• Keine Ergebnisse gefunden

Dichloro-[ 1-( hydroxyphenyl)-2-phenylethylenediamine]platinum(II) Complexes: Testing on the Human Ovarian Cancer Cell Lines NIH:

N/A
N/A
Protected

Academic year: 2022

Aktie "Dichloro-[ 1-( hydroxyphenyl)-2-phenylethylenediamine]platinum(II) Complexes: Testing on the Human Ovarian Cancer Cell Lines NIH: "

Copied!
7
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Dichloro-[ 1

Dichloro-[ 1-( hydroxyphenyl)-2-phenylethylenediamine]platinum(II) Complexes: Testing on the Human Ovarian Cancer Cell Lines NIH:

OVCAR 3 and SK OV 3*)

Giinther Bemhardt, Richard Muller, Ronald Gust, Herta Reile, Christoph Keller, Thilo SpruB, and Helmut Schonenberger

Institut f& Pharmazie, Lehrstuhl Pharmazeutische Chemie 11, Sonderforschungsbereich 234 (SFB 234), Universitiit Regensburg, Universit;itsstraBe 3 1, W- 8400 Regensburg, Germany

Received January 11,1991

The diastereoisomeric dichlore[ 1-(2-, 3- and 4-hydmxyphenyl)-2-phenyl- ethylenediamine]platinum(Il) complexes were tested on two human ovarian cancer cell lines N I H OVCAR-3 and SK-OV-3, both resistant against cis- platin Dichloro-[fhreo-1-(3-hydroxyphenyl)-2-p~nylethyle~iami~]plati- num(II) (rhreo-S-PtC12) proved to be the most active representative of the new series, producing cytocidal effects at a concentration range of 2.5 to 5.0 pM on the NM: OVCAR-3 cell Line. On the more resistant SK-OV-3 cell line, threo-5-PtC12 was only moderately active, while in combination with BSO, a GSH level lowering compound, fhreo-5-PtC12 showed a strong syn- ergistic effect.

Dichlor~[l-(hydroxyphenyl)-2phenylethylendlamin]piatin(II)-Kom- plexe. m n g an den mensehllchen Ovarialcarclnom-Zellinlen NIH:

OVCAR-3 und SK-OV-3

Die diastereomeren Dichloro-[ 1-(2-, 3- und 4-hydroxyphenyl)-2-phenyleth- ylendiamin]platin(lI)-Komplexe wurden an zwei cisplatinresistenten, menschlichen Ovarialcarcinom-Zellinien, NIH: OVCAR-3 und SK-OV-3, getestet. Dichloro-[rhreo-l-(3-hydroxyphenyl)-2-phenylethylendi~in]p~a- tin(II) (threo-J-PtCl2) envies sich als der aktivste Vertreter der Reihe. Threo- 5-PtC12 ~ f i im Konzentrationsbereich 2.5

-

5.0 an der NM: OVCAR-3

Zellinie cytocide Effekte hervor. An der resistenten SK-OV-3 Zellinie war rhreo-S-PtC12 nur d i g wirksam; es zeigte aber in Kombination mit BSO.

einer GSH-Spiegel emiedrigenden Substanz. stark synergistische Effekte.

- +

Compound X

I-PtC12 4-OH 2-PtC12 3-OH S-PtCIz 2-OH 5-PtCI

..

3-OH

i-PtC1:” 4-OH 4-PtCl2’ 2-OH

/vi3

C1 /pt\ C1

coo-

- I

I

I I

II I

I H,N-C - H

( C H I ! , H

c= s - C H 2 - C - H

N’i CH,

CH,

CisDlot in

But hi on ln- sulfoximln IBSO)

* Available asthreoform

I

. Available as r h ~ o and erythro form Scheme 1

X‘ In recent publicationsi4) we have shown that stereoisomeric dichloro-

4-OH (1,2-diphenylethylenediamine)platinurn(II) complexes with hydroxy

2-OH groups in the 4-,3- or 2-positions of both benzene rings (Scheme 1, com-

H pounds 1-PtC12 to 3-PtCld are of interest as antitumor agents. D,L-&PtCI2 ent Ehrlich ascites tumor of the mouse. In this experiment 70% of the tumor-bearing animals were cured by complex D,L-%Ptc12 but only 20%

by complex meso-3-PtC12).

