Cu Complexes-11

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/19404833

In Vivo Antitumor Activity and in Vitro Cytotoxic Properties of Bis[1,2-


bis(diphenylphosphino)ethane]gold(I) Chloride

Article in Cancer Research · December 1986


Source: PubMed

CITATIONS READS

183 223

10 authors, including:

Christopher Mirabelli Michael R Mattern


Leap Therapeutics Progenra Inc.
103 PUBLICATIONS 6,013 CITATIONS 118 PUBLICATIONS 8,481 CITATIONS

SEE PROFILE SEE PROFILE

Stanley Crooke
Ionis Pharmaceuticals
572 PUBLICATIONS 27,753 CITATIONS

SEE PROFILE

All content following this page was uploaded by Michael R Mattern on 15 June 2015.

The user has requested enhancement of the downloaded file.


In Vivo Antitumor Activity and in Vitro Cytotoxic Properties of
Bis[1,2-bis(diphenylphosphino)ethane]gold(I) Chloride
Susan J. Berners-Price, Christopher K. Mirabelli, Randall K. Johnson, et al.

Cancer Res 1986;46:5486-5493. Published online November 1, 1986.

Updated Version Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/46/11/5486

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, contact the AACR Publications
Department at permissions@aacr.org.

Downloaded from cancerres.aacrjournals.org on July 10, 2011


Copyright © 1986 American Association for Cancer Research
[CANCER RESEARCH 46, 5486-5493, November 1986]

In Vivo Antitumor Activity and in Vitro Cytotoxic Properties of


Bis[l ,2-bis(diphenylphosphino)ethane]gold(I) Chloride
Susan J. Berners-Price, Christopher K. Mirabelli,1 Randall K. Johnson, Michael R. Mattern, Francis L. McCabe,
Leo F. Faucette, Chiù-MeiSung, Shau-Ming Mong, Peter J. Sadler, and Stanley T. Crooke
Department of Chemistry, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, United Kingdom [S. J. B-P., P. J. S.J, and Department of
Molecular Pharmacology, Smith Kline and French Laboratories, Swedeland, Pennsylvania 19479 [C-M. S., R. K. J., M. R. M., F. L. M., L. F. F.,
S-M. M., C. K. M., S. T. CJ

ABSTRACT other tumor models (6).2 In addition, several of the phosphine


ligands (e.g., DPPE3) of these compounds demonstrated anti-
We have previously reported the cytotoxicity and antitumor activity of
bis(diphenylphosphino)ethane (DPPE) and a variety of its transition
tumor activity but were significantly less potent than the
gold(I) and gold(III) complexes. In the series [(AuCl)2(Ph2P-
metal complexes. During studies of the chemistry of a gold complex of
this group |( Vu( !).(' 'I'l'l )|. it was observed that this complex readily (CH2)„PPh2)],antitumor activity was maximized when n = 2
underwent ring closure on reaction with DPPE to form the tetrahedral or 3. The complex with a cw-ethylene bridging alkene also was
complex |Ati(I>PI'K),|*. Various counterion forms (<•.#., ( 'I ) of this cation very active, whereas the /ra/w-ethylene complex was inactive.
were isolated and were found to exhibit a remarkably high stability in Since these active ligands would be able to form stable 5- or 6-
solution. Evaluation of |Au(DPPE)JCl in mice bearing i.p. P388 leukemia membered rings by chelation, the possibility that a ring-closed
demonstrated that the compound produced an average of 87% increase form could be an intermediate in the metabolism of these
in life span at its maximally tolerated dose (2-3 ¿imol/kg/dayfor 5 days).
complexes was considered. However, gold(I) complexes gener
Activity was also seen in i.p. M5076 reticulum cell sarcoma (60% increase ally exhibit linear 2-coordinatimi, and 3- or 4-coordinate com
in life span) and s.c. mammary adenocarcinoma 16/c. Modest activity plexes are rare and often unattainable (7-9). They are often
was evident in i.p. B16 melanoma and 1,1210 leukemia. A subline of
P388 leukemia resistant to cisplatin was not cross-resistant to unstable species. During our studies of the chemistry of
|Au(Dl'l'l:),|( I. In addition, combination therapy of | \u(I)PPK)..|( I and [(AuCl)2(DPPE)], we observed, unexpectedly, that this complex
cisplatin against i.p. P388 demonstrated an advantage over single-agent readily underwent ring closure on reaction with DPPE, to form
therapy.
the tetrahedral complex [Au(DPPE)2]+ (10) (Fig. 1). Complexes
in vitro studies of (Au(DPPE)2)Cl showed that the compound: (a) is containing this cation were isolated and found to exhibit a
o tutuvii-to tumor cell lines; (b) is only minimally inhibited in its cytotoxic remarkably high stability.
activity by the presence of serum; (c) produces DNA protein cross-links This report describes our evaluation of the antitumor prop
and DNA strand breaks in cells; and (</)inhibits macromolecular synthe erties, cellular pharmacology, and chemical reactivity of
sis with a preferential inhibitory effect on protein synthesis relative to [Au(DPPE)2]Cl. The results of these studies are discussed in
DNA and RNA synthesis. "P nuclear magnetic resonance spectroscopy
the context of previously described data for other classes of
indicated that the compound is stable in the presence of serum proteins,
thiols, or disulfides and that it reacts with Cu(II) resulting in the forma gold(I) phosphines in an attempt to correlate their respective
tion of a Cu(I)DPPE complex. The results of these in vivo and in vitro pharmacological actions with their chemical properties.
experiments suggest that the contrasting pharmacological profile of
|Au( 1>1TK).|( Ãw̄ith respect to other gold(I) phosphine complexes may
MATERIALS AND METHODS
be related to both the kinetic stability of the complex and its stability in
the presence of thiols. Materials. Na[AuCU] was purchased from Johnson Matthey, Ltd.,
and DPPE was from Strem Chemicals. Thiodiglycol and glutathione
INTRODUCTION (reduced and oxidized) were purchased from Sigma and dried bovine
serum (Wellcomtrol I) was purchased from Wellcome.
Evidence is emerging to show that many gold(I) phosphine NMR. Proton-decoupled 3IP NMR (|'H| "P-NMR) spectra at 24.2
complexes are potent cytotoxic agents (1). The antiarthritic MHz were measured in 10-mm tubes using a JEOL FX60 spectrometer.
agent, auranofin [2,3,4,6-tetra-0-acetyl-l-thio-/3-D-glucopyra- When nondeuterated solvents were used, the sample was contained in
an 8 •mm tube inside a 10-mm tube containing D2O for a '!) lock.
nosato-S-triethylphosphine gold(I)] (Fig. 1), has been shown to
Eighty-five % H3PO4 in D2O (85:15, v/v) was used as an external shift
inhibit human lymphocyte responsiveness both in vivo and in
reference.
vitro (2, 3) and has shown in vivo antitumor activity against i.p.
Preparation of |Au(DPPE)j|Cl. Na[AuCU] 0.5H2O (0.5 g, 1.35 mmol)
P388 leukemia (4). Extensive evaluation of the in vivo antitumor was reduced by thiodiglycol (0.33 g, 2.70 mmol) in aqueous acetone
activity of auranofin in 15 tumor models revealed that the drug (2.5:1, 7 ml). The solution was cooled to 0-5°Cand added dropwise to
was active only when administered i.p. against i.p. P388 leu a solution of DPPE (1.07 g, 2.70 mmol) in acetone (about 30 ml). A
kemia (5). clear colorless solution resulted; this was stirred for 30 min; then the
In an attempt to identify a gold-containing complex with a solvent was concentrated to 10 ml by evaporation at room temperature.
spectrum of in vivo antitumor activity greater than that of The product was obtained as a white to cream-colored solid by addition
auranofin, we have synthetized a variety of gold complexes and of 11•<
) (about 10 ml) and was recrystallized from aqueous methanol
evaluated them in vitro and in in vivo tumor systems (1). A and dried in a vacuum (1.1 g, 79%). Analysis: (CszHsoAuClOP^ C, H,
series of gold(I) and gold(III) complexes incorporating biden- CI, P.
The complex was characterized by NMR and UV spectroscopy. The
tate phosphine ligands were found to be reproducibly more
active than auranofin in P388 leukemia and active as well in 2C. K. Mirabelli, D. T. Hill, L. F. Faucette, F. L. McCabe, G. R. Girard, B.
M. Sutton, and R. K. Johnson. In vivo antitumor activity of analogs of
Received 5/23/86; revised 7/31/86; accepted 8/5/86. bis(diphenylphosphino)ethane and their gold(I) coordination complexes, submit
The costs of publication of this article were defrayed in part by the payment ted for publication.
of page charges. This article must therefore be hereby marked advertisement in ' The abbreviations used are: DPPE, l,2-bis(diphenylphosphine)ethane; NMR,
accordance with 18 U.S.C. Section 1734 solely to indicate this fact. nuclear magnetic resonance; GSH, reduced glutathione; GSSG, oxidized gluta
1To whom requests for reprints should be addressed, at Birkbeck College, thione; DMSO, dimethyl sulfoxide; TCA, trichloroacetic acid; MTD, maximally
University of London, Malet Street, London, WCIE 7HX, United Kingdom. tolerated dose; ILS, increase in life span.
5486
Downloaded from cancerres.aacrjournals.org on July 10, 2011
Copyright © 1986 American Association for Cancer Research
ANTITUMOR ACTIVITY OF BIS(DPPE)GOLD(I) CHLORIDE

