Human Miillerian Inhibiting Substance Inhibits Tumor...

7
[CANCER RESEARCH 51. 2101-2106. April 15. 1991| Human Miillerian Inhibiting Substance Inhibits Tumor Growth in Vitro and in Vivo1 Taiwai Chin, Robert L. Parry, and Patricia K. Donahoe2 Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, 11217, Republic ofChina [T. CJ; Department of Surgery, National Naval .Medical Center, Bethesda, Maryland 14853 fR. L. P.]; and Pediatrie Surgical Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02II41T.L.R.L.P..P.K.D.I ABSTRACT Mullerian inhibiting substance (MIS) causes regression of the miiller- ian duct in the male fetus. Bovine MIS has been reported to inhibit the growth of some gynecological tumors. Recombinant human MIS (rhMIS) produced in transfected Chinese hamster ovary cells has been highly purified by immunoaffinity chromatography. The introduction of a salt wash prior to elution of MIS from the affinity column removes a growth- stimulating factor(s) derived from Chinese hamster ovary cells. This immunopurified rhMIS caused significant inhibition (34-59% survival) of A431 (a vulvar epidermoid carcinoma), HT-3 (a cervical carcinoma), HEC-l-A (an endometrial adenocarcinoma), NIH:OVCAR-3 (an ovarian adenocarcinoma), and OM431 (an ocular melanoma) human cell lines in colony inhibition assays. Two cell lines, Hep 3B (a hepatocellular carci noma) and RT4 (a bladder transitional cell papilloma), were unresponsive to immunopurified rhMIS. Using an in vivo subrenal capsule assay in irradiated CD-I mice, the growth of A431 and OM431 cells was inhibited by immunopurified rhMIS. We conclude that rhMIS inhibits the growth of certain tumor cell lines in vitro and in vivo. INTRODUCTION The miillerian duct in the female fetus develops into the fallopian tubes, uterus, and upper vagina. MIS' is produced by the fetal testis as a 140-kDa glycosylated disulfide-linked hom- odimer and causes regression of the miillerian duct in the male fetus. The protein can be enzymatically cleaved in vitro into two fragments, composed of dimers with subunits of M, 57,000 and 12,500. Since first described by Jost as a fetal regressor of the miillerian ducts more than 40 years ago, MIS has also been shown to play a role in inhibition of oocyte meiosis (1), testic- ular descent (2), inhibition of fetal lung development (3), inhi bition of autophosphorylation of the EGF receptor (4-5), and inhibition of tumor growth (6-9). The antitumor effect of MIS reported previously was elicited by using natural MIS extracted and partially purified from bovine testes. Since that time the bovine and human MIS genes have been cloned and the human protein has been expressed in CHO cells (10). Initial antipro- liferative studies against established cell lines using this recom binant human MIS (rhMIS) were inconclusive, but some inhib itory effects against primary tumor in vitro were observed (11). Since modifying the purification of rhMIS, the inhibitory effects of both partially and highly purified rhMIS can be demonstrated against established tumor lines both in vitro and in vivo. Received 11/5/90; accepted 2/6/91. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1This work was supported by National Cancer Institute Grant CA 17393 and American Cancer Society Grant PDT63048 to P. K. D. 2To whom requests for reprints should be addressed, at Pediatrie Surgical Research Laboratory, Warren 11. Massachusetts General Hospital. Boston. MA 02114. 'The abbreviations used are: MIS. miillerian inhibiting substance: CHO. Chinese hamster ovary: rhMIS. recombinant human MIS: FCS. fetal calf serum; DG-MIS, serial anión exchange and dye affinity-purified MIS: IAP-MIS, im- munoaffinily-purified MIS; IAP-salt. salt-elutcd fraction from immunoaffinity column: ELISA, enzyme-linked immunosorbent assay; «-MEM+. à -minimal essential medium with ribonucleosides and deoxyribonucleosides; EGF. epidermal growth factor. MATERIALS AND METHODS Production and Purification of rhMIS. After cloning MIS comple mentary DNA and genomic DNA, dihydrofolate reductase-deficient CHO cells were cotransfected with a linear transcript of both the human MIS and the dihydrofolate reducÃ-asegenes (10). The transfected CHO cells were amplified in methotrexate and grown at 37'C in «-minimal essential medium without ribonucleosides and deoxyribonucleosides. supplemented with 10% bovine MIS-free female FCS. Two different MIS purification protocols were used to provide either partially pure DG-MIS (1-10%) or homogeneous (90-95%) IAP-MIS. The first used serial anión exchange and dye affinity chromatography (12), and the second used immunoaffinity chromatography4 and an anti-human MIS monoclonal antibody (13). Previous immunoaffinity protocols resulted in rhMIS preparations that contained low levels of several other pro teins which counteract MIS activity in some in vitro systems. These proteins can be preeluted from the immunoaffinity columns with salt- containing buffers, prior to elution of highly purified rhMIS/ The biological activity of MIS was detected in vitro using the rat müllerianduct regression organ culture assay (14). MIS concentrations were estimated using an ELISA for MIS (13), and protein concentra tions were measured by the method of Bradford (15). Preparation of Monoclonal Antibody for MIS Absorption. Monoclo nal antibody was produced by immunizing female A/J mice (The Jackson Laboratory. Bar Harbor, ME) with immunoaffinity-purified rhMIS by methods previously described for bovine MIS (13, 16). Spleen cells producing anti-MIS antibodies were harvested, and hybridomas were produced as described by Kohler (17). A monoclonal line (6E11) was selected and amplified in Dulbecco's modified essential medium supplemented with 15% FCS. The antibody was precipitated from 6E11 conditioned medium with 50% (NH4hSOj and further purified by protein A-Sepharose CL-4B (Sigma, St. Louis, MO) chro matography. Cell Lines. A431, a cell line derived from a human vulvar epidermoid carcinoma; HT-3 from a human lymph node metastasis of a cervical carcinoma; NIH:OVCAR-3 from a human ovarian adenocarcinoma; HEC-l-A from a human endometrial adenocarcinoma: RT4 from a human bladder transitional cell papilloma; and Hep 3B from a human hepatocellular carcinoma were obtained from the American Type Cul ture Collection. OM431, from a human ocular melanoma, was obtained from Dr. James Epstein of Massachusetts General Hospital. The A431, HT-3, NIH:OVCAR-3, OM431, and Hep 3B cells were maintained with «-MEM+ supplemented with 10% female FCS and 2 m\l I -glutamine. HEC-l-A and RT4 were maintained with McCoy's 5A medium supplemented with 10% female FCS. Cells were passed 1:2 every 2-3 days, and experiments were performed at 70-80% confluency. Fluorescence-activated cell sorting analysis of this population in A431 cells shows a consistent phase distribution, with approximately 70% of the cells in S phase. This proliferating cell population was then centri- fuged at 1500 rpm for 5 min and resuspended with 10% female FCS- supplemented medium. Cells were counted using a hemocytometer. Semisolid Medium (Double-Layer) Colony Inhibition Assay. The ef fects of IAP-MIS and the IAP-salt were tested using A431. HT-3, HEC-l-A, NIH:OVCAR-3, OM431, and Hep 3B cells in the conven tional double-layer agarose colony inhibition assay (18. 19). The un- derlayer of the 35-mm culture dishes contained 1 ml of 0.6% agarose (Sigma) in 10% female FCS-supplemented «-MEM+. The overlayer consisted of 0.3% agarose in 10% female FCS-supplemented «-MEM+, 4 R. C. Ragin et al., manuscript in preparation. 2101 Research. on October 13, 2019. © 1991 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of Human Miillerian Inhibiting Substance Inhibits Tumor...

