Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PROBE
Document Type and Number:
WIPO Patent Application WO/1988/005054
Kind Code:
A1
Abstract:
An antibody or fragment thereof against a human mucin core protein which antibody or fragment has reduced or substantially no reaction with fully expressed human mucin glycoprotein.

Inventors:
TAYLOR-PAPADIMITRIOU JOYCE (GB)
GENDLER SANDRA (GB)
BURCHELL JOY (GB)
Application Number:
PCT/GB1988/000011
Publication Date:
July 14, 1988
Filing Date:
January 07, 1988
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
IMP CANCER RES TECH (GB)
International Classes:
A61K38/00; A61K39/395; A61K49/00; A61P35/00; C07K1/113; C07K1/22; C07K14/00; C07K14/435; C07K14/47; G01N33/53; C07K14/82; C07K16/00; C07K16/30; C07K16/32; C07K16/44; C07K19/00; C12N5/10; C12N5/20; C12N15/02; C12N15/09; C12P21/00; C12P21/08; C12Q1/68; G01N33/574; G01N33/577; C12R1/91; (IPC1-7): C07K15/00; C07K7/00; C12N5/00; C12P21/00; A61K39/395; G01N33/574; C07H21/00; C12Q1/68
Other References:
Proc. Natl. Acad. Sci, USA, volume 84, September 1987, S.J. Gendler et al.: "Cloning of partial cDNA encoding differentiation and tumor-associated mucin glyco-proteins expressed by human mammary epithelium", pages 6060-6064
Nature, volume 328, 2 July 1987, D.M. Swallow et al.: "The human tumor-associated epithelial mucins are coded by an expressed hypervariable gene locus PUM", pages 82-84
Chemical Abstracts, volume 106, no. 19, 11 May 1987, (Columbus, Ohio, US), D.M. Swallow et al.: "Detection of the urinary 'PUM' polymorphism by the tumor-binding monoclonal antibodies Cal, Ca2, Ca3, HMFG1, and HMFG2", see page 513
Chemical Abstracts, volume 104, no. 17, 28 April 1986, (Columbus, Ohio, US), N. Mian et al.: "Characterization of core polypeptides of human bronchial mucins", see pages 268-269
Chemical Abstracts, volume 107, no. 23, December 1987, (Columbus, Ohio, US), J. Burchell et al.: "Development and characterization of breast cancer reactive monoclonal antibodies directed to the core protein of the human milk mucin"
Biological Abstracts/RRM, volume 32, 1986, J. Taylor-Papadimitriou et al.: "Patterns of expression of keratins and mucins by mammary epithelial cells"
Biological Abstracts/RRM, volume 32, 1986, S.J. Gendler et al.: "Molecular cloning and characterization of complementary DNA species coding for differentiation and tumor-associated mucin glycoproteins expressed by humanbreast epithelium"
Download PDF:
Claims:
CLAIMS
1. An antibody or fragment thereof against a human mucin core protein which antibody or fragment has reduced or substantially no reaction with fully expressed human mucin glycoprotein.
2. Human polymorphic epithelial mucin core protein.
3. A polypeptide comprising 5 or more amino acid residues in a sequence corresponding to the sequence (I) Val Thr Ser Ala Pro Asp Thr Arg Pro Ala Pro Gly Ser Thr Ala Pro Pro Ala His Gly Val Thr Ser Ala Pro Asp Thr Arg Pro Ala Pro Gly Ser Thr Ala Pro Pro Ala His Gly (I) .
4. A polypeptide according to claim 3 having 20 or more amino acid residues in a sequence corresponding to the sequence (I) Val Thr Ser Ala Pro Asp Thr Arg Pro Ala Pro Gly Ser Thr Ala Pro Pro Ala His Gly Val Thr Ser Ala Pro Asp Thr Arg Pro Ala Pro Gly Ser Thr Ala Pro Pro Ala His Gly (I) .
5. A polypeptide according to claim 3 or claim 4 wherein at least one amino acid residue bears a linkage sugar substituent.
6. A polypeptide according to claim 5 wherein the linkage sugar bears an oligosaccharide moiety. SUBSTITUTE SHEET .
7. A polypeptide according to claim 5 or claim 6 wherein amino acid bearing a substituent is a serine or threonine and the linkage sugar is Nacetyl galactosamine.
8. A polypeptide according to any one of claims 5 to 7 linked to a carrier protein.
9. An antibody or fragment thereof against a polypeptide according to any one of claims 3 to 8 which antibody or fragment has reduced or substantially no reaction with fully processed human mucin glycoprotein.