CDC13. The ('H) 3IP NMR spectra of both the supernatant and the
CHÃ--0-C-CHj precipitate were recorded.
-0 Evaluation in i.p. P388 Leukemia. P388 leukemia cells (10") main
tained by a serial transplantation in syngeneic DBA/2 mice were
S-Au-P(CzHg)s
inoculated i.p. in C57BL/6 x DBA/2 F, (hereafter called B6D2F,)
AURANOFIN mice. Twenty-four h later, if the tumor inoculum proved to be free of
H,C-C-0\ 0
bacterial contamination (as determined by 24-h incubation in thiogly-
0 \0-C-CH,
colate broth), animals were randomized into groups of 6 and housed in
0-C-CHS shoe box cages. [Au(DPPE)2]Cl was dissolved in 95% ethanol. Water
was added in sufficient quantity such that the desired dose was delivered
in 0.5 ml. The final concentration of ethanol was «10%.Formulation
were prepared immediately prior to injection. The drug was adminis
tered i.p. on Days 1 through 5 (i.e., treatment was initiated 24 h after
[(AuCI)2(dppe)] tumor inoculation). Each experiment includes 3 groups of 6 animals as
untreated controls and animals treated with a postive control, cisplatin
AuCI AuCI (Sigma Chemical Co., St. Louis, MO), at 2 dose levels.
Other in Vivo Tumor Models. Other murine tumor models were
maintained by serial transplantation in syngeneic mice and were im
planted i.p. and/or s.c. for evaluation of drug activity. Tumor models
included: M5076 reticulum cell sarcoma, B16 melanoma, and Lewis
lung carcinoma (maintained in C57BL/6 mice and evaluated in B6D2F,
mice); Madison 109 lung carcinoma and colon carcinoma 26 (main
tained and evaluated in BALB/c mice); mammary adenocarcinomas
16/c and 13/c (maintained and evaluated in C3H mice); LI210 leuke
[Au(dppe)2]CI mia and a cisplatin-resistant subline of P388 leukemia (maintained in
DBA/2 mice and evaluated in B6D2F, mice). All tumors were obtained
from the National Cancer Institute tumor bank at Frederick Cancer
Research Center, Frederick, MD.
For evaluation in the slower growing solid tumor models,
Fig. I. Structure of gold coordination complexes described in this paper. [Au(DPPE)2]Cl was administered i.p. daily for 10 days beginning 1 day
after tumor implantation. The drug was administered in each tumor
molar conductance of the complex in CH3CN is consistent with that of model at 5 logarithmically spaced dosage levels which encompassed the
a 1:1 electrolyte. In the 'H NMR spectrum, the phenyl protons are maximally tolerated dose. Activity was assessed by inhibition of tumor
slightly shielded with respect to free DPPE, and this presumably arises growth at the site of implant for s.c. tumors and prolongation of median
as a result of ring-current effects between adjacent aromatic rings in life span for i.p. tumors. Tumor volume was determined when tumors
the tetrahedral complex. The X-ray crystal structure of the chloride in untreated control mice averaged about 1000 mm3 (2 to 3 weeks after
complex has recently been described by Bates and Waters (11). We inoculation). Tumor volume was estimated by measurement of perpen
have previously reported the X-ray crystal structure of the SbF6~ dicular diameters with vernier calipers using the equation of 0.5 x
complex (10), and both have identical 31P NMR chemical shifts in length x (width)2.
CDCI3. Cell Culture Techniques. B16 melanoma cells were maintained as
[Au(DPPE)2]Cl is a white solid. It is stable to light and heat and is monolayer cultures in minimal essential medium and P388 and 1.1210
soluble in a range of solvents, including DMSO, methanol, ethanol, leukemia cells were maintained in RPMI 1640 as suspension cultures.
CHC13, and acetone. Its solubility in H2O is extremely low (<1 ng/ml). All cell lines were grown in medium supplemented with 10% calf serum,
The stability of [Au(DPPE)2]Cl in solution was monitored using 3IP penicillin (100 units/ml), and streptomycin (100 /¿g/ml)in a 5% ('<>.•-
humidified incubator at 37°C.All media and calf sera were purchased
NMR. No decomposition was observed in any solvent after 24 h in
solution, and the complex was stable for at least 4 days in metha- from Grand Island Biological Co., Grand Island, NY.
nol:saline(l:l, v/v). In Vitro Cytotoxicity Assays. Monolayer clonogenic assays were
Reaction of | \u(l)l'l'K):|( I with GSH, GSSG, Bovine Serum, and performed using asynchronous populations of B16 cells as described
CuSO4. Aliquots of a solution of [Au(DPPE)2]Cl in DMSO were added previously (5). The in vitro chemosensitivity of asynchronous popula
tions of P388 cells was determined in a soft agar colony-forming assay
to solutions containing 1, 5, 10 and 50 molar equivalents of GSH in
in 0.17% agarose as described by Metcalfe et al. (12).
D2O at pH 7 (meter reading) to give final gold concentrations of 10
min in D2O:DMSO (1:1, v/v). ('H) 31PNMR spectra were recorded at Inhibition of Macromolecular Synthesis. B16 cells were harvested and
transferred into 96-well, flat-bottomed microtiter plates at a cell con
l h and 8 days of reaction time. centration of 5 x IO4 cells/well in 10(1 //I of medium. Following
One-mi aliquots of GSSG in 0.1 M phosphate buffer (pH 7) were
overnight incubation to allow attachment of the cells, specified concen
added to 23 mM solutions of [Au(DPPE)2]Cl in methanol (1 ml) giving trations of the test compound and/or 25 n\ of [3H]thymidine (80 /xCi/
gold:GSSG ratios of 2:1, 1:1, and 1:5. ('H) 31P NMR spectra were ml), [3H]uridine (80 nCi/ml), or [3H]leucine (160 /zCi/ml) were added
recorded after 24 h and 7 days of reaction time. to the appropriate wells, and the plates were returned to the incubator
Lyophilized bovine serum was reconstituted with D2O. Fifty n\ of a for specified time periods (5 through 90 min). The medium was then
DMSO solution of [Au(DPPE)2]Cl (1.03 mg) were added with gentle aspirated, and each well was washed once with 200 >i\of 0.15 M NaCl.
shaking to 1 ml of the serum to give a final drug concentration of 1 Cells were lysed with 25 n\ of 0.2 M NaOH for 20 min at room
mM with 5% (v/v) DMSO present. The ('H) 3IP NMR spectrum was
temperature, followed by the addition of 100 *ilof 15% TCA. After 1 h
recorded after an average reaction time of 12 h. at 4"( '. the T< A precipitated material was transferred to glass micro-
CuSO4-5H2O (5.0 mg, 0.02 mmol) in D2O (0.5 ml) was added to a fiber Illters using a multisample cell harvester and 5% TCA. The filters
solution of |Au(1)1M'I ),|(1 (21.0 mg, 0.02 mmol) in methanol (1 ml). were dried and placed into scintillation vials with 2 ml of Econofluor
The solution became cloudy and after stirring for 30 min a thick white (New England Nuclear, Boston, MA), and the incorporation of 3H
precipitate had formed. This was removed by centrifugation, washed precursors into TCA-precipitable materials was measured in a Beckman
thoroughly with 11•(>,
and dried in a vacuum. A small amount of the LS 9800 counter (Beckman Instruments). Inhibition of precursor up
solid was digested in concentrated HNO3 and the [Au]:[Cu] ratio was take into macromolecules was measured in parallel with cells incubated
determined by atomic absorption. The remainder was dissolved in in the absence of drug.
5487
Downloaded from cancerres.aacrjournals.org on July 10, 2011
Copyright © 1986 American Association for Cancer Research
ANTITUMOR ACTIVITY OF BIS(DPPE)GOLD(I) CHLORIDE