This result prompted us to a thorough study of the stereo- isomeric [ 1,2-bis(2-hydroxyphenyl)ethylenediamine]dichlo- roplatinum(II) complexes (meso-, D,L-, (+)-, and (-)-3- PtC12) on the

NIH

: OVCAR-3 ovarian cancer cell line, which exhibits a multidrug resistance phenotype (compare ref?) and footnote 1). The NIH :

OVCAR

3 cell line was established from a patient with ovarian cancer who was treated unsuccessfully with cisplatin, adriamycin and cyclo- phosphamide’! Among the studied stereoisomeric 3-PtC12 complexes the (S),(S)configurated, levorotatory compound proved to be the most active one. Since non-toxic concen- trations of this compound produces cytocidal effects on the NIH: OVCAR 3 cell line, it is of interest for further evalu- ation for the therapy of ovarian cancer.

This publication will report on the testing of the structu- rally related diastereomeric dichloro-[ 1-(hydroxypheny1)-2-

3-OH

H

H and especially ~,~.-3-Ptc12 led to a marked inhibition of the cisplatin-resist-

1. Cisplatin based combination chemotherapy regimens produce complete remissions in 6040% of patients with advanced stage ovarian cancer. However, their effectiveness is limited by the development of acquired resistance”. Therefore, the development of new platinum complexes, which overcome the cisplatin resistance, is of great interest: (a) in combination with cisplatin for first-line treatment of ovarian cancer to avoid or retard the development to resistance. (b) for the second-line therapy of ovarian cancer after development of resistance against cisplatin.

= Dedicated to Professor Dr. Dr. E. Mutschler on the occasion of his 60th birthday.

Arch. Phurm. (Weinheim) 325.93-99 (1992) 0VCH Verlagsgesellschaft mbH, D-6940 Weinheim, 1992 0365-6233/92/0202-03 $3.50

+

.25/0

(2)

94 Schonenberger and coworkers

phenylethylenediamine]platinum(II) complexes on the human epithelial ovarian cancer cell lines NIH: OVCAR-3 Derivatives which are OH-substituted in 2-, 3-, and 4-PO- sition (4-PtC12, 5-PtCl2, and 6-PtC12; Scheme 1) were used in this study. With the exception of eryrhro-4-PtCl2, the synthesis which was not successful (cf. ref?)), all com- pounds were available as threo- and erythro-forms.

For characterization of both cell lines a cytogenetic ana- lysis as well as an investigation of growth kinetics and of the sensitivity against the therapeutically most used drug cisplatin were performed.

The cytogenetic analysis demonstrated that the NIH:

OVCAR-3 cell line included cells with a hypotriploid to a hypohexaploid karyotype and the SK-OV-3 cells with a hy- podiploid to a hypotetraploid karyotype. The modal chro- and SK-OV-3.

-

u Y

24 1 1

21

18

15

12

9

6

3 0

57 60 63 66 69 73 126 131

mosome numbers amount to 63 for NIH: OVCAR-3 (Fig. 1) and to 43 and 83 for SK-OV-3 (Fig. 2). The karyograms of both cell lines show numerous chromosome anomalies.

Their morphological and cytogenetic characteristic proved to be constant in the course of the study.

Fig. 3A and B illustrate examplarily the growth curves and the corresponding doubling times at any time of incuba- tion for the MH: OVCAR-3 cell line in the passages 30 and 32 (see footnote 2)). Under the given experimental condi- tions the logarithmic phase covers only a small fraction of the overall growth curve. In a plot doubling time versus time of incubation exponential growth is characterized by a parallelism of the graphs with the X-axis. The NIH:

OVCAR-3 cell line grows exponentially for about two generations. The same is true for the SK-OV-3 cell line in the passages 38 to 40, which were used for the evaluation of

14 12

Lo 10

-

Y

Lo .c

g a

E

6 4 2 0

34 42 45 69 75 79 82 85 88 163

C h r m s m N W r C h r m s m Nmber

Fig. 1: Chromosome distribution of NIH-OVCAR 3 cells. (0) 29th passage from origin, (m) 51th passage from origin: The cells were passaged weekly.