10. An antibody or fragment thereof according to claim 1 or claim 9 against a human polymorphic epithelial mucin core protein.
11. An antibody or fragment thereof according to claim 10 against human polymorphic epithelial mucin core protein as expressed by a human colon, lung, ovary or breast carcinoma.
12. An antibody or fragment thereof according to any one of claims 1 and 9 to 11 which has no significant reaction with mucin glycoprotein expressed by pregnant or lactating human mammary epithelial tissue.
13. A monoclonal antibody or fragment thereof according to any one of claims 1 and 9 to 12.
14. A hybridoma cell capable of secreting a monoclonal antibody according to claim 13.
15. A hybridoma cell of the cell line designated HSM3 (ECACC 87010701) .
16. A monoclonal antibody secreted by HSM3 (ECACC 87010701) .
17. A nucleic acid fragment comprising at least 17 nucleotide bases the fragment being hybridisable with at least one of a) the DNA sequence 5' * ACC GTG GGC TGG GGG GGC GGT GGA GCC CGG GGC CGG CCT GGT GTC CGG GGC CGA GGT GAC ACC GTG GGC TGG GGG GGC GGT GGA GCC CGG 3' GGC CGG CCT GGT GTC CGG GGC CGA GGT GAC b) DNA of sequence 5' GTC ACC TCG GCC CCG GAC ACC AGG CCG GCC CCG GGC TCC ACC GCC CCC CCA GCC CAC GGT GTC ACC TCG GCC CCG GAC ACC AGG CCG GCC * 3« CCG GGC TCC ACC GCC CCC CCA GCC CAC GGT c) RNA having a sequence corresponding to the DNA sequence of a) and d) RNA having a sequence corresponding to the DNA sequence of b) .
18. A nucleic acid fragment according to claim 17 comprising a portion of at least 30 nucleotide bases capable of hybridising with at least one of sequences (a) to (d) .
19. A DNA fragment according to claim 17 or 18.
20. A double stranded DNA fragment comprising antiparallel paired portions having respectively sequences (a) and (b) as defined in claim 17.
21. An antibody or fragment thereof according to any one of claims 1, 9 to 13 and 16 bearing a detectable label or a therapeutically or diagnostically effective moiety.
22. An antibody or fragment thereof according to any one of claims 1, 9 to 13, 16 and 21 for use in a method of therapy or diagnosis practised on the human or animal body.
23. Human polymorphic epithelial mucin core protein bearing a detectable label or a therapeutically or diagnostically effective moiety.
24. Human polymorphic epithelial mucin core protein according to claim 2 or claim 23 for use in a method of therapy or diagnosis practised on the human or animal body.
25. A polypeptide according to any one of claims 3 to 8 bearing a detectable label or a therapeutically or diagnostically effective moiety.
26. A polypeptide according to any one of claims 3 to 8 and 25 for use in a method of therapy or diagnosis practised on the human or animal body.
27. A nucleic acid fragment according to any one of claims 17 to 20 bearing a detectable label or a therapeutically or diagnostically effective moiety.
28. A nucleic acid fragment according to any one of claims 17 to 20 and 27 for use in a method of therapy or diagnosis practised on the human or animal body.
29. An assay method comprising contacting a sample suspected to contain abnormal human mucin glycoproteins with an antibody or fragment thereof according to any one of claims 1, 9 to 13, 16 and 21.
30. A diagnostic or therapeutic method practised on the human or animal body comprising administering an antibody or fragment thereof according to any one of claims 1, 9 to 13, 16, 21 and 22.
31. A diagnostic or therapeutic method practised on the human or animal body comprising administering human polymorphic epithelial mucin core protein according to any one of claims 2, 23 or 24.
32. A diagnostic or therapeutic method practised on the human or animal body comprising administering a polypeptide according to any one of claims 3 to 8, 25 and 26.
33. A diagnostic or therapeutic method practised on the human or animal body comprising administering a nucleic acid fragment according to any one of claims 17 to 20, 27 and 28.
Description:
PROBE