Alkaline Elution Assay. The DNA in exponentially growing 1,1210 average of 87% ILS over 21 separate dose-response studies.
cells was radioactively labeled by incubation with [2-14C]-or [methyi-
•'H]thymidine(50 mCi/mmol and 73 Ci/mmol, respectively; New Eng This prolongation in life span at the maximally tolerated dose
land Nuclear) for 16-20 h at 37°C.14Clabeling was with 0.02 MCi/ml corresponds to a net cell kill of approximately 2 logs; i.e., at
and 3H labeling (internal standard cells) was with <0.1 »iCi/ml. the end of the 5-day course of treatment, the tumor cell burden
Unincorporated radiolabel was removed by centrifugation prior to
was reduced by 99% compared to the tumor cell burden at the
drug treatment or cell irradiation. Cells were incubated at 37°Cin drug- start of therapy.
free medium for at least 60 min before this medium was replaced with The influence of the route of administration of [Au(DPPE)2]-
37°Cmedium containing [Au(DPPE)2]Cl. Following the time period Cl on its activity against i.p. P388 leukemia was evaluated. The
allotted for drug exposure, cells were then centrifuged and washed with complex was less toxic when given i.V., s.c., or p.o. but was
phosphate-buffered saline at 0-4°C. In experiments which measured
inactive in this tumor system by these three routes of adminis
reversibility of drug effect, treated cells were centrifuged, washed, and tration (data not shown). [Au(DPPE)2]Cl was also inactive
resuspended in 37°Cdrug-free growth medium and incubated for 30
against systemic P388 leukemia (i.v. inoculated tumor) when
min. DNA single-strand breaks and DNA-protein cross-links were
the compound was administered either i.p. or i.v. (data not
measured by DNA filter elution assays as described by Kohn et al. ( 13).
Briefly, for measurement of DNA single-strand breaks 5 x IO5treated shown).
or control [l4C]thymidine-labeled cells were mixed with approximately The activity of [Au(DPPE)2]Cl against i.p. implanted M5076
equal numbers of }H-labeled cells that had been irradiated with 300 reticulum sarcoma is shown in Fig. 3. At a MTD of 1.9 ¿¿mol/
rads. The cells were deposited onto polycarbonate membrane filters (2- kg/day, [Au(DPPE)2]Cl administered i.p. daily for 10 days
¿impore diameter, Nucleopore Corp.) and lysed with 0.1 M glycine- produced 59% ILS. As in P388 leukemia, there was a dose-
0.025 M d¡M u!iuni EDTA-2% sodium dodecyl sulfate, pH 10, with or dependent prolongation of life span. [Au(DPPE)2]Cl showed
without proteinase K (0.5 mg/ml; Sigma). Elution was carried out with modest activity against B16 melanoma with an average of 39%
tetrapropylammonium hydroxide-EDTA-0.1% sodium dodecyl sulfate, ILS at the MTD in three dose-response studies (data not
pH 12.1, at 0.04 ml/min. Fractions were collected every 3 h for a total shown). [Au(DPPE)2]Cl also exhibited significant activity
of 18 h. Single-strand break frequency and percentage of break resealing
against the s.c. implanted tumor, mammary adenocarcinoma
were calculated as described previously (13). Results were expressed as
"rad equivalents" of DNA strand breakdage. The amount of "protein- 16/c, when administered either i.p. or i.v. on an intermittent
concealed" DNA strand breakage was determined in experiments in treatment schedule (Table 1). The compound was less toxic to
which duplicate drug-treated cultures were assayed, one with and the mice when given i.v., and the greater inhibition of tumor growth
other without proteinase K in the lysis solution. To measure DNA- by this route may be the result of the higher dose levels. The
protein cross-linking treated or control cells were 7-irradiated at ice compound also was evaluated in a series of other transplantable
temperature with 3000 rads and then deposited onto 2 nM polyvinyl in vivo murine tumor models. The resulting overall spectrum
chloride filters (Millipore Corp.) and lysed with a solution consisting of in vivo activity of [Au(DPPE)2]Cl is summarized in Table 2.
of 2 M NaCl, 0.2% Sarkosyl, and 0.04 M disodium EDTA, pH 10 (5
ml). DNA was eluted, following a wash with 0.04 M EDTA, pH 10,
with tetrapropylammonium hydroxide-EDTA, pH 12.1, at 0.04 ml/ 60
min. Fractions were collected as described above, and the DNA-protein
80
cross-linking frequency was calculated according to the procedure de
UJ
scribed by Kohn (13). The data were expressed in rad equivalents of 40-
DNA-protein cross-linking for comparison with the amount of DNA I 40
single-strand breakage, and the values obtained for untreated control m
t»
cells were subtracted from those obtained for the treated samples. U)
m
20
RESULTS
In Vivo Antitumor Activity. The activity of [Au(DPPE)2]Cl ¡
against ip P388 leukemia in mice is shown in Fig. 2. A MTD 048 095 191 382 7.64
of 2.9 jiinol/kg, administered i.p. daily for 5 days, produced an DOSE (>jmol/kg/doy, ip., Days I-IO)
Fig. 3. Dose-response curve for [Au(DPPE)j]CI in i.p. M5076 reticulum celi
24 sarcoma. Each point is the average of median survival times at a particular dose
in 3 experiments. Bars, SD.
3)
20 - R
801 Table 1 Activity of(Au(DPPE)JCl against mammary adenocarcinoma 16/c
implanted s.c.
UJ
16
60 â„¢
5 Optimal Mean tumor
4O o Route of dose volume* Inhibition
Compound administration" (fimol/kg) (mm3) (%) NP'
12
Experiment
1ControlCisplatin
tr ±99930
n
(ft i.p.[Au(DPPE)2]CI ±6496
m i.p.Experiment 14311±
co
o
g l 2ControlCisplatin
13 ±626097921001/246/84/70/238/8
i.p.208201187
O 036 072 14 29 57
DOSE (pmol/Kg/day, i p.Days 1-5) [Au(DPPE)2]Cl 6 430 ±291 61 0/8
Fig. 2. Activity of [Au(DPPE)2]Cl in mice bearing i.p. P388 leukemia. Tumor- 11 181 ±163 84 2/8
bearing mice were treated with drug on Days 1 and 5 with a single daily dose i.p. * All dosages were given every 4 days for 5 doses.
Each point is the average of median survival times at a particular dose in 21 *OnDay21,±SD.
experiments. Bars, SD. ' Proportion of mice without palpable tumors on Day 21.