For each passage the chromosomes from 50 well-spread metaphase plates

were counted. counted.

Fig. 2: Chromosome distribution of SK-OV-3 cells, (0) 32th passage from origin, (B) 42th passage from origin: The cells were passaged weekly. For each passage the chromosomes from 50 well-spread metaphase plates were

A B

1 I I I i

0 1 0 0 2 0 0 300

Fig. 3: Growth curves and corre- sponding doubling times of the NIH OVCAR-3 cell line in de- pendence on the passage number.

Inoculum: 100 pl/well at a density of 19,25 cells/microscopic field for

0 1 0 0 2 0 0 300 h passages 30,32.

Arch. Pharm. (Weinheim) 325. 93-99 (1992)

(3)

Dichloro-[ 11)-Complexes

0 100 2 0 0 300 h

0 1 cio 2 0 9 300

Fig. 4B: Variation of time of drug exposure: Effect of cisplatin (A: 1 pM) on the proliferation of NIH: OVCAR-3 ovarian cancer cells. Plot of correc- ted T/C values versus time; Inoculum: 100 pvwell at a density of 17 cells (37th passage)/microscopic field; preincubation 61 h; drug incubation 1.3.6, and 12 h; final incubation without drug; duration of the experiment 244 h.

Fig. 4 A Effect of cisplatin on the proliferation of NIH: OVCAR-3 ovarian cancer cells. Plot of corrected T/C values versus time of drug exposure.

Inoculum: 100 pVwell at a density of 19 cells (30th passage) / microscopic field; preincubation 73.5 h, drug incubation 256 h.

the antitumor activity of the new platinum complexes. A doubling time of about 35 h was estimated for the NIH.

OVCAR-3 line and one of about 28 h for the SK-OV-3 line.

In order to get informations on the sensitivity of the NIH:

OVCAR-3 cell line against cisplatin, the most active drug in the therapy of ovarian cancer, the cells were incubated with this compound at therapeutically relevant concentrations for 256 h. Only at the highest concentrations (1 and 5 mM) a cytocidal effect, which we consider essential for a curative effect in vivo, could be detected (Fig. 4A). However, a rela- tively long period of contact between cells and cisplatin (6 h) is required to achieve cell kill (compare Fig. 4B). Due to the marked toxic side effects and the pharmacokinetics of cis- platin it is difficult to meet this demand. The SK-OV-3 ovarian cancer cell line proved to be less sensitive against cisplatin than the NIH: OVCAR-3 line (Fig. 5). In a concen- tration of 2.5 pM, which produced cytocidal effects on the NIH: OVCAR-3 cell line, cisplatin showed merely cyto- static activity on the SK-OV-3 cell line. After an incubation period of about 140 h development of resistance of the SK- OV-3 cells occurred.

In long term experiments (256 h drug incubation) the dia- stereoisomeric dichloro-[ l -(hydroxyphenyl)-2-phenylethy- lenediamine]platinum(II) complexes showed a strong con- centrationdependent activity on the cisplatin-resistant NIH:

OVCAR-3 cell line (Fig. 6-10).

In the threo series the 2- and 3-hydroxy-substituted com- plexes (threo-CPtC12 and fhreo-5-PtCl2) produced cytocidal effects at a concentration of 5 pM (Fig. 6 and 7). The 4-hy- droxy-substituted complex (threo-CPtCl2) was merely cy- tostatically active at this concentration (5 pM, Fig. 9). Com- parison of the activity of fhreu-rl-PtC12 and threo-5-PtCl2 at lower concentrations reveals the superiority of rhreo-5-PtCl~.

100

-

.Y

u 60

\ I- 3 aJ

4-J V aJ L V b 20

0

0 100 200 h

Fig. 5: Effect of cisplatin on the proliferation of SK-OV-3 ovarian cancer cells. Plot of corrected T/C values versus time of drug exposure. Inoculum:

100 pl/well at a density of 7 cells (38th passage) / microscopic field;

preincubation 77 h, drug incubation 162 h.