[CANCER RESEARCH 51. 2101-2106. April 15. 1991|

Human Miillerian Inhibiting Substance Inhibits Tumor Growth in Vitroand in Vivo1

Taiwai Chin, Robert L. Parry, and Patricia K. Donahoe2

Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, 11217, Republic ofChina [T. CJ; Department of Surgery, National Naval .Medical Center,Bethesda, Maryland 14853 fR. L. P.]; and Pediatrie Surgical Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts02II41T.L.R.L.P..P.K.D.I

ABSTRACT

Mullerian inhibiting substance (MIS) causes regression of the miiller-ian duct in the male fetus. Bovine MIS has been reported to inhibit thegrowth of some gynecological tumors. Recombinant human MIS (rhMIS)produced in transfected Chinese hamster ovary cells has been highlypurified by immunoaffinity chromatography. The introduction of a saltwash prior to elution of MIS from the affinity column removes a growth-stimulating factor(s) derived from Chinese hamster ovary cells. Thisimmunopurified rhMIS caused significant inhibition (34-59% survival)of A431 (a vulvar epidermoid carcinoma), HT-3 (a cervical carcinoma),HEC-l-A (an endometrial adenocarcinoma), NIH:OVCAR-3 (an ovarianadenocarcinoma), and OM431 (an ocular melanoma) human cell lines incolony inhibition assays. Two cell lines, Hep 3B (a hepatocellular carcinoma) and RT4 (a bladder transitional cell papilloma), were unresponsiveto immunopurified rhMIS. Using an in vivo subrenal capsule assay inirradiated CD-I mice, the growth of A431 and OM431 cells was inhibitedby immunopurified rhMIS. We conclude that rhMIS inhibits the growthof certain tumor cell lines in vitro and in vivo.

INTRODUCTION

The miillerian duct in the female fetus develops into thefallopian tubes, uterus, and upper vagina. MIS' is produced bythe fetal testis as a 140-kDa glycosylated disulfide-linked hom-odimer and causes regression of the miillerian duct in the malefetus. The protein can be enzymatically cleaved in vitro into twofragments, composed of dimers with subunits of M, 57,000 and12,500. Since first described by Jost as a fetal regressor of themiillerian ducts more than 40 years ago, MIS has also beenshown to play a role in inhibition of oocyte meiosis (1), testic-ular descent (2), inhibition of fetal lung development (3), inhibition of autophosphorylation of the EGF receptor (4-5), andinhibition of tumor growth (6-9). The antitumor effect of MISreported previously was elicited by using natural MIS extractedand partially purified from bovine testes. Since that time thebovine and human MIS genes have been cloned and the humanprotein has been expressed in CHO cells (10). Initial antipro-liferative studies against established cell lines using this recombinant human MIS (rhMIS) were inconclusive, but some inhibitory effects against primary tumor in vitro were observed (11).Since modifying the purification of rhMIS, the inhibitory effectsof both partially and highly purified rhMIS can be demonstratedagainst established tumor lines both in vitro and in vivo.

Received 11/5/90; accepted 2/6/91.The costs of publication of this article were defrayed in part by the payment

of page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1This work was supported by National Cancer Institute Grant CA 17393 andAmerican Cancer Society Grant PDT63048 to P. K. D.

2To whom requests for reprints should be addressed, at Pediatrie Surgical

Research Laboratory, Warren 11. Massachusetts General Hospital. Boston. MA02114.

'The abbreviations used are: MIS. miillerian inhibiting substance: CHO.

Chinese hamster ovary: rhMIS. recombinant human MIS: FCS. fetal calf serum;DG-MIS, serial anión exchange and dye affinity-purified MIS: IAP-MIS, im-munoaffinily-purified MIS; IAP-salt. salt-elutcd fraction from immunoaffinitycolumn: ELISA, enzyme-linked immunosorbent assay; «-MEM+. à-minimalessential medium with ribonucleosides and deoxyribonucleosides; EGF. epidermalgrowth factor.