5488
Downloaded from cancerres.aacrjournals.org on July 10, 2011
Copyright © 1986 American Association for Cancer Research
ANTITUMOR ACTIVITY OF BIS(DPPE)GOLD(I) CHLORIDE

Table 2 Spectrum of activity of [Au(DPPE)JCl in transplantable murine tumors Table 3 Combination chemotherapy of moderately advanced i.p. P3SS leukemia
cisplatin[Au(DPPE)2]Cl
with [Au(DPPE)JCl and
Response of tumor to
[Au(DPPE)2]CI (%) at following cisplatin doses
Tumor model Oimol/kg/day)»0510

i.p. tumors 0<mol/kg/day)°00.35


P388 leukemia
M5076 reticulum cell sarcoma
B16 melanoma 25 40 5560 95
LI210 leukemia 0.7 30 45 90
Colon carcinoma 26 1.4 3045e1.04-15
40 50 70 105
BI6 melanoma (FIO subline) 2.95.7IIS" 502.15 604.245 808.390 13516.7C

i.v. tumors " Based on median survival time of groups of 8 mice.


P388 leukemia * Administered i.p. on Days 3 through 7.
' Toxic dose.
s.c. tumors
Mammary adenocarcinoma 16/c
Mammary adenocarcinoma 13/c
ADJ-PC6 plasmacytoma ±
M5076 reticulum cell sarcoma ±
Colon carcinoma 26
Colon carcinoma 07/A
" ++, >50% ILS in i.p. tumors and >90% tumor growth inhibition in s.c.
tumor models; +, >30% ILS in i.p. tumors and >75% tumor growth inhibition
in s.c. tumor models; ±,unreproducible activity; -, <30% ILS in i.p. tumors and
<75% tumor growth inhibition in s.c. tumor models.

A. B.
5OCO \(-.\\\|
200

40i_£>>Co0
g 11i\\\\•H\\\\\
100

80
P388 PJ88
30|O

60

05«20in\-

40 \\
\\
20 \\
\\T0
1.25 2.5 5 10 20 038 075 1.5 3
[Au(dppe)2Ki
\ai
Cisplotin
>jmol/kg,QDx5 >imol/Kq,QDx5
Fig. 4. Dose-response curves for cisplatin (A) and [Au(DPPE)2]Cl (B) in i.p.
P388 and the i.p. cisplalin-resistant P388 tumors, till x 5, daily for 5 days.
i i
468 10
The in vivo antitumor activity of [Au(DPPE)2]Cl was also UM of [Au(dppe).]CI
evaluated in a subline of P388 leukemia which is resistant to
Fig. 5. Cytotoxic activity of [Au(DPPE)2]CI against B16 (<J) and P388 (H)
cisplatin. As shown in Fig. 4, this subline is refractory to cells as measured in the colony formation assays following a 2-h exposure to the
cisplatin when compared to the response of the parental tumor drug. Points, means of 3 assays; bars, SE.
line. However, [Au(DPPE)2]Cl was active against both the
parental and the cisplatin-resistant line of P388 producing produced 90% ILS. [Au(DPPE)2]Cl was less effective in these
similar increases in life span (85 and 77%, respectively) at the advanced tumor-bearing mice with 45% ILS at its MTD. The
MTD of 2.9 ixmol/kg. drugs could be combined at their respective maximally tolerated
The antitumor activity of the combination of [Au(DPPE)2]Cl doses with no lethality. There was a clear-cut advantage of this
with cisplatin in an in vivo tumor model was assessed. P388 combination over cisplatin alone, producing 135% increase in
leukemia was used because of its sensitivity to both classes of life span at a nontoxic dose. It was also noted that low-dose
drug. In order to provide more of a therapeutic challenge, combinations, ineffective individually, were effective in pro
treatment was initiated on Day 3 so that a moderately advanced longing life span.
tumor was present. Drugs were administered i.p. concurrently, Effect of [Au(DPPE)2]Cl on Tumor Cells in Vitro. The effect
and nontumorous mice were treated in a parallel with tumor- of [Au(DPPE)2]Cl on the clonogenicity of B16 and P388 cells
bearing mice so that an accurate assessment of the toxicity of following a 2-h treatment in vitro is shown in Fig. 5. Both
the combinations could be made. The results are summarized survival curves were characterized by a shoulder; with subse
in Table 3. Cisplatin at its MTD of 8.3 /¿mol/kg/day for 5 days quent exponential decreases in cell survival with increases in
5489
Downloaded from cancerres.aacrjournals.org on July 10, 2011
Copyright © 1986 American Association for Cancer Research
ANTITUMOR ACTIVITY OF BIS(DPPE)GOLD(I) CHLORIDE