Erythro-5-PtCl2, which also brought about cytocidal effects at the highest concentration (5 pM), was less active than its diastereoisomer rhre0-5-PtQ (compare Fig. 7 and 8). The antitumor activity of eryrhro-5-PtCl2 was comparable with that of threo-CPtCl2. In contrast to the diastereoisomeric pair 5-PtCl2 (threo-Ei-PtClz is more active than erythro-5-PtC12) the eryrhro-6-PtQ proved to be somewhat more active than its counterpart thre0-6-PtQ (compare Fig. 9 and 10). The comparison of the test results of rhreo-5-PtC1.2, the most ac- tive representative of the new series, with those of (-)-3- 2. Comparable growth kinetics were also observed for the higher passage numbers of the NIH: OVCAR-3 cell line which were used in the test series.

Arch. Pharm. (Weinheim) 325.93-99 (1992)

(4)

96 Schonenberger and coworkers

100

-

be

-

u 60

\ I-

J W il W V L

u"

L 20 0

1 0 0 M

-

v

60

J P, U

U 0, L

s

L 20

0

100

- 5

v 60

\ I-

3 aJ

Y V 01 L

s

L 2 0

0

threo-_5-PtC12

0 1 0 0 Z O O 300 h

0 1 0 3 2 O C ? O C k

100

V t-

\ 60

L

s

L 20

0

0 1 0 0 200 300 h 0 1 0 0 200 300 h

100

-

bp

-

t 60 U

0 )

u

U aJ

L

u"

L- 2 0

0

100

-

M u

v

2

60 J

0, U aJ

L L V 0

U

20

0

erythro-5-PtC l 2

0 1 0 0 200 300 h

0 1 0 0 2 0 0 3 0 0 h Fig. 6-11: Effect of rhreo-4-PtCI2, threo-S-F'tCI2, eryrhro-5-PtCI2. rhreOaPtCl2, erythro-6-PtCIz, and (-)-3-W12 on the proliferation of NIH OVCAR-3 ovarian cancer cells. Plot of corrected T/C values versus time of drug exposure. Inoculum: 100 pVwell at a density of 19 cells (30th passage) /microscopic field; preincubation 73.5 h; drug incubation 265 h.

PtC12, which we used as positive control, showed a higher potency of the latter (compare Fig. 7 and 11).

To find out the time which is necessary to achieve a cyto- cidal effect of threo-5-PtCl2, the most active representative of the new platinum complexes, the

NIH:

OVCAR-3 cells were preincubated with this drug at concentrations of 2.5 and 5.0 pM for 1, 3, 6, and 12 h, respectively, and the growth curves were registered after removal of the drug by medium exchange (duration of the experiment 244 h). In a concentration of 5.0 pM rhreo-S-PtCl2 produced cytocidal effects after a drug-cell-contact of at least 3 h (Fig. 12A).

However, at a concentration of 2.5 pLM only a drug incuba- tion time of 12 h led to cytocidal effects (Fig. 12B).

Comparative experiments with (-)-3-PtC12 (which was used as control) revealed that at a drug concentration of 2.5

pM

in-

cubation times of 2 3 h were necessary to bring about cyto- cidal effects (Fig. 13). From this experiment, too, the supe- riority of (-)-3-PtC12 compared with rhreo-5-PtC12 becomes evident.

On the SK-OV-3 ovarian cancer cell line, which showed a marked resistance against cisplatin, only the most active representative of the new series, rhreo-S-PtClz was testet.

Even at the highest concentration (5 pM) only a weak cy- tostatic effect was obvious (Fig. 14). The comparison com- pound (-)-3-PtC12 displayed a similar low activity, which was even limited in time. After a drug incubation of 150 h.

cells grow unlimited (Fig. 15).

In the development of the resistance of ovarian cancer against cisplatin the ability of malignant cells to elevate its content of glutathione (GSH) is supposed to be of great importance5'. Glutathione inactivates drugs like cis-

Arch. Pharm. (Weinheim) 32.c. 93-99 (1992)

(5)

Dichloro-[ 1

100

‘ -

60

V \ t- U

c)

u W 0 U a d

2

2 0 0

B

100

<

e-2

-

u \

J

c) U

u aa

L L 0

Y

t- 6 0

v 2 0

0

0 100 200 300 h 0

drug incubation (h) drug incubation (h)

threo-_5-PtCI2 (5,O UM

ug i n c u b a t i o r

0 1 0 0 200 300

Fig. 13: Variation of the time of drug exposure: Effect of (-)-3-PtC12 (2.5 pM) on the proliferation of NIH: OVCAR-3 ovarian cancer cells. Plot of corrected T/C values versus time; Inoculum: 100 pl/well at a density of 17 cells (37th passage)/microscopic field; preincubation 61 h; drug incubation 1,3,6, and 12 h; final incubation without drug: duration of the experiment 244 h.

