. Geller et al., Prostate 2 (1981) 309 – 314.
553. R. C. Stein et al., Cancer Chemother. 583. E. A. Peets, M. F. Henson, R. O. Neri,
Pharmacol. 26 (1990) 75. Endocrinology 94 (1974) 532 – 540.
554. ICI, US 4935437, 1990 (P. N. Edwards, M. S. 584. R. Neri, N. Kassem, Prog. Cancer Res. Ther.
Lange). 31 (1984) 507 – 518.
555. Ciba Geigy AG, US 4617307, 1986 (L. J. 585. F. Labrie et al., Oncol. 2 (1988) 597 – 619.
Browne). 586. F. Labrie, A. Dupont, M. Ciguere, Brit. J.
556. J. Prous, Drugs of the Future 19 (1994) 4, Urol. 61 (1988) 341 – 346.
335 – 337. 587. A. Belanger, A. Dupont, F. Labrie, J. Clin.
557. J. Castañer, Annu. Drug Data Rep. 13 (1991) Endocrinol. Metab. 59 (1984) 422 – 426.
8, 716. 588. J.-P. Raynaud, C. Bonne et al., Prostate 5
558. A. B. Foster et al., J. Med. Chem. 28 (1985) (1984) 299 – 311.
200. 589. T. Ojasoo, Drugs of the Future 12 (1987)
559. J. T. Pento, Drugs of the Future 14 (1989) 9, 763 – 770.
843 – 845. 590. L. Proulx, F. Labrie, Prostate 5 (1984)
560. J. Prous et al., Drugs of the Future 20 (1995) Abst. 429.
1, 30 – 32. 591. F. Labrie, A. Dupont, A. Belanger, Prostate 4
561. R. E. Steele et al., Steroids 50 (1987) 147. (1983) 579.
562. M. Dukes et al., Proc. Am. Assoc. Cancer Res. 592. C. Harnois, A. Dupont, F. Labrie, Brit. J.
33 (1992) Abst. 1677. Opthalmol. 70 (1986) 471 – 473.
58 Cancer Chemotherapy
593. H. Tucker, G. J. Chesterson, J. Med. Chem. 31 618. B. Kutscher et al., Angew. Chem. Int. Ed. Engl.
(1988) 885 – 887. 36 (1997) 2148 – 2161.
594. H. Tucker, J. W. Crook, G. J. Chesterson, J. 619. R. L. Barbieri, Trends Endocrinol. Metab. 3
Med. Chem. 31 (1988) 954 – 959. (1992) 30 – 34.
595. B. J. A. Furr et al., J. Endocrinol. 113 (1987) 620. R. Schmidt, K. Sundaram, R. B. Thau, C. W.
R7 – 9. Badrin, Contraception 29 (1984) 283 – 289.
596. S. N. Freeman, W. I. P. Mainwaring, B. J. A. 621. S. Bajusz et al., Int. J. Pept. Protein Res. 32
Furr, J. Endocrinol. (1986) Suppl. III, 155. (1988) 425 – 435.
597. C. J. Tyrell, Prostate (Suppl. 4) 20 (1992) 97. 622. J. Pinski et al., Int. J. Pept. Protein Res. 45
598. G. Wilding et al., Proc. Am. Assoc. Clin. (1995) 410 – 417.
Oncol. 10 (1991) Abst. 593. 623. A. Kleemann et al., Proc. Akabori Conf. Ger.
599. A. V. Schally, S. M. McCann, Fertil. Steril. 64 Jpn. Symp. Pept. Chem. 4th 1991, 96 – 101;
(1995) 452 – 453. b) F. R. Kunz, T. Müller, K. Drauz, Proc.
600. A. V. Schally, A. Arimura, A. J. Kastin,
Akabori. Conf. Ger. Jpn. Symp. Pept. Chem.
Science 173 (1971) 1036 – 1038.
5th 1994, 15 – 16.
601. H. Matsuo, Y. Baba, M. Nair, A. Arimura,
624. A. Ljugquist et al., Proc. Natl. Acad. Sci. USA
A. V. Schally, Biochem. Biophys. Res.
85 (1988) 8236 – 8240.