MATERIALS AND METHODS

Production and Purification of rhMIS. After cloning MIS complementary DNA and genomic DNA, dihydrofolate reductase-deficientCHO cells were cotransfected with a linear transcript of both the humanMIS and the dihydrofolate reducíasegenes (10). The transfected CHOcells were amplified in methotrexate and grown at 37'C in «-minimal

essential medium without ribonucleosides and deoxyribonucleosides.supplemented with 10% bovine MIS-free female FCS. Two differentMIS purification protocols were used to provide either partially pureDG-MIS (1-10%) or homogeneous (90-95%) IAP-MIS. The first usedserial aniónexchange and dye affinity chromatography (12), and thesecond used immunoaffinity chromatography4 and an anti-human MIS

monoclonal antibody (13). Previous immunoaffinity protocols resultedin rhMIS preparations that contained low levels of several other proteins which counteract MIS activity in some in vitro systems. Theseproteins can be preeluted from the immunoaffinity columns with salt-containing buffers, prior to elution of highly purified rhMIS/

The biological activity of MIS was detected in vitro using the ratmüllerianduct regression organ culture assay (14). MIS concentrationswere estimated using an ELISA for MIS (13), and protein concentrations were measured by the method of Bradford (15).

Preparation of Monoclonal Antibody for MIS Absorption. Monoclonal antibody was produced by immunizing female A/J mice (TheJackson Laboratory. Bar Harbor, ME) with immunoaffinity-purifiedrhMIS by methods previously described for bovine MIS (13, 16). Spleencells producing anti-MIS antibodies were harvested, and hybridomaswere produced as described by Kohler (17). A monoclonal line (6E11)was selected and amplified in Dulbecco's modified essential medium

supplemented with 15% FCS. The antibody was precipitated from6E11 conditioned medium with 50% (NH4hSOj and further purifiedby protein A-Sepharose CL-4B (Sigma, St. Louis, MO) chromatography.

Cell Lines. A431, a cell line derived from a human vulvar epidermoidcarcinoma; HT-3 from a human lymph node metastasis of a cervicalcarcinoma; NIH:OVCAR-3 from a human ovarian adenocarcinoma;HEC-l-A from a human endometrial adenocarcinoma: RT4 from ahuman bladder transitional cell papilloma; and Hep 3B from a humanhepatocellular carcinoma were obtained from the American Type Culture Collection. OM431, from a human ocular melanoma, was obtainedfrom Dr. James Epstein of Massachusetts General Hospital.

The A431, HT-3, NIH:OVCAR-3, OM431, and Hep 3B cells weremaintained with «-MEM+ supplemented with 10% female FCS and 2m\l I -glutamine. HEC-l-A and RT4 were maintained with McCoy's

5A medium supplemented with 10% female FCS. Cells were passed 1:2every 2-3 days, and experiments were performed at 70-80% confluency.Fluorescence-activated cell sorting analysis of this population in A431cells shows a consistent phase distribution, with approximately 70% ofthe cells in S phase. This proliferating cell population was then centri-fuged at 1500 rpm for 5 min and resuspended with 10% female FCS-supplemented medium. Cells were counted using a hemocytometer.

Semisolid Medium (Double-Layer) Colony Inhibition Assay. The effects of IAP-MIS and the IAP-salt were tested using A431. HT-3,HEC-l-A, NIH:OVCAR-3, OM431, and Hep 3B cells in the conventional double-layer agarose colony inhibition assay (18. 19). The un-derlayer of the 35-mm culture dishes contained 1 ml of 0.6% agarose(Sigma) in 10% female FCS-supplemented «-MEM+. The overlayerconsisted of 0.3% agarose in 10% female FCS-supplemented «-MEM+,

4 R. C. Ragin et al., manuscript in preparation.

2101

Research. on October 13, 2019. © 1991 American Association for Cancercancerres.aacrjournals.org Downloaded from

MÃœLLERIAN INHIBITING SUBSTANCE ANTITUMOR EFFECT

the cells to be tested (50,000 cells/ml for A431, HT-3, and OM431; cell clot thus formed was cut into approximately 100 fragments (125,000 cells/ml for HEC-l-A. NIH:OVCAR-3, and Hep 3B), 10 ng/ml EGF (Sigma), and one of the following: IAP-MIS (final concentration, 30 nM); IAP-salt (final protein concentration, 19.3 ng/ml); orvehicle buffer as a negative control. The dishes were incubated in humidair with 5% CO2 at 37°Cfor 10-21 days. Colonies with more than 30

cells were counted with an inverted microscope (Nikon). The resultsare expressed as percentage survival relative to a control group

No. of colonies in the test group x 100no. of colonies in the control group

Liquid Medium Colony Inhibition Assay. Single-cell suspensions wereplaced and grown in 24-well culture plates (no. 3047; Falcon, Oxnard,CA). After cell attachment, only those with good single-cell dispersionwithout clumping were used for further study. Agents to be tested wereadded in a volume less than 1/10 of the total volume in a well and weretested in triplicate. The cells were incubated in humid air with 5% CO2at 37°C.Colonies that formed in 5-7 days were stained with Giemsa

solution, and those with more than 30 cells were counted with aninverted microscope or by a computer-based image analyzer.5 The assay

was used to compare the response of different cell lines to variouspreparations of rhMIS and to establish a dose-response curve to rhMIS

in A431 cells.Antibody Absorption of MIS. MIS monoclonal antibody (6E11) and

normal rabbit IgG (Lot 108; Dako Corporation, Denmark) were di-alyzed into serum-free «-MEM+ before use. Previous experimentsshowed maximum absorption of MIS activity at a MIS:antibody ratioof 1:3 (13). Therefore, 17.4 Mgof 6E11 were added to 5.6 Mgof DG-MIS. Normal rabbit IgG was diluted with culture medium and addedto MIS in the same 1:3 ratio to determine nonspecific absorption. Anequivalent amount of protein purified from conditioned medium ofuntransfected wild-type CHO cells served as a negative control whenmixed 1:3 with antibody as above. The preparations were mixed at 4°C

for 12 h. Protein A-Sepharose CL-4B (Sigma), after being washed inserum-free medium, was added and incubated at 4°Cfor another 12 h.

The mixtures were centrifuged and the supernatants were tested in theliquid medium colony inhibition assay using A431 cells (25,000 cells/ml; EGF, 50 ng/ml). The percentage survival of each group wascalculated by comparing the number of colonies in each group to thewild-type negative control.