platin, which produce their effect by reaction with nucleophilic targets of the tumor cell (e.g. DNA). Therefore, drugs causing a GSH reduction in the cells should lead to a potentiation of the effect of platinum complexes. In this context buthionine sulfoximine (BSO), an inhibitor of the y-gluta- mylcsteine synthetase, the key enzyme of GSH biosynthesis. has met great interest. At a concentration range of 15 to 25 pM it produces areduction of the GSH level of about 85% in N I H OVCAR-3 cells and, as a consequence, markedly elevates the inhibitory effect of cisplatin on this cell line5’.

Due to these findings the combined administration of BSO and platinum complexes is being thoroughly studied. Our experiments on the SK-OV-3 cell line revealed a strong effect of fhreo-5-PtClz in a concentration of 2.5

pM

after preincubation with BSO (50 pM, 48 h; Fig. 17).

This is a remarkable finding, since both substances, rhree 5-PtC12 (2.5

pM)

and BSO (50

pM)

are ineffective when

Fig. 12: Variation of the time of drug expo- sure: Effect of rhreo-IPtClz (A. 2.5 pM: B:

5.0 pM) on the proliferation of NIH. OV- CAR-3 ovarian cancer cells. Plot of correc- ted T/C values versus time. Inoculum: 100

@/well at a density of 17 cells (37th passa- ge) / microscopic field, preincubation 61 h;

drug incubation 1,3,6, and 12 h; final incu- bation without drug: duration of the experi- 1 0 0 2 0 0 300 h ment244h.

applied singly (compare Fig. 14 and 16). (-)-3-PtC12 was markedly less active under the same conditions (Fig. 18). It causes only weak cytostatic effects.

Also with cisplatin, the antitumor effect was only slightly increased by BSO. At the therapeutically relevant cisplatin concentration (0.5

pM)

a combination with

BSO

(50 pM) gave no cytocidal effects.

Only at therapeutically non-relevant cisplatin concentra- tions (2.5 yM), which are themselves markedly cytostatic, the additional treatment with BSO ends up in a vast cell kill (Fig. 19). The strongly synergistic effect of the fhreo-5- PtClflSO combination could be of great interest in the therapy of the cisplatin resistent ovarian cancer.

Further investigations using in vifro and in vivo ovarian cancer models are planned.

The technical assistance of E. Aichinger, L. Gortswinrer, D. Krisam, S . Paulus, P. Pisfor, and P. Richfhanuner is gratefully acknowledged.

Thanks are also due to the Deutsche Forschungsgemeinschaft (SFB 234).

the Mafrhias Lackas-Stiftung fiir Krebsforschung and the Fonds der che- mischen Industrie for financial support.

Biological methods

NIH : OVCAR-3 ovarian cancer cell line. The NIH OVCAR-3 (ATCC no. HTB 161), a human adenocarcinoma of the ovary’), was obtained from the American Type Culture Collection in passage 17. Cell-line-banking and quality con- trol were performed according to the “see stock concept“ re- viewed by Hay’’. The cells were maintained in RPMI-1640 (Sigma) containing NaHC03 (2 gfl), gentamicin (50

ma),

10%

BMS

(Seromed) and insulin (10 yglml) (Sigma) in 75- cmz flasks at 37°C in a H,O-saturated atmosphere of 95% air and 5% COz. The cells were serially passaged weekly fol- lowing trypsinization using trypsin/EDTA (Boehringer). For chemosensitivity testing the cells (in passage 24,30,32,37, and 49, respectively, were plated in 96-well microplates (100 pl/well at a density of about 17-28 cells/microscopic field (Leitz Diavert, 320x)) and were allowed to attach. After 48- 80 h, the medium was removed by suction and replaced with

Arch. Pharm. (Weinheim) 325,93-99 (1992)

(6)