drug concentration. The concentrations of [Au(DPPE)2]Cl re in the number of DNA single-strand breaks during the incuba
quired to inhibit the clonogenic capacity of B16 and P388 cells tion in drug-free medium. In addition to the DNA strand
by 50% following a 2-h exposure were 2 and 6 MM,respectively. breaks, [Au(DPPE)2]Cl, at low concentrations (5 MM),produced
The cytotoxic potency of [Au(DPPE)2]Cl was not markedly a significant number of DNA-protein cross-links. However,
decreased when cells were incubated with the compound in the using increasing concentrations, DNA-protein cross-links de
presence of fetal calf serum. The 50% inhibitory concentration creased and only strand breaks were evident.
for a 2-h exposure as measured in a clonogenic assay using B16 The effects of [Au(DPPE)2]Cl on the incorporation of 3H-
cells was only 2-fold lower in medium not containing serum as labeled precursors into DNA, RNA, and protein by B16 mela
compared to drug exposure in medium containing 10% fetal noma cells are shown in Fig. 7. The compound inhibited the
calf serum. This is in contrast to the cytotoxic monophosphine uptake of [3H]thymidine, [3H]uridine, and [3HJleucine into
gold(I) complex, auranofin (5), for which cytotoxic potency was DNA, RNA, and protein, respectively, In H16 cells in both a
decreased 10-fold in the presence of serum. concentration- and time-dependent manner. There was a some
Previous studies have shown that the cytotoxic effect of what greater effect on protein biosynthesis than on DNA and
auranofin against cells in culture occurs rapidly, is not the result RNA biosynthesis by [Au(DPPE)2]Cl. For example, 15 MM
of direct damage to DNA or chromatin, and does not occur as [Au(DPPE)2]Cl essentially shut down leucine incorporation
a result of preferential inhibition of DNA, RNA, and/or protein within 90 min, whereas inhibition of uridine and thymidine
synthesis (5, 14). To assess the nature of cell kill (i.e., acute incorporation was less pronounced.
versus delayed) produced by [Au(DPPE)2]Cl, the ability of P388 Chemical Reactivity of | Am I)l>l'l ),]( 1 in Model Systems.
cells to exclude trypan blue as a function of [Au(DPPE)2]Cl Previous work with auranofin and other monophosphine gold
treatment was evaluated and compared to clonogenicity. Fol complexes has shown that these compounds are reactive with
lowing a 2-h incubation (under experimental conditions iden serum proteins, cellular proteins, glutathione, and other small
tical to those shown in Fig. 5), there was no increase in the molecular weight thiols (15-18). These interactions are domi
percentage of cells stained by trypan blue following treatment nated by the reactivity of these gold complexes with sulfhydryl
with (Au(DPPE)2]Cl as high as 40 MM.Therefore, the concen groups contained in the respective biological ligands and these
tration of [Au(DPPE)2]Cl required to produce acute cell death
is at least 20-fold greater than that required to inhibit the
30 r
clonogenic capacity of B16 cells.
The effects of the drug on intracellular chromatin structure
were assessed by alkaline elution. [Au(DPPE)2]Cl produced a
concentration-dependent increase in DNA single-stranded 20
breaks in L1210 cells treated for l h at 37°C(Fig. 6). The
ability of the cells to repair this DNA damage was assessed by o.
u
IO
washing the cells which had been treated with the compound
for 1 hr and subsequently incubating the cells in drug-free
medium. The results indicate that there was a further increase
30 60 90
Minutes

40

O 30
x
E 20
o.
IO

30 60 90
Minutes
S
•o
. C
s 30

20

o.
IO

20 40 60 80 100
>jM of [Au(dppe)2] CI
30 60 90
Fig. 6. DNA strand breaks U.I and DNA-protein cross-links (9) produced in
Minutes
LI210 cells treated with increasing concentrations of [Au(DPPE)2]Cl Tor l h at
37*C. In addition, cells were treated with 25 and 50 »AI concentrations of the Fig. 7. Effect of [Au(DPPE)2]Cl on DNA, RNA and protein synthesis in B16
compound for 1 h, and the cells were then washed and resuspended in drug-free melanoma cells. Cells were treated with 0 (•),S (O), IO (D), and 15 (A) ,;\i
medium and incubated at 37*C for I h and strand breaks were measured (A). concentrations of the gold complex and the incorporation of ('HJthymidine (A),
Strand breaks and DNA-protein cross-links were measured by alkaline elution ['Hhiridine (B), and [3H]leucine (Q into TCA-precipitable material was measured
(see "Materials and Methods"). as a function of the duration of drug exposure.
5490
Downloaded from cancerres.aacrjournals.org on July 10, 2011
Copyright © 1986 American Association for Cancer Research
ANTITUMOR ACTIVITY OF BIS(DPPE)GOLD(I) CHLORIDE