Multicellular Tumor Spheroid Assay. Multicellular tumor spheroidsof HT-3 and Hep 3B cells were produced by the method described byYuhas et al. (21). In brief, IO6cells of either HT-3 or Hep 3B in 10 mlof 10% female FCS-supplemented a-MEM+ were plated on the top of1% agarose in a 10-cm culture dish and incubated in humid air with5% CO2 at 37°C.Spheroids usually formed in 2-5 days. Agarose (0.5

ml of 1%) was added to each well of a 24-well culture plate (Falcon) toform a bottom layer before use. Individual spheroids of similar size(approximately 250 pm diameter) were selected under a dissectingmicroscope and transferred by a micropipet at 1 spheroid/well containing 0.5 ml of 10% female FCS-supplemented «-MEM+ on top of theagarose layer. The sizes of the spheroids on day 0 were measured bythe longest diameter (L) and the diameter perpendicular to the longestone ( H7);the volume was then expressed as L x W x W. Six spheroid-containing wells of each cell type were treated by either DG-MIS (finalconcentration, 7 nM) or vehicle buffer. Volumes were measured againat the 3rd, 6th, and 9th days. The size ratio of each spheroid at differentintervals was obtained by comparing it to the size of the same spheroidat day 0. The average size ratio of each group was plotted versus timein a growth curve and compared to the other groups.

Subrenal Capsule Assay. Following the method of Bogden (22), whichwas later modified by Fingert (23), A431 and OM43I cells were tested.The cells (IO7) were centrifuged at 1500 rpm for 5 min to form a pellet.Fibrinogen (15 M') (Sigma; 20 mg/ml dissolved in phosphate-bufferedsaline, pH 7.4) was added to the pellet, followed by 8 /jl of thrombin(Sigma; 20 units/ml dissolved in double-strength Dulbecco's modifiedessential medium). The mixture was incubated at 37°Cfor 15 min. The

' R. L. Parry, T. Chin, and P. K. Donohoe, Biotechniques, in press, 1991.

mm1, each containing approximately IO5 cells) in preparation for

implantation.MIS was delivered by an Alzet mini-osmotic pump (no. 2001; Alza,

Palo Alto, CA) placed in the peritoneal cavity at the time of tumorimplantation. These pumps have a fill volume of 209 ±6 (SD) n\ andrelease their contents at a rate of 1.03 ±0.04 ¿il/hfor approximately 8days of delivery time. The pumps were filled either with IAP-MIS(MIS, 159 /jg/ml by ELISA) or with vehicle buffer. A total MIS doseof approximately 33 ^g (230 nM) was given to each mouse in the MISgroup over the course of the experiment.

Virus- and pathogen-free female CD-I mice (10 weeks old; averageweight, 3.5 g; Charles River Breeding Laboratory, Wilmington, MA)were given whole-body irradiation of 640 rads by a Mark-1 137Csirradiator 16-24 h before the experiment (24). After induction ofanesthesia with an i.p. injection of 0.3 ml of 10% pentobarbital (AbbottLaboratory, North Chicago, IL), an incision was made in the left flankof the mouse, and the left kidney was exteriorized. A subcapsular spacewas developed with a 19-gauge needle trocar. A cell clot was introducedinto the space with a segment of 5-0 nylon suture (approximately 1 mmlong), which was used both to calibrate ocular micrometer measurements and to localize the tumor. Twenty-four mice were implantedwith A431 cell clots and 12 mice with OM431 cell clots. The longestdiameter (/.,) of the implant, the one perpendicular to the longest one(Wi), and the length of the suture were measured with the ocularmicrometer of a dissecting microscope. The animals were treated byeither IAP-MIS or vehicle buffer delivered by the Alzet pumps placedin the peritoneal cavity. Blood samples at 6, 24, 48, 120 (fifth day), and192 h (eighth day) were obtained from selected animals by orbitalbleeding, and serum MIS levels were measured by ELISA. The animalswere sacrificed on the eighth day. The longest diameter (L2) of thetumor, the one perpendicular to the longest one (H^), and the lengthof the suture were measured blindly by two independent investigators.After calibration of the measurements, the graft size ratio was represented by

L2 x W2 x W2Lt x Wi x Wi

Histológica! sections of the kidneys were also obtained and examined.Tumors with cystic change were excluded.

Statistical Analysis. The results of the liquid medium and semisolidmedium colony inhibition assays were analyzed by the x2 test with or

without Yates correction, while the multicellular spheroid and thesubrenal capsule assays were tested by Student's f test (P < 0.05 was

considered statistically significant).

RESULTS

Semisolid Medium (Double-Layer) Colony Inhibition Assay.The percentage survival of the various cell lines after incubationwith 30 nM of IAP-MIS was 45% for A431, 47% for HT-3,54% for HEC-l-A, 59% for NIH:OVCAR-3, 34% for OM431,and 114% for Hep 3B. When compared to controls, the survivalafter treatment with MIS in all cell lines except Hep 3B wassignificantly inhibited by IAP-MIS (P < 0.05). The growth ofHep 3B was not inhibited by MIS. The percentage survival afterincubation with IAP-salt was 172% for A431, 93% for HT-3,92% for HEC-l-A, 120% forOM431, 105% for NIH:OVCAR-3, and 173% for Hep 3B. The stimulatory effect was significantfor A431, OM431, and Hep 3B cells (P < 0.05) (Fig. 1).

Effect of IAP-MIS and IAP-salt on Colony Formation, by theLiquid Medium Colony Inhibition Assay. The percentage survival was 55.9% for A431, 36.8% for OM431, 100.5% for HT-3, and 115.8% for RT4 when treated with 30 nM IAP-MIS.The percentage survival was 169.0% for A431, 226.7% forOM431, 101.6% for HT-3, and 96.5% for RT4 when treatedwith IAP-salt (final protein concentration, 19.3 ¿¿g/ml).The

2102

Research. on October 13, 2019. © 1991 American Association for Cancercancerres.aacrjournals.org Downloaded from

MÃœLLER1ANINHIBITING SUBSTANCE ANTITUMOR EFFECT

Semisolid Medium Colony Inhibition Assay

Fig. 1. Semisolid medium (double-layer) colony inhibition assay. Left, effectof IAP-MIS. Colony formation of A431, HT-3, HEC-l-A, NIH:OVCAR-3, andOM431 cells was significantly inhibited by IAP-MIS (30 nM). Right, effect of thesalt fraction preeluted from the immunoaffinity column (IAP-salt.). Stimulationof A431, OM431, and Hep 3B colony formation was seen when treated with thesalt fraction. *, P < 0.05 when compared with control.