The present invention relates to DNA probes for detecting a tandemly-repeated nucleotide sequence in the gene encoding mucin glycoprotein expressed by human mammary epithelial cells, to the use of the probe in diagnosis and in "fingerprinting" individuals, to the polypeptides expressed by the corresponding mucin gene, to antibodies against the polypeptides and to the use of the polypeptides and antibodies in the diagnosis and therapeutic treatment of cancer.

Normal and malignant human mammary epithelial cells express high molecular weight glycoproteins (gps) which are extensively glycosylated and very antigenic. As a result, many of the monoclonal antibodies (MAbs) selected for reactivity with human breast cancer and other carcinomas are found to react with molecules which are produced in abundance by the fully differentiated human mammary tissue and are found in the milk fat globule (MFG) and in milk. However, the level of expression of a particular antigenic determinant may be different in the gps produced by the normal differentiated cell and in the similar molecules produced by breast cancers. This means that some antibodies can show a certain specificity for reacting with tumour gps.

The molecules bearing the epitopes recognised by these antibodies are complex and have been difficult to analyse τ both because they are large and heavily glycosylated (>250,000 daltons) and because of the complex pattern of expression. Two of the MAbs, HMFG-1 and -2, react with a component in human milk which appears to be greater than 400,000 daltons, whereas the molecules found in sera and tumours are smaller, although the dominant components are still greater than 200,000 daltons on im unoblots. The large glycoprotein produced by the differentiated mammary epithelial cells found in human milk or in the milk fat globule has been purified and shown to have some of the characteristics of the mucins. This component contains a large amount of carbohydrate joined in O-linkage to serine and threonine residues via the linkage sugar N-acetylgalactosamine. Moreover, the core protein contains high levels of serine, threonine and proline and low levels of aromatic and sulphur containing amino acids.

These mucin-like glycoproteins are also secreted by a number of other normal epithelial cells. The monoclonal antibody HMFG-1 is highly reactive with the milk mucin and evidence suggests that the epitope recognised by this antibody is more abundant on the fully processed mucin, characteristic of normal differentiation.

In tumours, the molecular weight of the molecules carrying these antigenic determinants differs among individual tumours and, in the case of the components

recognised by the HMFG-2 antibody, can range from 80-400K daltons. Although it appears that the differences observed . in the mobility of the high molecular weight bands are due to genetic polymorphism this probably does not explain variations in the size of the lower bands. It has been proposed that these may be the result of aberrant processing occurring in the tumour cell possibly within the glycosylation pathways.

For the majority of the monoclonal antibodies reacting with this group of molecules the exact nature of the antigenic epitopes remains unclear but circumstantial evidence has suggested that carbohydrate may at least be partly involved in many of the epitopes. Moreover, from previously available data it was not known whether the mucin found in the normal differentiated cells, and that observed in the tumours, contain the same core protein, or just carry common carbohydrate determinants.

Mucin has now been isolated from human milk by affinity chromatography enabling identification of the core protein and the gene encoding the protein. This has been found to be a highly polymorphic gene defined by the peanut urinary mucin (PUM) locus [see Swallow e_t al♦ , Disease Markers, 4_ j _ 247, (1986) and Nature, 327, 82-84 (1987)] . The gene product, which is hereafter referred to as human polymorphic epithelial mucin or HPEM, has been detected in breast tumours and other carcinomas as well as in some normal epithelial tissues.