98 Schonenberger and coworkers

1 0 0

bp

U

\ t 3 W 2

V aJ

L L U 0

-

60

20

0

threo-5-PtCl

5,O IJM

0 1 0 0 200

100

-

t-e V

\ I-

3 Q) V a

L L 0 V

-

60

Y

20

0

0 1 0 0 200 h

Fig. 14 and 15: Effect of rhreo-5-PtClz and (-)-3-PtC12 on the proliferation of SK-OV-3 ovarian cancer cells. Plot of corrected T/C values versus time of drug exposure. Inoculum: 100 hl/well at a density of 7 cells (38th passage) / microscopic field;

preincubation 77 h drug incubation 162 h.

-0 aJ

Y V 01 L

s

L 2 5

0

0 1 0 0 2 0 0 h

Fig. 1 6 Effect of BSO (30 and 60 pM) on the proliferation of SK-OV-3 ovarian cancer cells. Plot of corrected T/C values versus time of drug expo- sure. Inoculum: 100 pl/well at a density of 100 cells (39th passage) / mi- croscopic field preincubation 44 h, drug incubation 160 h.

fresh medium (200 kl/well) containing drug (drugs were added as a 1000-fold stock solution) or pure solvent. The platinum complexes were dissolved in DMF. On every plate two rows (n = 16) acted as controls, whereas two vertical rows (n = 16) per drug concentration and time point were used. After various times of incubation the cells were fixed with glutaraldehyde and stored under

PBS

at 4OC. All plates were stained with crystal violet simultaneously. The process- ing procedure and data analysis were performed as described by Reile et a1.l'). Drug effects were calculated as corrected T/C values according to: T/C,, = (T

-

C&(C

-

Co) 100

[%I,

where T is the absorbance of treated cells, C the absorbance of the controls and C, the absorbance at the time (t = 0) when drug was added. The experimental errors for T/C,, range

100

6 0

20

0

0 60 120

Fig. 17: Effect of rheo-5-PtC12 (2.5 pM) on the proliferation of SK-OV-3 ovarian cancer cells after preincubation with BSO (50 pM, 48 h). Plot of corrected T/C values versus time. Inoculum: 100 pl/well at a density of 10 cells (39th passage) / microscopic field; preincubation without drugs 44 h:

duration of the experiment 120 h.

from approximately f 20% after short times of incubation (small values for T and C compared to

G)

to It 5% with pro- longed incubation.

SK-OV-3 ovarian cancer cell line. The experiment with the SK-OV-3 cell line (ATCC no. HTB 77)'') were per- formed in the same manner as with NIH: OVCAR-3. Con- trary to this procedure cells in passage 38 were plated at a density of about 7 cells/microscopic field (Leitz Diavert, 320x) for chemosensitivity testing. After 77 h the medium was replaced by fresh medium containing drug or pure sol- vent (control).

Cyrogenetic analysis. The cells were grown to about 50%

confluence on microscopic slides. The slides were prepared

Arch. Pharm. (Weinheini) 325. 93-99 11992)

(7)

1 0 0

A-

U W I

L (-)-3-PtC12 2,s UM

+ ESO 50 UM

0 60 1 2 0 h

-

ts V

\

I- D

0, U

V L W L 0 U

. d

100

60

20

0

Cisplatin 2,5 ~ f l

Cisplatin 2,5 PI

0 60 120 h

Fig. 18 and 19: Effect of (-)-3-PtCI2 (2.5 pM) and cisplatin (2.5 pM) on the proliferation of SK-OV-3 ovarian cancer cells after preincubation with BSO (50 pM, 48 h). Plot of corrected T/C values versus time of drug exposure. Inoculum: 100 pUwell at a density of 10 cells (39th passage) / microscopic field; preincubation without drugs 44 h: duration of the experiment 120 h.

as described”’. So that spindle formation could be inhibited, the slide chambers were inoculated with colcemid solution (Serva) to a final concentration of 0.04 pgml and incubated for 3 h at 37OC. The medium was removed by suction and replaced with 0.075 M KC1. After 30 min of incubation at 37°C an equal volume of cold, freshly made fixative (absol.

methanol/glacial acetic acid, 3/1) was added. This hypo- tonic/fixative mixture was removed immediately and re- placed twice by ice-cold, fresh fixative. The slides were removed from the dish and air-dried at 6OOC. The chromo- somes were stained for 8 min with 10 ml Giemsa plus 90 ml of 0.025 M KH,PO,, pH 6.8.