Liquid Medium Colony Inhibition Assay*

_>°>1_

3w

S?240-220-200-180-160-140-120-100-80-60-LiquKControlHH

*

20

Fig. 2. Liquid medium colony inhibition assay. Left, effect of IAP-MIS. Thecolony formation of A431 and OM431 cells was significantly inhibited by IAP-MIS (30 nM). Right, effect of the salt fraction pre-eluted from the immunoaffinitycolumn (IAP-salt). The colony formation of A431 and OM431 cells was stimulated by IAP-salt. IAP-MIS and IAP-salt showed no effect on HT-3 and RT4cells. The result was expressed as percentage survival relative to the vehicle buffercontrol (*,/•<0.05). Cell concentrations were as follows: A431, 25,000 cells/ml;OM431, 25,000 cells/ml; HT-3, 5,000 cells/ml; and RT4. 5,000 cells/ml. Forcolony growth in the OM431 and A431 cell lines, EGF (50 ng/ml) was required.

inhibitory effect of IAP-MIS and the stimulatory effect of IAP-salt were significant for A431 and OM431 colony formation (P< 0.05) (Fig. 2).

Dose-dependent Inhibition of A431 Colony Formation by MIS.The percentage survival was 103.4, 81.4, 61.3, and 26.5%,respectively, for DG-MIS concentrations of 0.9, 1.8, 3.5, and7.0 nM. Significant inhibitions were seen with DG-MIS concentrations of 3.5 and 7.0 nM (P < 0.05). The percentage survival

was 109.7, 71.1, 56.6, and 33.3%, respectively, for IAP-MISconcentrations of 12, 24, 48, and 96 nM. Significant inhibitionswere seen at IAP-MIS concentrations of 24, 48, and 96 nM(Fig. 3). DG-MIS thus was 10-14 times more potent than IAP-MIS.

Antibody Absorption of MIS. The MIS levels, measured byELISA for MIS, were 9.0 nM for the positive control (MISalone), 8.8 nM after normal IgG absorption (nonspecific), and0.62 nM after the monoclonal antibody absorption (specific).All the wild-type negative controls had an MIS level equal to0. The percentage survival was 42.9% for MIS alone, 38.0%for normal IgG (nonspecific absorption), and 74.2% for themonoclonal antibody (specific absorption). There was no significant nonspecific absorption of MIS activity (P > 0.05 compared with negative control). The percentage survival after themonoclonal antibody absorption was significantly higher thanthat of positive control (MIS alone) and normal IgG (Fig. 4).

The Salt-eluted Fraction (IAP-salt) Inhibits IAP-MIS. Thepercentage survival of A431 cells was 51.3% for IAP-MIS alone

Dose Dependent Inhibition by MIS

0.9 48 96

MIS concentration (nM)Fig. 3. Dose-dependent inhibition of A431 colony formation by MIS. Left,

DG-MIS tested in increasing doses with the liquid medium colony inhibitionassay. Significant inhibition was seen at MIS concentrations of 3.5 and 7.0 nM.At right, IAP-MIS caused significant inhibition at concentrations of 24, 48, and96 n\i. *, P < 0.05 when compared with buffer control.

100-

80

3CO

Antibody Absorption of MIS activity

MIS alone MIS+normal IgG MIS+monoclonal Ab

Fig. 4. Antibody absorption of DG-MIS. The inhibitory effect of MIS on A431colony formation was significantly reduced after absorption with the MIS-specificmonoclonal antibody (6EI1), but not after nonspecific absorption by normal IgG.*,P< 0.05 when compared with MIS alone.

2103

Research. on October 13, 2019. © 1991 American Association for Cancercancerres.aacrjournals.org Downloaded from

MLILLERIAN INHIBITING SUBSTANCE ANTITUMOR EFFECT

120 n

100 -

80-

Inhibition MIS Effect by IAP-sa!t

^

i

lAP-salt IAP-MIS

abc

Combination

B

31

22

14Fig. 5. A, effects of various ¡mmunopurified fractions on A431 cells (25,000

cells/ml; EGF. 50 ng/ml) in the liquid colony inhibition assay. IAP-MIS wasmixed with an equal volume of IAP-salt (protein concentration. 0.199 mg/ml) togive a final M IS concentration of 50 nM. The IAP-MIS. aftersalt and acid elution,inhibited colony growth, whereas the salt-eluted fraction stimulated growth. Thecombination of these two preparations reduces MIS-inhibitory effect. The percentage survival of the combination was significantly higher than that of MISalone. *, P < 0.05 when compared with IAP-MIS alone, although the MISconcentrations were the same. B, sodium dodecyl sulfate gel electrophoresis afterreduction of immunopurified preparations. Lane a, salt-eluted fraction; Lanes band c, two separate IAP-MIS (preparations 180 and 181) after both salt and acidelution. Holo MIS reduced is recognized at M, 70.000 and its cleavage productsat M, 57,000 and 12,500. Bands of protein from M, 14.000 to M, 43,000, presentin the salt-clutcd fraction, were absent in IAP-MIS.

(50 HM), 116.9% for IAP-salt alone, and 81.6% for the combination of these two (MIS, 50 nM). The difference between IAP-MIS alone and the combination of IAP-MIS/IAP-salt wassignificant (P < 0.05) (Fig. 5A). Fig. 5B shows the sodiumdodecyl sulfate gel electrophoresis after reduction of IAP-saltand two separate preparations of IAP-MIS. Several bands ofprotein in the IAP-salt were seen in the region of Mr 14,000-43,000 but were absent in the IAP-MIS.