It has now been found that the HPEM core protein has epitopes which also appear in the aberrantly processed gps produced by adenocarcinoma cells. Certain of these epitopes are not exposed in the fully processed mucin glycoprotein produced by the lactating mammary gland.

In one aspect the present invention therefore provides an antibody against a human mucin core protein which antibody substantially does not react with a fully processed human mucin glycoprotein.

As used herein the term "antibody" is intended to - include fragments of antibodies bearing antigen binding sites such as the F(ab')- fragments.

Antibodies according to the present invention react with HPEM core protein, especially as expressed by colon, lung, ovary and particularly breast carcinomas, but have reduced or no reaction with the corresponding fully processed HPEM. In a particular aspect the antibodies react with HPEM core protein but not with fully processed HPEM glycoprotein as produced by the normal lactating human mammary gland.

Antibodies according to the present invention have no significant reaction with the mucin glycoproteins produced by pregnant or lactating mammary epithelial tissues but react with the mucin proteins expressed by mammary epithelial adenocarcinoma cells. These antibodies show a much reduced reaction with benign breast tumours and are therefore useful in the diagnosis and localisation of

breast cancer as well as in therapeutic methods.

The antibodies may be used for other purposes including screening cell cultures for the polypeptide expression product of the human mammary epithelial mucin gene, or fragments thereof, particularly the nascent expression product. In this case the antibodies may conveniently be polyclonal or monoclonal antibodies.

Antibodies according to the present invention may be produced by innoculation of suitable animals with HPEM core protein or a fragment thereof such as the peptides described below. Monoclonal antibodies are produced by the method of Kohler & Milstein (Nature 256, 495-497/1975) by immortalising spleen cells from an animal innoculated with the mucin core protein or a fragment thereof, usually by fusion with an immortal cell line (preferably a myeloma cell line) , of the same or a different species as the innoculated animal, followed by the appropriate cloning and screening steps.

In a particular aspect the present invention provides the monoclonal antibodies designated SM3 against the HPEM core -protein. In another aspect the invention provides the hybridoma cell line which secretes the antibodies SM3 and has been designated HSM3. Samples of HSM3 have been deposited with ECACC on 7th January 1987 under accession number 87010701. using antibodies according to the invention it has been possible to screen a phage library constructed

from mRNA isolated from a human breast cancer cell line to identify sequences coding for portions of the mucin core protein. Complementary DNA sequences have been constructed and from these it has surprisingly been found that the gene encoding the core protein contains multiple tandem repeats of a 60 base sequence leading to considerable polymorphism sufficiently extensive that cDNA fragments corresponding to the repeat sequence would be useful for fingerprinting DNA. The fingerprinting thus made possible has applications in for instance ascertaining whether bone marrow growth after transplants is from the host or the donor and in forensic medicine for identifying individuals using body tissues or fluids.

Accordingly the present invention also provides a nucleic acid fragment comprising at least 17 nucleotide bases the fragment being hybridisable with at least one of a) the DNA sequence

5 « *

ACC GTG GGC TGG GGG GGC GGT GGA GCC CGG-

GGC CGG CCT GGT GTC CGG GGC CGA GGT GAC-

*

ACC GTG GGC TGG GGG GGC GGT GGA GCC CGG-

3' GGC CGG CCT GGT GTC CGG GGC CGA GGT GAC b) DNA complementary to the DNA of a) , i.e. of sequence

5'

GTC ACC TCG GCC CCG GAC ACC AGG CCG GCC- *

CCG GGC TCC ACC GCC CCC CCA GCC CAC GGT-

GTC ACC TCG GCC CCG GAC ACC AGG CCG GCC- * 3"

CCG GGC TCC ACC GCC CCC CCA GCC CAC GGT

c) RNA having a sequence corresponding to the DNA sequence of a) and d) RNA having a sequence corresponding to the complementary DNA sequence of b) .