Doubling rime analysis. Curve fitting of experimental data of the growth curves is accomplished by a polynomal re- gression fit applying the least-squares method”).

References

1 B. Wappes, M. Jennenvein, E. von Angerer, H. Schonenberger, J.

Engel, M. Berger, and K.H. Wrobel, J. Med. Chem. 27,1280(1984).

M. Jennenvein, R. Gust, R. Miiller, H. Schonenberger, J. Engel, M.

Berger, D. S c h m a , S. Seeber, R. Osieka, G. Atassi, and D. Marechal- De Bock, Arch. Pharm. (Weinheim) 322.67 (1989).

R. Miiller, R. Gust, G. Bemhardt, Ch. Keller, H. Schbnenberger, S.

Seeber. R. Osieka, A. Eastman, and M. Jemerwein, J. Cancer Res.

Clin. Oncol. 116.237 (1990).

G. Bemhardt, R. Gust, H.-D. vom Orde, R. Miiller, Ch. Kelier, Th.

SpruB, and H. Schbnenberger, J. Cancer Res. Clin. Oncol., in press.

R.F. Ozols, Th.C. Hamilton, and R.C. Young, Pharmacologic reversal of drug resistance in ovarian cancer: in Multimodal treatment of ovari- an cancer, p. 313, edited by P.F. Conte, N. Ragni, R. Rosso, and J.B.

Vermorken, Raven Press, NY 1989.

R.F. 0zolsandR.C. Young, Semin. Oncol. 11,251 (1984).

R. Miiller, R. Gust, and H. Schonenberger, Chem. Ber., submitted.

T.C. Hamilton, R.C. Young, W.M. Mc Koy, K.R. Grotzinger, J.A.

Green, E.W. Chu, J. Whang-Peng, A.M. Rogan, W.R. Green, and R.F.

Ozols, Cancer Res. 43,5379 (1983).

R.J. Hay,Anal. Biochem. 171,225 (1988).

10 H. Reilk, H. BimMck, G. Bernhardt, Th. SpruB, and H. Schonenber- ger, Anal. Biochem. 187,262 (1990).

11 D.E. Rooney and B.H. Czepulkowski in: Human Cytogenectics, a practical approach, p. 10 (D.E. Rooney and B.H. Czepulkowoski, eds.), IRL Press, OxfordlWashington D.C. 1986.

12 B.T. Hill, D.H. Whelan, E.M. Gibby, D. Sheer, L.K. Hesking, S.A.

Shellard, and H.T. Rupniak, Int. J. Cancer39.219 (1987). [Ph913]

Arch. Pharm. (Weinheim) 325.93-99 (1992)

Referenzen

ÄHNLICHE DOKUMENTE

• This was a randomized controlled trial to assess the outcomes of pri- mary debulking surgery followed by platinum- based chemotherapy compared to platinum- based

obtained fromthe carbene transfer reaction led to the isolation of the previously undetected mono- carbene complex [Pt(Cl) 2 L] (10) which contains only one

The coupling of the tricarboxylic acids, 1,3,5-benzene-tricarboxylic acid, cis,cis-1,3,5-cyclohex- ane-tricarboxylic acid and of citric acid, with rac-N 1 ,N

In continuation of these studies we report in the fol- lowing the coupling of various tricarboxylic acids with the useful reagent 1,2,4-triaminobutane [9, 11] to give

A detailed assignment of the IR spectra (4000-150 cm Ð1 ) of the complexes, supported by an approximate normal coordinate analysis, has been performed. The com- plexes are

This specifically confirmed the induction of apoptosis in the two TRAIL-resistant cell lines PEA-1 and KK as well in the partially sensitive cell line PEA-2 [figure 14-16]

1) The prognostic relevance of the expression of CSC surface markers in CRC clinical specimens. 2) The “in vivo” tumorigenicity of primary CRC derived cells, as related

The results can be summarized as follows: 1) Of the five IRA’s tested, only compound 5 had a significant growth-inhibitory effect on HCT-116 p53wt cells, which in addition was