Mull ¡cellularSpheroid Assay. The average size ratios of HT-3 spheroids in the control group (n = 6) were 1.15 ±0.04, 1.44±0.02, and 1.96 ±0.11 at days 3, 6, and 9, respectively, whilein the MIS group (n = 6) they were 0.98 ±0.04, 0.94 ±0.04,and 1.08 ±0.06 (Fig. 6A). The average size ratios of Hep 3Bspheroids in the control group (n = 6) were 2.56 ±0.05, 5.64±0.53, and 13.07 ±1.09, respectively, at days 3, 6, and 9,while in the MIS group (n = 6), they were 2.36 ±0.25, 5.77 ±0.54, and 14.30 ±0.54. The growth of Hep 3B spheroids wasfaster and uninhibited by MIS (Fig. 6B).

Subretta! Capsule Assay. The MIS levels in mouse serumwere relatively stable from 24 h to the eighth day after theimplantation of the MIS-filled Alzet pumps (Fig. 7/4). Theaverage MIS level of 14 blood samples from day 2 to day 8 was19.1 ±2.7 ng/ml (approximately 140 pM). The serum MISlevels of the control mice were undetectable. The graft size ratioof the implanted A431 tumor was 3.70 ±0.31 in the controlgroup (n = 12) versus 1.50 ±0.26 in the MIS group (n = 12).The graft size ratio of the OM431 tumor was 3.93 ±0.49 inthe control (n = 7) versus 1.42 ±0.44 in the MIS group (n =5). The growth of the tumors in the MIS group was significantlylower than the control in both cell lines (P < 0.05) (Fig. IB).

DISCUSSION

In the male fetus, MIS causes the regression of the müllerianduct, which is the anlagen of the fallopian tubes, uterus, cervix,and upper vagina. The müllerianduct is formed by an invagination of the coelomic epithelium overlying the genital ridge.The serosal surface of the normal ovary, the tissue of origin ofthe majority of primary ovarian tumors, is also derived fromthis same coelomic epithelium. We hypothesized that MISmight suppress the growth of malignant tumors derived frommüllerianduct and hence coelomic epithelial structures (6).Using MIS partially purified from bovine testes we demonstrated an inhibitory effect on endometrial and ovarian carcinoma cell lines both in vitro and in vivo (7, 8). This antitumoreffect was also seen in a large series of primary human coelomicepithelium/müllerian duct-derived tumors in vitro using highlypurified bovine MIS (9). The human MIS gene has since beencloned, expressed, and amplified in CHO cells (10) so thatsufficient human MIS has become available for testing. Thus,rhMIS from the conditioned media of the amplified CHO cellline, purified by either DG-MIS or IAP-MIS, was examinedagainst various cell lines in a number of growth assays.

The reliable and reproducible double-layer colony inhibitionassay was used, but the lengthy incubation time (10-21 days)and the relatively large quantity of sample required for assessment proved to be disadvantageous. To overcome some of thesedifficulties, a more rapid (5-7-day) liquid medium colony inhibition assay using less test material was developed. Whilesome cell lines failed to grow colonies in the liquid mediumcolony inhibition assay, when so formed their counting can beautomated.5 OM431 and A431 required EGF for colony formation. This EGF-dependent colony formation is paradoxicalin view of the fact that EGF is known to inhibit the A431 cellin monolayer (20). Overall, the results of the liquid and semi-solid assays were quite similar, with the exception of the HT-3(cervical carcinoma) cell line, which was inhibited by highlypurified MIS in the semisolid assay but not in the liquid assay.The multicellular spheroid assay was used to recapitulate tumormicroregions with cell-cell interactions and nutrient-affectedgrowth patterns (25, 26). HT-3 cells formed satisfactory spheroids that were inhibited by MIS. No MIS effect was noted onthe spheroid growth of Hep 3B (hepatocellular carcinoma). Theavailability of all three in vitro assays permits selection ofoptimal conditions for each tumor.

Not all cell lines respond to MIS. Initial studies includedonly cell lines and tumors derived from tissue of coelomicepithelium/müllerian duct origin, in which the majority wereinhibited by MIS (6-9). As we expanded our investigations toevaluate tumor cell lines unrelated to müllerian-derivedtissue,we found that a vulvar epithelial and an ocular melanoma tumor

2104

Research. on October 13, 2019. © 1991 American Association for Cancercancerres.aacrjournals.org Downloaded from

MÃœLLERIAN INHIBITING SUBSTANCE ANTITUMOR EFFECT

Fig. 6. Spheroid assay. DC-MIS (7 n.\i) inhibited the growth of HT-3 spheroids whencompared with control (P< 0.05) (A) but didnot affect the growth of Hep 3B spheroids (B).

HT-3 Spheroids Hep 3B spheroids

—O— Control"— MIS

day 0 day3 day6Time

day9 day 0 day3 day6Time

d.iy'3

cell line were also inhibited by MIS, thereby suggesting thatother diverse tumor lines may have sensitivity to MIS.

DG-MIS, although less purified, consistently showed inhibition of cell growth in vitro. The specificity of the MIS effectwas demonstrated by blocking the inhibitory effect with an MISmonoclonal antibody (13). Recent findings suggest that MIS isprocessed by at least one requisite posttranslational proteolyticcleavage and that both the amino- and carboxy-terminal fragments are required for full biological activity (27, 28). Themonoclonal antibody to holo-MIS thus may not recognize allMIS moieties present in the preparations, which could accountfor the incomplete inhibition of the response.

Wallen et al. (11) reported only a minimal antiproliferativeactivity when an immunopurified MIS preparation was testedagainst a variety of established cell lines. We observed the samepoor response with IAP-MIS before the salt elution step wasadded prior to elution with l M acetic acid. In addition, the saltfraction eluted from the immunoaffinity column actuallyshowed a growth-stimulating effect in some cell lines. Electro-phoresis of this salt fraction shows several bands in the regionof M, 14,000-43,000, which are absent in the fraction subsequently eluted by 1 M acetic acid (Fig. 5B). When the concentrated culture medium of untransfected wild-type CHO cellswas subjected to the same purification process, the salt fraction,which also showed a stimulatory effect on A431 cells (Figs. 1and 2), revealed the presence of the same bands at M, 14,000-43,000. These protein products of the untransfected CHO cellsare being studied as putative growth-stimulating factors thatmight serve to mask the MIS effect. This hypothesis wassupported when the salt-eluted fraction added to the acetic acid-

eluted fraction altered the MIS inhibition (Fig. 5/1). Transforming growth factors and other polypeptides are known to be

produced as autocrine growth factors by certain malignant cells(29-32).