The sequences in (a) and (b) each include a doubl tandem repeat sequence of 120 bases. Fragments according t the invention may correspond to any portion of this sequenc including portions bridging the start point of the repeat.

Fragments according to the invention will hybridise under conditions of low stringency with the DNA and RNA sequences (a) to (d) above. Preferred fragments are those which also hybridise under conditions of high stringency. The most preferred fragments of the invention are those which have sequences exactly identical to, or exactly complementary to the sequences (a) to (d) above.

Normally a given DNA or RNA fragment according to the invention will be capable of hybridising with both DNA according to a) and RNA according to c) or with both DNA according to b) and RNA according to d) above.

Preferably the nucleic acid fragment according to the present invention will comprise a portion of at least 30 nucleotide bases capable of hybridising with at least one of a) to d) above, more preferably at least 50 such bases and most preferably the fragment contains a sequence of 60 bases exactly complementary to one of the repeat sequences of a) , b) c) or d) above. Other fragments of the invention may comprise two or more repeats of such a sequence, optionally

SUBSTITUTE SHEET

with minor variations by way of substitution. Preferably such fragments include an integral number of such repeat sequences. Further fragments of the invention comprise the tandem repeat sequence and additional coding or non-coding 5' and/or 3' flanking sequences corresponding to the HPEM gene or a portion thereof.

When the existence of a tandem repeat sequence was first identified it was believed that the sequence consisted of 59 base pairs corresponding with the sequences indicated in (a) and (b) above except for the lack of the base indicated with "*".

Many fragments according to the invention as originally defined in British Patent Application No. 8700269 also conform with the new definition of fragments as set out herein and those fragments of sequences defined under (a) , (b) , (c) or (d) above which do not include the bases marked "*" form a particular aspect of the present invention. Such fragments have sequences corresponding to at least a portion of the sequences a* ) GTG GGC TGG GGG GGC GGT GGA GCC a") CGG GGC CGG CCT GGT GTC CGG GGC CGA GGT GAC AC b r ) DNA complementary to the sequence of a 1 ) or a 11 ) / c 1 ) RNA having a sequence corresponding to the sequence of a' ) or a' ' ) and d 1 ) RNA having a sequence corresponding to one of the complementary DNA sequences of b')

In the human genome the DNA tandem repeat sequence

comprises antiparallel double stranded DNA, one strand having sequence (a) and being paired with a strand having sequence (b) .

As mentioned above the nucleic acid fragments of the invention may be used as a probe for detecting one or other strand of the DNA tandem repeat sequence in the human genome, or RNA transcribed from either strand and hence for identifying the gene or genes for human mucin core proteins, mRNA transcribed therefrom and complementary DNA and RNA. For such purposes it may be convenient to use the complete normal gene comprising at least one tandem repeat sequence, or mRNA transcribed therefrom or to attach non-complementary fragments to either or both the 5' and 3' ends of a fragment according to the invention and/or to attach detectable labels (such as radioisotopes , fluorescent or enzyme labels) to the probe or to bind the probe to a solid support. All of these may be achieved by conventional methods and the nucleic acid fragments of the invention may be produced de novo by conventional nucleic acid synthesis techniques.

The nucleic acid fragments of the present invention may also be used in active immunisation techniques. In such methods the fragment codes for a polypeptide chain substantially identical to a portion of the mucin core protein and may be extended at either or both the 5' and 3 1 ends with further coding or non-coding nucleic acid sequences including regulatory and promoter sequences, marker sequences and splicing or ligating sites. Coding

sequences may code for corresponding portions of the mucin core protein chain or for other polypeptide chains. The fragment according to the invention, together with any necessary or desirable flanking sequences is inserted, in an appropriate open reading frame register, into a suitable vector such as a plasmid or a viral genome (for instance vaccinia virus genome) and is then expressed as a polypeptide product by conventional techniques. In one aspect the polypeptide product may be produced by culturing appropriate cells transformed with a vector, harvested and used as an immunogen to induce active immunity against the mucin core protein. In another aspect the vector, particularly in the form of a virus, may be directly innoculated into a human or animal to be immunised. The vector then directs expression of the polypeptide _ir_ vivo and this in turn serves as an immunogen to induce active immunity against the mucin core protein.