Commun. 43 (1971) 1334 – 1339.
625. J. Leal et al., Drugs of the Future 16 (1991)
602. K. Amoss et al., Biochem. Biophys. Res.
529 – 537.
Commun. 44 (1971) 205 – 210.
603. Y. Baba, H. Matsuo, A. V. Schally, Biochem. 626. A. Janecka, T. Janecki, C. Bowers, K. Folkers,
Biophys. Res. Commun. 44 (1971) 459 – 463. J. Med. Chem. 37 (1994) 2238 – 2241.
604. G. Emons, A. V. Schally, Hum. Reprod. 9 627. A. Janecka et al., Med. Chem. Res. 1 (1991)
(1994) 1364 – 1370. 306 – 311.
605. P. M. Conn, W. Crowley, New Engl. J. Med. 628. A. Janecka, T. Janecki, C. Bowers, K. Folkers,
324 (1991) 93 – 103. Int. J. Pept. Protein Res. 44 (1994) 19 – 23.
606. G. F. Weinbauer, E. Nieschlag in K. Höffgen 629. P. Theobald et al., J. Med. Chem. 34 (1991)
(ed.): Peptides in Oncology Springer, 2395 – 2402.
Heidelberg, 1992, pp. 113 – 136. 630. J. Rivier et al., J. Med. Chem. 35 (1992)
607. A. S. Dutta, Drugs of the Future 13 (1988) 4270 – 4278.
43 – 57. 631. J. E. Rivier et al., J. Med. Chem. 38 (1995)
608. M. Filicori, C. Flamingi, Drugs 35 (1988) 2649 – 2662.
63 – 82. 632. K. Stoeckemann, J. Sandow, J. Cancer Res.
609. J. J. Nestor, B. H. Vickery, Annu. Rep. Med. Clin. Oncol. 119 (1993) 457 – 462.
Chem. 23 (1988) 211 – 220. 633. R. Deghenghi, F. Boutignon, P. Wüthrich V.
610. M. T. Goulet, Annu. Rep. Med. Chem. 30 Lenaerts, Biomed. Pharmacother. 47 (1993)
(1995) 169 – 178. 107 – 110.
611. T. Reissmann et al., Hum. Reprod. 20 (1995) 634. J. Nester, et al., J. Med. Chem. 35 (1992)
1974 – 1981. 3942 – 3948.
612. M. J. Karten in W. F. Crowley, P. M. Conn 635. J. Mulzer in E. Ottow, U. Schöllkopf, B. G.
(eds.): Modes of Action of GnRH and GnRH Schulz (eds.): Stereoselective Synthesis ,
Analogs, Springer, Heidelberg 1992, Springer, Heidelberg, 1994, pp. 37 – 61.
pp. 277 – 297. 636. J. Mulzer et al., Angew. Chem. 106 (1994)
613. G. Flouret et al., Pept. Sci. 1 (1995) 89 – 105. 1813 – 1815; Angew. Chem. Int. Ed. Engl. 33
614. P. M. Conn, W. F. Crowley, Annu. Rev. Med. (1994) 1737 – 1739.
45 (1994) 391. 637. F. Haviv et al., J. Med. Chem. 32 (1989)
615. A. V. Schally in J. F. Holand et al. (eds): 2340 – 2344.
Cancer Medicine 3rd ed., Lee & Febiger, 638. F. Haviv et al., J. Med. Chem. 36 (1993)
Philadelphia, PA 1993, pp. 827 – 840. 928 – 933.
616. A. V. Schally in P. Belfort, J. Pinotti, T. K.
639. Abbott Laboratories, PCT/US 95/02410, 1995
Eskes (eds.): Advances in Gynecology and
(F. Haviv).
Obstetrics Vol. 6, Parthenon, Cornforth 1989,
640. Abbott Laboratories, WO 95/04540, 1995 (F.
pp. 3 – 22; b) Scrip 22, 1995, 2066.
Haviv).
617. A. Friedrich, G. Jaeger, K. Radscheit, R.
641. Abbott Laboratories, WO 94/14841, 1994 (F.
Uhmann, Pept. Proc. Eur. Pept. Symp. 22nd
Haviv).