When the dose responses to highly purified IAP-MIS andDG-MIS were compared, it was noted that DG-MIS was 10-fold more potent than the more highly purified IAP-MIS. It ispossible that purification of the cleaner preparation may haveremoved the proteases required to cleave and activate MIS.Furthermore, irreversible aggregation may occur; to reducethese effects, EDTA and a nontoxic concentration of NonidetP-40 (0.001%) were added to the eluted IAP-MIS. In addition,the relatively harsh immunoaffinity column elution protocolsmay denature a binding protein essential for the antiproliferative effect of MIS. Such binding proteins have been shown tobe essential for the activity of other biological systems (32).

The subrenal capsule assay was used to test the effect of MISin vivo. By delivering MIS via a constant i.p. infusion Alzetpump inserted at the time of tumor implantation, the growthof a vulvarepidermoid carcinoma cell line, A431, and an ocularmelanoma line, OM431, were inhibited in vivo. It is essentialin evaluating this assay that histology be documented for eachdifferent tumor and that the experiment be completed beforecentral necrosis occurs. Thus the assay was terminated on day8, since longer duration led to variable cystic change. Suchchanges can vary the graft size ratio enough to make comparisons unreliable due to imbibition of fluid and cystic changes.By this careful analysis, the postimplantation characteristics ofeach tumor cell line can be established, and artifacts can beavoided. The in vivo effect in this subrenal assay is achieved atphysiological or picomolar concentrations, as was also observedin vivo in suppression of lung surfactant in late embryo rats (3).In vitro studies, on the other hand, require 50- to 500-foldhigher levels, which may reflect failure of activation.

Fig. 7. A, MIS serum levels. Alzet pumpsloaded with 33 t¡gMIS were implanted in CD-I mice. A relatively constant level of MIS wasachieved 24 h after implantation. B, inhibitionof tumor growth in vivo by MIS. A431 andOM431 cells were implanted in the subrenalcapsule space of CD-I mice. The graft sizeratios of both tumors were significantly inhibited in the MIS-lreated group. *. P < 0.05

when compared with controls.

E 20-

100 150

Time (hours)

2105

A431 OM431

Research. on October 13, 2019. © 1991 American Association for Cancercancerres.aacrjournals.org Downloaded from

MÃœLLER1AN INHIBITING SUBSTANCE ANTITUMOR EFFECT

Although we do not yet know the maximum tolerable doseof systemic MIS, no obvious toxicity to animals was observed.Much must be done to understand the mechanism of action ofMIS, but the in vivo environment appears to create the conditions appropriate for MIS to exert its maximal antiproliferativeeffects. The results of these experiments encourage continuedevaluation of this biological modifier as an effective chemo-therapeutic agent.

REFERENCES

1. Takahashi. M . Koidc, S. S.. and Donahoe. P. K. Mulini.m inhibitingsubstance as oocyte meiosis inhibitor. Mol. Cell. Endocrinol.. 47: 225-234.1986.

2. Hutson. J. M., and Donahue. P. K. The hormonal control of tcsticulardesceñí.Endocr. Rev.. 7: 270-283. 1986.

3. C'atlin. E., Manganaro. T.. and Donahoe. P. K. Mullerian inhibiting sub

stance depresses accumulation in vitro of disaturated phosphatidylcholine infetal rat lung. Am. J. Obstet. Gynecol.. 159: 1299-1303. 1988.

4. Coughlin. J. P.. Donahoe. P. K.. Budzik. Ci. P.. and MacLaughlin. D. T.Mullerian inhibiting substance blocks autophosphorylation of the EGF receptor by inhibiting tyrosine kinase. Mol. C'ell. Endocrinol.. 49: 75-86. 1987.

5. Cigarroa. F. G.. Coughlin. J. P., Donahoe. P. K.. White. M. F.. Uitvlugt. N..and MacLaughlin. D. T. Recombinant human mullerian inhibiting substanceinhibits epidermal growth factor receptor tyrosine kinase. Growth Factors./: 179-191. 1989.

6. Donahoe. P. K.. Swann. D. A., Hayashi. A., and Sullivan. M. D. Mullerianduct regression in the embryo correlated with cytotoxic activity againsthuman ovarian cancer. Science (Washington DC). 205: 913-915, 1979.

7. Donahoe. P. K.. Fuller. A. F., Jr.. Scully. R. E., Guy, S. R., and Budzik. Ci.P. Mullerian inhibiting substance inhibits growth of a human ovarian cancerin nude mice. Ann. Surg.. 194:472-480. 1981.

8. Fuller. A. F., Jr., Guy. S. R.. Budzik. G. P.. and Donahoe. P. K. Mullerianinhibiting substance inhibits colony growth of a human ovarian cell line. J.Clin. Endocrinol. Metab.. 54: 1051-1055. 1982.

9. Fuller. A. F.. Jr., Krane, I. M., Budzik. G. P.. and Donahoe, P. K. Mullerianinhibiting substance reduction of colony growth of human gynecologic cancers in a stem cell assay. Gynecol. Oncol.. 22: 135-148. 1985.

10. Cate. R. L., Mattaliano. R. J.. Hession. C.. Tizard. R.. Farber. N. M.. Chang.A.. Ninfa, E. G., Frey. A. Z.. Gash. D. J.. Torres. G., Wallner. B. P.,Ramachandran. K. L.. Ragin. R. C.. Manganaro. T. F.. MacLaughlin. D. T..and Donahoe. P. K. Isolation of the bovine and human genes for mullerianinhibiting substance and expression of the human gene in animal cells. Cell.45:685-698. 1986.

11. Wallen. J. W.. Cate. R. L.. Kiefer. D. M.. Riemen. M. K.. Martinez. D..Hoffnar. R. M.. Donahoe. P. K., von Hoff. D. D.. Pepinsky. B.. and Oliff.A. Minimal antiproliferative effect of recombinant mullerian inhibiting substance on gynecological tumor cell lines and tumor expiants. Cancer Res..49:2005-2011. 1986.