reactions clearly play an important role in their pharmacologi Cudldppe


cal activities (5, 19). To gain insight into the potential role of
sulfhydryl interactions on the in vitro and in vivo pharmacology
of [Au(DPPE)2]Cl, its reactivity in the presence of thiols, disul-
fides, and serum proteins was investigated using 31P NMR
spectroscopy. The 31P NMR resonance of [Au(DPPE)2]Cl was
unaffected by the addition of GSH after 8 days in solution.
There was no change in the chemical shift (24.1 ppm), and the
resonance remained sharp, even in the presence of a large excess
(50 mol equivalents) of GSH showing that the thiol does not A.
bind to the complex or displace DPPE from the gold coordi
nation sphere. The bis(triethylphosphine) gold(I) complex
[Au(PEt3)2]Cl also has been reported to be unreactive towards
thiols in model reactions (20), but it does react with disulfides
by reductively cleaving —S—S— bonds with the formation of
Et3PO as the phosphine oxidation product. [Au(DPPE)2]Cl was
I , I I , I
found to be unreactive towards GSSG, as judged by NMR even 60 4O 20 -20
after 7 days of reaction time.
8/ppm
Incubation of 1 mM [Au(DPPE)2]Cl in bovine serum for 24
h caused no change in the 31P NMR resonances of phosphate Fig. 9. ('H) "P NMR spectra showing the reaction of [Au(DPPE)2]CI with
CuSO4. A, 20 mM [Au(DPPE)2]CI in methanol:D2O (3:1, v/v); B, supernatant
and phospholipids in the serum (Fig. 8). The [Au(DPPE)2]Cl from the reaction of [Au(DPPE)2]Cl with 1 mol equivalent of CuSO4 in metha-
resonance at 23.9 ppm broadened slightly (AT(i= 15 Hz). The nol:D2O (3:1, v/v); C, the precipitate from this reaction redissolved in CDC13.
The complex [Au(DPPE)2]2* has a 10-membered ring structure containing two
broadening may be indicative of some binding of the Au(I) ions.'
[Au(DPPE)2]+ cation to certain serum components (.e.g, fatty
acids) either by ion pairing or by hydrophobic interactions with Cu(I)phosphine complexes (28),4 it seems likely that the major
the phenyl rings of the complex. resonance at —
10.7 ppm corresponds to a Cu(I):DPPE complex.
A variety of structurally diverse antitumor drugs, e.g., bleo-
Indeed, atomic absorption measurements substantiated that the
mycin (21), doxorubicin (22), 1,10 phenanthroline (23), and
precipitate contains a substantial amount of copper; the
amsacrine (24, 25), are able to interact with and/or form [Au]:[Cu] ratio in the precipitate was 1:45. The 3IP NMR
coordination complexes with transition metal ions. We have spectrum of the water-soluble product(s) of the reaction of
demonstrated that the in vitro cytotoxic activity and in vivo
[Au(DPPE)2]Cl with CuSO4 is shown in Fig. 9B. The single
potency of DPPE are increased in the presence of Cu(II) salts
resonance at 39.5 ppm can be assigned to the annular complex
(26, 27). In addition, in vitro and in vivo evaluation of a series [Au2(DPPE)2]2+ which has an identical chemical shift in
of DPPE analogues provided evidence for a relationship be aqueous methanol.5
tween the ability of Cu(II) to potentiate the cytotoxic activities
of the DPPE analogues and their having in vivo antitumor
activity (26). In view of these observations, the potential for the DISCUSSION
reaction of [Au(DPPE)2]Cl with Cu(II) was examined in vitro.
DPPE and a number of closely related diphosphines and gold
The reaction of [Au(DPPE)2]Cl with 1 mol equivalent of
complexes thereof have potent cytotoxicity to tumor cells in
CuSO4 in aqueous methanol gave a thick white precipitate. The
31P NMR spectrum of this product, when resolved in chloro culture and have reproducible activity in animal tumor models
(6, 26).2 [Au(DPPE)2]Cl is unique relative to previously de
form (Fig. 9C), consisted of two broad resonances at -10.7 and
scribed gold(I) bisphosphines in that it is an unusually stable 4-
34.4 ppm. By comparison with our previous NMR studies of
coordinate, tetrahedral complex. Tumors sensitive to
[Au(DPPE)2]Cl in vivo include P388 leukemia, M5076 reticu-
lum cell sarcoma, B16 melanoma, and the mammary adenocar-
cinoma 16/c. Activity in the latter system demonstrates that
the compound is active when administered systemically against
a solid tumor.
[Au(dppe)2] + [Au(DPPE)2]Cl exhibits cytotoxic activity against tumor cells
in vitro at micromolar concentrations. The survival curves (Fig.
5) were characterized by a shoulder at low concentrations, and
exponential cell kill was seen with further increases in drug
concentration. This survival pattern is similar to that produced
by radiomimetic alkylating agents (29) but dissimilar to the
monophasic and exponential survival curves produced by au-
ranofin (5). The lack of immediate toxicity to cells by
[Au(DPPE)2]Cl, as measured by the ability of cells to exclude
trypan blue after l h exposure to the compound at concentra
tions determined to be cytotoxic in the clonogenic assay, sug
30 10 -10 gests that the cytotoxic effect of [Au(DPPE)2]Cl may be medi-
S/ppm 4 S. J. Berners-Price and P. J. Sadler. Manuscript in preparation.
Fig. 8. ('H) "P NMR spectrum at 24.2 MHz of 1 mM [Au(DPPE)2]CI in * S. J. Berners-Price and P. J. Sadler. Gold(I) complexes with bidentate tertiary
bovine serum after 24 h. The lyophilized serum was reconstituted in IM) before phosphine ligands: formation of annular versus tetrahedral chelated complexes,
addition of the compound. /'/.. phospholipids; Pi, inorganic phosphate. submitted for publication.
5491
Downloaded from cancerres.aacrjournals.org on July 10, 2011
Copyright © 1986 American Association for Cancer Research
ANTITUMOR ACTIVITY OF BIS(DPPE)GOLD(I) CHLORIDE