1992/1993, 47 – 49.
Cancer Chemotherapy 59
642. Abbott Laboratories, WO 94/13313, 1994 (J. 662. D. Rea et al., Proc. 7th NCI-EORTC Symp. On
Greer). New Drugs in Cancer Ther. Amsterdam
643. Tap Pharmaceuticals, US-A 5300492, 1994 (F. (1992), p. 62.
Haviv). 663. D. Ingber et al., Nature 348 (1990) 555.
644. F. Haviv et al., J. Med. Chem. 37 (1994) 664. T. Ozsuka et al., J. Antibiotics 45 (1992) 348.
701 – 707. 665. Y. Reiss et al., Cell 62 (1990) 81 – 88.
645. T. Beckers, K. Marheineke, H. Reiländer, P. 666. S. Omura, D. van der Pyl, Cell 46 (1993) 222.
Hilgard, Eur. J. Biochem. 231 (1995) 667. S. M. Sebti, A. D. Hamilton, Drug Discovery
535 – 543. Technol. 3 (1998) 26 – 33.
646. R. P. Millar, C. A. Flanagan, R. C. Milton, J. A. 668. A. Levitski, A. Gazit, Science 267 (1995)
King, J. Biol. Chem. 264 (1989) 1782 – 1788.
21007 – 21013. 669. A. Levitski, Eur. J. Biochem. 226 (1994)
647. G. A. McPherson, J. Pharmacol. Methods 14 1 – 13.
(1985) 213 – 228. 670. R. T. Abraham, M. Aquarone, A. Anderson,
648. J. Trachtenberg, A. Pont, Lancet 2 (1984) Biol. Cell. 83 (1995) 105.
433 – 435. 671. D. J. Hanahan, Ann. Rev. Biochem. 55 (1986)
649. S. Bhasin et al., Endocrinology 118 (1986) 483 – 509.
1229 – 1232. 672. W. E. Berdel, Onkologie 13 (1990) 245 – 250.
650. B. De et al., J. Med. Chem. 32 (1989) 673. W. E. Berdel et al., Anticancer Res. 1 (1981)
2036 – 2038. 345 – 352.
651. Abbott Laboratories, US-A 4992421, 1991 (B.
674. G. Rodriguez et al., Proc. Am. Assoc. Cancer
De).
Res. 33 (1992) 262.
652. McNeillab Inc., US-A 4678784, 1987 (C. Y.
675. C. Unger, H. Eibl, Lipids 26 (1991) 1412.
Ho).
676. P. Hilgard, J. Engel, Drugs Today Suppl. B.,
653. Takeda Chemical Industries, WO 95/28405,
(1994) 30.
1995 (S. Furuya).
654. Takeda Chemical Industries, WO 96/34012A1, 677. P. Hilgard et al., Cancer Chemother.
1996 (C. Kitada). Pharmacol. 32 (1993) 90 – 95.
655. Merck, WO 97/21435, 1997 (M. Goulet). 678. P. Hilgard, J. Stekar, C. Unger, Proc. Annu.
656. Takeda Chemical Industries, WO 97/14697, Meet. Am. Assoc. Cancer Res. 31 (1990)
1997 (S. Furuya). A2457.
657. C. Unger, Drugs of the Future 22 (1997) 12, 679. J. Engel et al., Drugs of the Future 13 (1988)
1337 – 1345. 10, 948 – 951.
658. G. Powis, Pharmacol. Ther. 62 (1994) 57 – 95. 680. C. Geilen et al., Eur. J. Cancer 27 (1991) 12,
659. W. J. Fantl, D. E. Johnson, L. T. Williams, 1650 – 1653.
Annu. rev. Biochem. 62 (1993) 453 – 481. 681. R. Hass et al., Cancer Res. 52 (1992)
660. F. McCormick, Nature 363 (1993) 15 – 16. 1445 – 1450.
661. C. A. Lang-Carter et al., Science 260 (1993) 682. R. Becher et al., Onkologie 16 (1993) 1, 11.
315 – 319.