12. Budzik. G. P., Powell. S. M.. Kamagata. S.. and Donahoe. P. K. Mullerianinhibiting substance fractionation by dye affinity chromatography. Cell. 34:307-314. 1983.

13. Hudson. P. L.. Dougas. I., Donahoe, P. K.. Cale. R. L.. Epstein. J.. Pepinsky.R. B.. and MacLaughlin. D. T. An immunoassay to detect human mullerianinhibiting substance in males and females during normal development. J.Clin. Endocrinol. Metab.. 70: 16-22. 1990.

14. Donahoe, P. K., Ito, Y., Marfatia, S., and Hendren. W. H. A graded organculture assay for the detection of mullerian inhibiting substance. J. Surg.Res.. 23: 14Ì-I48.1977.

15. Bradford. M. M. A rapid and sensitive method for the quantitation ofmicrogram quantities of protein utilizing the principle of protein dye binding.Anal. Biochem.. 72: 248-254, 1976.

16. Mudgett-Hunter, M., Budzik. G. P., Sullivan, H.. and Donahoe. P. K.Monoclonal antibody to mullerian inhibiting substance. J. Immunol.. 128:1327-1333. 1982.

17. Kohler, G. The technique of hybridoma production. Immunol. Methods, 2:285-298. 1981.

18. Hamburger. A. W., and Salmon, S. E. Primary bioassay of human tumorstem cells. Science (Washington DC), 197:461-463. 1977.

19. Hamburger, A. W., and Salmon. S. E. Primary bioassay of human myelomastem cells. J. Clin. Invest., 60: 846-854. 1977.

20. Lee. K.. Tanaka. M.. Hatanaka. M., and Kuze, F. Reciprocal effects ofepidermal growth factor and transforming growth factor tf on the anchorage-dependent and -independent growth of A431 epidermoid carcinoma cells.Exp. Cell Res.. 173: 156-162, 1987.

21. Vuhas, J. M., Li. A. P., Martinez, A. O., and Ladman, A. J. A simplifiedmethod for production and growth of multicellular tumor spheroids. CancerRes.. 37: 3639-3643. 1977.

22. Bogden. A. Z., Haskell, P. M., LePage, D. J.. Kelton, D. E., Cobb, W. R.,and Esber, H. J. Growth of human tumor xenografts implanted under therenal capsule of normal immunocompetcnt mice. Exp. Cell Biol.. 47: 281-293. 1979.

23. Fingert, H. J.. Chen. Z., Mizrahi, N., Gajewski. W. H.. Bamberg, M. P.. andKradin. R. L. Rapid growth of human cancer cells in a mouse model withfibrin clot subrena! capsule assay. Cancer Res.. 47: 3824-3829. 1987.

24. Gajewski, W. H., Fingert, H. J., Fuller. A. F.. Jr.. and Donahoe. P. K. Wholebody irradiation improves growth and histology of human tumor grafts inthe mouse subrenal capsule assay. Surg. Forum, 38: 468-470. 1987.

25. Sutherland. R. M., McCredie. J. A., and Such. W. R. Growth of multicelispheroid in tissue culture as a model of nodular carcinomas. J. Nati. CancerInst., 46: 113-120, 1971.

26. Sutherland. R. M. Cell and environment interactions in tumor microregions:the multiceli spheroid model. Science (Washington DC). 240:177-184,1988.

27. Pepinsky. R. B.. Sinclair. L. K., Chow, E. P., Mattaliano, R. J.. Manganaro.T. F.. Donahoe, P. K., and Cate, R. L. Protcolytic processing of mullerianinhibiting substance produces a transforming growth factor-/i-like fragment.J. Biol. Chem., 263: 18961-18964. 1988.

28. Cate, R. L., Donahoe. P. K.. and MacLaughlin, D. T. Mullerian inhibitingsubstance. In: M. B. Sporn and A. B. Roberts (eds.). Handbook of Experimental Pharmacology. Vol. 95/11. pp. 179-210. Berlin: Springer-Verlag.1990.

29. Todaro, G. J.. and De Larco, J. E. Growth factors produced by sarcomavirus-transformed cells. Cancer Res.. 38: 4147-4154. 1978.

30. Todaro. G. J., Frying, C., and De Larco, J. E. Transforming growth factorsproduced by certain human tumor cells: polypeptides that interact withepidermal growth factor receptors. Proc. Nati. Acad. Sci. USA, 77: 5258-5262. 1980.

31. Marquardt. H.. Hunkapiller, M. W.. Hood, L. E., Twardzik. D. R.. De Larco.J. E., Stephenson, J. R.. and Todaro, G. J. Transforming growth factorsproduced by retrovirus-transformed rodent fibroblasts and human melanomacells: amino acid sequence homology with epidermal growth factor. Proc.Nati. Acad. Sci. USA, «0:4684-4688. 1983.

32. Richmond, A.. Lawson. D. H., Nixon. D. W.. and Chawla. R. K. Characterization of autostimulatory and transforming growth factors from humanmelanoma cells. Cancer Res., 45:6390-6394. 1985.

33. Schumann. R. R.. Leong. S. R.. Flaggs. G. W.. Gray. P. W., Wright, S. D.,Mathison, J. C., Tobias, P. S., and Ulevitch, R. J. Structure and function oflipopolysaccharide binding protein. Reports. 21: 1429-1431, 1990.

2106

Research. on October 13, 2019. © 1991 American Association for Cancercancerres.aacrjournals.org Downloaded from

1991;51:2101-2106. Cancer Res   Taiwai Chin, Robert L. Parry and Patricia K. Donahoe 

in Vivo and VitroinHuman Müllerian Inhibiting Substance Inhibits Tumor Growth

  Updated version

  http://cancerres.aacrjournals.org/content/51/8/2101

Access the most recent version of this article at:

   

   

   

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

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/51/8/2101To request permission to re-use all or part of this article, use this link

Research. on October 13, 2019. © 1991 American Association for Cancercancerres.aacrjournals.org Downloaded from