ated by interference with the reproductive capacity of the cell. is consistent with our 3IP NMR studies which demonstrate that
This result is in contrast with that found for auranofin under high concentrations of [Au(DPPE)2]Cl are stable in serum,
equivalent experimental conditions in which its cytotoxic effects consistent with the model reactions where no ligand exchange
appeared to be acutely mediated (5). with thiols or reactions with disulphide linkages were observed.
Incorporation studies demonstrated that [Au(DPPE)2]Cl in The complex is unlikely to react directly with other functional
hibited the biosynthesis of DNA, RNA, and protein. However, groups on proteins such as methionine SCH3, histidine N, or
whereas the rates of incorporation of the precursors into DNA aspartic acid and glutamic acid CO2~ groups. Electrostatic
and RNA were inhibited leucine incorporation into protein interactions may occur with negatively charged oxyanions and
appeared to be effectively terminated within 30 min of exposure hydrophobic interactions (of the van der Waals type) with ligand
of cells to 15 fÃ-M [Au(DPPE)2]Cl. Further experiments will be side chains.
required to determine if this apparent selective inhibition of
Previous studies from our laboratory have demonstrated that
protein synthesis by the compound is the result of an inhibition
DPPE has in vitro cytotoxic and in vivo antitumor activity
of amino acid transport into the cells, to a more direct inhibition
suggesting that the pharmacological activities of [Au(DPPE)2]-
of protein synthesis, or is secondary to other effects of the drug.
The observation that [Au(DPPE)2]Cl produces DNA-protein Cl may be the consequence of the delivery of DPPE to the
cross-links and DNA single-strand breaks in cells suggests that appropriate biological targets (26, 27). Phosphines are strong
the effects observed on macromolecular synthesis may be the reducing agents (37), and the reduction of essential cellular
result of chromai in damage which leads to inhibition of gene components or the generation of reactive radical species during
replication, transcription, and translation. The relative signifi phosphine oxidation, perhaps involving redox-active metal ions
cance of the DNA-protein cross-links and DNA single-strand such as copper and iron, may play a role in the cytotoxic activity
breaks in the cytotoxic effects of [Au(DPPE)2]Cl are not yet of DPPE or [Au(DPPE)2]Cl. Indeed, there are data to suggest
determined; however, the DNA-protein cross-links appeared to that copper is involved in the cytotoxic and antitumor ac
be the more dominant lesion in the cytotoxic concentration tion of DPPE (26). Our investigation of the reaction of
range of the compound with single-strand breaks evident only [Au(DPPE)2]Cl with CuSO4 shows that the complex is reactive
at supralethal concentrations of [Au(DPPE)2]Cl. towards Cu(II) ions, and a Cu(I)DPPE complex is one of the
Gold(I) phosphine complexes generally undergo rapid ligand products (Fig. 9). Cu(I) complexes may offer potential as anti-
exchange reactions so that the 31P NMR spectra of complexes cancer agents (38), and the chemical and pharmacological eval
of type [Au(PR3)n]Cl (n = 2, 3, or 4) in the presence of excess uation of this Cu(I)-containing reaction product is currently
1'k i consist of a single resonance at ambient temperature. under way in our laboratories. In addition, our studies have
Solutions usually must be cooled to below — 70°Cfor the
shown that [Au(DPPE)2]Cl is more stable in solution than
resolution of separate resonances for free and bound phosphine DPPE alone, which slowly undergoes solvent-dependent autox-
ligands (30-32). In contrast, we have reported that idation to the mono- and bis-phosphine oxides (1-10 days).6 In
[Au(DPPE)2]+ undergoes slow exchange in the NMR time scale
toto, these experimental observations led us to propose that the
with excess DPPE so that separate resonances are resolved for role of gold in the [Au(DPPE)2]Cl complex may be to protect
free and bound DPPE in the 3IP NMR spectrum at 300°K
the ligand from oxidation in vitro and in vivo prior to its
(10). Therefore, although 4-coordinate gold(I) complexes are
interactions with extracellular (and/or intracellular) redox com
rare and generally unstable species, tetrahedral gold (I) com ponents and critical cellular target(s). This would account for
plexes with bidentate chelated phosphine ligands appear to be the increased in vitro and in vivo cytotoxic potency of
more inert toward phosphine exchange than 2- or 3-coordinate
[Au(DPPE)2]Cl with respect to DPPE itself. While the identity
gold(I) complexes containing monodentate ligands. This unu
sual chemical stability of the bis-chelated complex may contrib of the critical cellular target(s) is unclear, the lipophilic nature
of both DPPE and [Au(DPPE)2]Cl may selectively target these
ute to the contrasting pharmacological properties compared to
molecules to cellular membrane organdÃ-es or allow the com
those of the R3PAu Y type of complexes.
pounds access to intercellular targets.
These studies have shown that [Au(DPPE)2]Cl does not read
In conclusion, the tetrahedral complex [Au(DPPE)2)Cl has
ily undergo substitution reactions with thiols in aqueous media.
This is to be expected in view of the high stability of gold- demonstrated significant and reproducible antitumor activity in
phosphorus bonds. The high coordination number of Au(I), 4, a broad spectrum of murine tumor models and potent cytotoxic
also may introduce considerable activation barriers to exchange. activity to tumor cells in vitro. In vitro and in vivo studies have
However, complexes of type R3PAuY, where Y is a good leaving provided evidence which suggests that the cytotoxic mecha
group, for instance chlorine (20) or 7V-imide (33), readily nism^) of this compound are different from those of auranofin
undergo substitution reactions with thiols in aqueous media, and cisplatin. In addition, the contrasting pharmacological
and there is fast exchange between free and bound thiol on the profile of [Au(DPPE)2]Cl with respect to other gold(I) phos
NMR time scale. Reactions between thiols and gold drugs in phine complexes may be related to both the kinetic stability of
vivo are likely to play a role in the mechanism of action of these the complex and its stability in the presence of thiols.
drugs in rheumatoid arthritis, perhaps by gold binding to sulfhy-
dryl groups in membranes (18), enzyme active sites (34), or the ACKNOWLEDGMENTS
metal storage protein metallothionein (35, 36). This notion is
consistent with data measured for auranofin in which the cy S. J. B. P. and P. J. S. are grateful to Smith Kline & French Labo
totoxic activity of the drug was reduced 10-fold by the addition ratories and the Medical Research Council for support. We thank
of 10% fetal calf serum during the exposure of the drug to cells Susanne Young and Shirley Peterson for their excellent secretarial
assistance in preparing this manuscript and Drs. Judith Hempel and
in culture (5). This result is attributed to the rapid binding of
auranofin-derived gold to serum proteins. The effect of 10% James McArdle for their helpful discussions.
fetal calf serum on the cytotoxic potency of [Au(DPPE)2]Cl was
significantly less than that evidenced for auranofin. This result 6 S. J. Bemers-Price and P. J. Sadler, unpublished observations.
5492
Downloaded from cancerres.aacrjournals.org on July 10, 2011
Copyright © 1986 American Association for Cancer Research
ANTITUMOR ACTIVITY OF BIS(DPPE)GOLD(I) CHLORIDE
20. Malik, N. A., Otiko, G., and Sadler, P. J. Control of intra- and extra-cellular
REFERENCES sulphydryl-disulphide balances with gold phosphine drugs: "I' nuclear mag
netic resonance studies of human blood. J. Inorg. Biochem., /-': 317-322,
1. Mirabelli, C. K., Johnson, R. K., Hill, D. T., Faucette, L. F., Girard, G. R.,