The invention therefore provides nucleic acid fragments as hereinbefore defined for use in methods of treatment of the human or animal body by surgery or therapy and in diagnostic methods practised on the human or animal body. The invention also provides such methods for treatment of the human or animal body by surgery or therapy and diagnostic methods practised _in vivo as well as e_x vivo and _in vitro.

The invention further provides a polypeptide comprising a series of residues encoded by the DNA tandem

repeat sequence, the sequence shown at (b) above being the coding sequence. Polypeptides according to the invention are selected from any of those having 5 or more amino acid residues represented by the following amino acid sequence

Val Thr Ser Ala Pro Asp Thr Arg Pro Ala Pro Gly Ser Thr Ala Pro Pro Ala His Gly*Val Thr Ser Ala Pro Asp Thr Arg Pro Ala Pro Gly Ser Thr Ala Pro Pro Ala His Gly ("*" marks the start of the repeat in the peptide) .

Polypeptides according to the invention may have a sequence cor esponding with any portion of the 40 residue sequence above and may include the start point of the repeat sequence.

Other polypeptides according to the invention include three or more repeats of the 20 amino acid repeat sequence. Such polypeptides may include minor variations by way of substitution of individual amino acid residues. The invention further provides polypeptides as defined above modified by addition of N-acetyl galactosamine (a linkage sugar) on serine and/or threonine residues and by addition of oligosaccharide moieties to that or via other linkage sugars and/or fragments linked to carrier proteins such as keyhole limpet haemocyanin, albumen or thyroglobulin.

Preferably the polypeptide comprises at least 10 amino acid residues of the sequence above, more preferably 20 residues. The polypeptide may comprise the full

sequence above. Such polypeptides may further comprise additional amino acid residues, preferably conforming to the amino acid sequence of HPEM core protein.

In a further aspect the present invention provides the HPEM core protein. This is encoded by the PUM gene and may be produced by recombinant DNA techniques and expressed without glycosylation in human or non-human cells. Alternatively it may be obtained by stripping carbohydrate from native human mucin glycoprotein which itself may be produced by isolation from samples of human tissue or body fluids or by expression and full processing in a human cell line. The HPEM core protein may be used for raising antibodies in animals for use in passive immunisation, diagnostic tests and tumour localisation and in active immunisation of humans.

The invention further provides antibodies (monoclonal or polyclonal) , and fragments thereof, against any of the polypeptides described above. Such antibodies may be obtained by conventional methods and are useful in diagnostic and therapeutic applications.

The invention further provides antibodies (monoclonal or polyclonal), or fragments thereof, linked to therapeutically or diagnostically effective ligands. For therapeutic use of the antibodies the ligands are lethal agents to be delivered to cancerous breast or other tissue in order to incapacitate or kill transformed cells. Lethal agents include toxins, radioisotopes and 'direct

killing agents' such as components of complement as well as cytotoxic or other drugs. Further therapeutic uses of the antibodies inclusive passive immunisation.

The invention further provides therapeutic methods comprising the administration of effective non-toxic amounts of such antibodies or fragments thereof and antibodies or fragments thereof for use in therapeutic treatment of the human or animal body. Especially in therapeutic applications it may be appropriate to modify the antibody by coupling the Fab region thereof to the Fc region of antibodies derived from the species to be treated (e.g. such that the Fab region of mouse monoclonal antibodies may be administered with a human Fc region to avoid immune response by a human patient) or in order to vary the isotype of the antibody.

In the diagnostic field the antibodies may be linked to ligands such as solid supports and detectable labels such as enzyme labels, chromophores and fluorophores as well as radioisotopes and other directly or indirectly detectable labels. Preferably monoclonal antibodies or fragments thereof are used in diagnosis.