Keu, G. Y., Sung, C-M., and Crooke, S. T. Correlation of the in vitro 1980.
cytotoxic and in vivo antitumor activities of gold(I) coordination complexes. 21. Sausville, E. A., Peisach, J., and Horwitz, S. F. A role for ferrous iron and
J. Med. Chem., 29: 218-223, 1986. oxygen in the degradation of DNA by bleomycin. Biochem. Biophys. Res.
2. Lorber, A., Simon, T. M., Leeb, J., Peter, A., and Wilcox, S. A. Effects of Commun., 91: 871-877, 1976.
chrysotherapy on parameters of immune response. J. Rheumatol., 6 (Suppl. 22. Someya, A., and Tanaka, N. DNA strand scission induced by Adriamycin
5): 81-90, 1979. and aclacinomycin A. J. Antibiot. (Tokyo), 32:839-844, 1979.
3. Finkelstein, A. E., Burrone, O. R., Walz, D. T., and Misher, A. Effect of 23. Sigman, D. S., Graham, D. R., Aurora, V. D., and Stern, A. Oxygen-
auranofin on DNA and protein synthesis in human lymphocytes. J. Rheu dependent cleavage of DNA by the 1,10-phenanthroline-cuprous complex. J.
matol., 4: 245-251, 1977. Biol. Chem., 254: 12269-12276, 1979.
4. Simon, T. M., Kunishima, D. H., Vilbert, G. J., and Lorber, A. Screen trial 24. Wong, A. L., Huang, C. H., and Crooke, S. T. Deoxyribonucleic acid breaks
produced by 4'-(9-acridinylamino)methanesulfon-m-anisidide and copper.
with a coordinated gold compound auranofin using mouse lymphocytic
leukemia P388. Cancer Res., 41:94-97, 1981. Biochemistry, 23: 2939-2945, 1984.
5. Mirabelli, C. K., Johnson, R. K., Sung, C-M., Faucette, L. F., Muirhead, K., 25. Wong, A. L., Huang, C. H., and Crooke, S. T. Mechanism of deoxyribonu-
cleic acid breakage induced by 4'-(9-acridinylamino)methanesulfon-m-anisi-
and Crooke, S. T. Evaluation of the in vivo antitumor activity and in vitro
cytotoxic properties of auranofin, a coordinated gold compound in murine dide and copper. Biochemistry, 23:2945-2952, 1984.
tumor models. Cancer Res., 45: 32-39, 1985. 26. Snyder, R. M., Mirabelli, C. K., Johnson, R. K., Sung, C-M., Faucette, L.
6. Johnson, R. K., Mirabelli, C. K., Faucette, L. F., McCabe, F. L., Sutton, B. F., McCabe, F. L., Zimmerman, J. P., Whitman, M., Hempel, J. C., and
M., Bryan, D. L., Girard, G. R., and Hill, D. T. Antitumor activity of Crooke, S. T. Modulation of antitumor and biochemical properties of
compounds related to bis(diphenylphosphine)ethane and its chlorogold(I) bis(diphenylphosphine) with metals. Cancer Res., 46: 5054-5060, 1986.
27. Mirabelli, C. K., Johnson, R. K., Bartus, J. O., Sung, C-M., and VonHoff,
coordination complex. Proc. Am. Assoc. Cancer Res., 26: 254, 1985.
7. Jones, P. G. X-ray structural investigations of gold compounds. Gold Bull., D. D. Cytotoxic properties and metal interactions of bis(diphenyl-
14: 102-117, I98I. phosphine)ethane and related antineoplastic bisphosphines. Proc. Am. Assoc.
8. Jones, P. G. Is regular tetrahedral geometry possible in gold(I) phosphine Cancer Res., 26: 256, 1985.
complexes? X-ray crystal structures of three modifications of 28. Berners-Price, S. J., Brevard, C., Pagelot, A., and Sadler, P. J.
(PPh3)5Au*BP.,~. J. Chem. Soc. Chem. Commun., 1031-1033, 1980. [Cu(Ph2PCH2CH2PEt2)2]Cl: a chelated copper(I) complex with tetrahedral
9. Elder, R. C., Kelle Zeiher, E. H., Onady, M., and Whittle, R. R. Nearly stereochemistry. Rate of inversion compared with those of isostructural
regular tetrahedral geometry in a gold(I) phosphine complex. X-ray crystal silver(I) and gold(I) complexes. Inorg. Chem., in press, 1986.
structure of a tetrakis(methyldiphenylphospnine)gold(I) hexafluorophos- 29. Drewinko, B., Roper, P. R., and Barlogie, B. Patterns of cell survival
phate. J. Chem. Soc. Chem. Commun., 900-901, 1981. following treatment with antitumor agents in vitro. Ear. J. Cancer, 15: 93-
10. Berners-Price, S. J., Mazid, M. A., and Sadler, P. J. Stable gold(I) complexes 98, 1979.
with chetate rings: solution studies of bis(phosphino)ethane complexes and 30. Mays, M. A., and Vergnano, P. A. Structure and bonding in gold(I) com
X-ray crystal structure of bis[l,2-bis(diphenylphosphinoethane))gold(I) hex- pounds. Part 4. A phosphorus-31 nuclear magnetic study of the structure of
afluoroamimonate acetone (1/1). J. Chem. Soc. Dalton Trans., 969-974, some gold(I) phosphine complexes in solution. J. Chem. Soc. Dalton Trans.,
1112-1114, 1979.
1984. 31. Parish, R. V., Colburn, C. V., Hill, W. E., and McAuliffe, C. A. "P NMR
11. Bates, P. A., and Waters, J. M. A tetrahedral complex of gold(l). Crystal and
molecular structure of Au(Ph2PCH2CH2PPh2)2CI • 2H2O. Inorg. Chim. Acta, study of tertiary phosphine complexes of gold(I). J. Chem. Soc. Chem.
«7:151-156, 1984. Commun., 218-219, 1979.
12. Metcalfe, S. A., Whelan, R. D. H., Masters, J. R. W., and Hill, B. T. In vitro 32. Parish, R. V., Parry, O., McAuliffe, C. A. Characterization of two-,
three-, and four-coordinate gold(I) complexes by "'An Mossbauer "I' '111
responses of human prostate tumor cell lines to a range of antitumor agents.
Int. J. Cancer, 32:351-358, 1983. nuclear magnetic resonance spectroscopy. J. Chem. Soc. Dalton Trans.,
13. Kohn, K. W., Ewig, R. A. G., Erickson, L. C., and Zwelling, L. A. In: E. C. 2098-2103, 1981.
Friedberg and P. C. Hanawalt (eds.). Repair: a Laboratory Manual of 33. Berners-Price, S. J., DiMartino, M. J., Hill, D. T., Kuroda, R., Mazid, M.
Research Techniques, pp. 379-401. New York: Marcel Dekker, 1981. A., and Sadler, P. J. Tertiary phosphine complexes of gold(I) and gold(III)
14. Mirabelli, C. K., Sung, C-M., Zimmerman, J. P., Hill, D. T., Mong, S., and with imido igands: 'H, "!'. and 15N NMR spectroscopy, antiinflammatory
Crooke, S. T. Interactions of gold coordination complexes with DNA. activity and X-ray crystal structure of (phthalimidoXtriethylphosphine)
Biochem. Pharmacol., 35: 1427-1433, 1986. gold(I). Inorg. Chem., 24: 3425-3434, 1985.
15. Carlock, M. T., Shaw, F. C., Ill, Eidsness, M. K., Watkins, J., and Elder, R. 34. Allaudeen, H. S., Snyder, R. M., Whitman, M. H., and Crooke, S. T. Effects
C. On the reactions of auranofin and Et3PAuCl with serum albumin. Inorg. of coordinated gold compounds on in vitro and in situ DNA replication.
Chem., 25: 333-339, 1986. Biochem. Pharmacol., 34: 3243-3250, 1985.
16. Razi, M. T., Otiko, G., and Sadler, P. J. Ligand exchange reactions of gold 35. Laib, J. E., Shaw, C. F., Ill, and Petering, D. H. Formation and characteri
drugs in model systems and in red cells. In: S. J. Lippard (ed.). Platinum, zation of aurothioneins: Au, Zn, Cd-thionein, Au, Cd-thionein, and (thiom-
Gold and Other Metal Chemotherapeutic Agents, ACS Symposium Series alato-Au),-thionein. Biochemistry, 24: 1977-1986, 1985.
209, pp. 371-384. Washington, DC: American Chemical Society, 1983. 36. Butt, T. R., Sternberg, E. J., Mirabelli, C. K., and Crooks, S. T. Regulation
17. Grootveld, M. C., Razi, M. T., and Sadler, P. J. Progress in the characteri of metallothionein gene expression in mammalian cells by gold compounds.
zation of gold drugs. Clin. Rheumatol., 3: 5-16, 1984. Mol. Pharmacol., 29: 204-210, 1986.
18. Snyder, R. M., Mirabelli, C. K., and Crooke, S. T. Cellular association 37. Cadogan, J. I. G, and Mackie, R. K. Tervalent phosphorus compounds in
intracellular distribution and efflux of auranofin via sequential ligand ex organic synthesis. Chem. Soc. Rev., 387: 87-137, 1974.
change reactions. Biochem. Pharmacol., 35: 923-932, 1986. 38. Sadler, P. J., Nasr, M., and Narayanan, V. L. The design of metal complexes
19. Walz, D. T., Griswold, D. E., Dimartino, M. J., and Humbler. E. Effect of as anticancer drugs. In: M. P. Hacker, E. B. Douple. and I. H. Krakhoff
auranofin dose regimen changeup on cell-associated gold in rheumatoid (eds.). Platinum Coordination Complexes in Cancer Chemotherapy, pp. 290-
arthritic patients. J. Rheumatol., 8:829-832, 1981. 304. The Hague: Martinus Nijhoff Publishers, 1984.

5493
Downloaded from cancerres.aacrjournals.org on July 10, 2011
View publication stats
Copyright © 1986 American Association for Cancer Research

You might also like