The invention further provides a diagnostic assay method comprising contacting a sample suspected to contain abnormal human mucin glycoproteins with an antibody as defined above. Such methods include tumour localisation involving administration to the patient of the antibody or fragment thereof bearing a detectable

label or of an antibody or fragment thereof and, separately simultaneously or sequentially in either order a labelling entity capable of selectively binding the antibody or fragment thereof. The invention also provides antibodies or fragments thereof for use in diagnostic methods practised on the human or animal body.

Particular uses of the antibodies include diagnostic assays for detecting and/or assessing the severity of breast, ovary and lung cancers.

Diagnostic test kits are provided for use in diagnostic assays and comprise antibody or a fragment thereof, optionally suitable labels and other reagents and, especially for use in competitive assays, standard sera.

The invention will now be illustrated by the following Examples and with reference to the figures of the accompanying drawings in which

Figure Leg end s

Figure 1 ; Purification of the milk mucin by i_ uno ffinity chromatography usi g the antibody H-TFG-I . Milks from aeτeral indiτiduals were combined and absorbed to a B-FG-I -Sepharoie column as described in Methods . The materiel alnting at low pH was iodinated and subjected to PAGE •lectrophorββis and aπtoradiography (track 1 ) . The iodinated material was precipitated using the Protein A asethod with antibodies HVΣFG-1 (track 5) , 10_FG-2 (track 2) , ET254 (track 3) and RPMI 4 20% PCS (track 4) .

125

Figure 2: Comparison of the I labelled purified milk mucin with i___unoblot of human skimmed milk. A, human skimmed milk was subjected to SDS polyacrylemide electropboresis, transferred to nitrocellulose paper, the blot probed with the monoclonal antibody H_ G-1 and binding detected using an ΣLISA method. B, after purification on an H-PG-1 affinity column followed by G75 Sephadez chromatography the milk mucin was iodinated by the Balton and Hunter method and subjected to SDS polyacrylaaide electrophoresis and autoradiography.

Figure 5; Autoradiography of the iodinated milk mucin after treatment trith hydrogen fluoride. The purified milk mucin was treated with HP for 5 hours at room temperature (track 1) or 1 hour at 4*C (track 2) and the resulting preparations were then iodinated and run on EDS polyacrylaaide gels.

SUBSTITUTE SHEET

- 1

Figure 4: Reactivity of the intact, partially stripped or extensively stripped milk mucin with iodinated lectinβ. The purified intact milk mucin (track 1), the mucin treated with HF for 1 hour at 4 * C (track 2) and the mucin treated for S hours at room temperature (track 5) were aubjectβd to SDS polyaerylamide electrophoresis and then transferred to nitrocellulose paper. The paper was then probed with 125I PNA

(peanut agglutinin), 125 I WGA (wheat germ agglutinin), or 125 I HPA

(Helix pomatia agglutinin).

Figure 5: Imπuncprecipitεtion and of the partially t_nd βxtenεively stripped mucin. A, the 125I extensively stripped mucin was i__sunoprβeipitεtβd with EM-3 (track 3), E__?G-2 (track 2) or λ'S2 mediu-ϋ as a control (track 1) by the protein A plate method (see

Materials and Methods). B, the partially stripped mucin (track ) or extensively stripped mucin (track 2) was run on SDS polyaerylamide gels and trsnsferred to nitrocellulose paper. The blot was then reacted with a cocktail of SM-3 and SM-4 monoclonal antibodies and the binding detected using an ΣLISA method.

Figure 6: Reactivity of monoclonal antibodies SU-3 and H_u*G-2 with βethacara fixed breast tisβue and tumour sections using an indirect imαunoperoxidase staining method. Infiltrating ductal carcinoma showing atrong reactivity with both SM-3 (A) and HMFG-2 (B) . Fibroadenoma ahowing no reactivity with SM-3 (C) and atrong heterogeneous staining of the epithelium with HVEFG-2 (D). Papilloma ahowing very weak reactivity with SM-3 (E) and strong positivity with HJ£FG-2 (F). Both normal resting breast (G) and lactating breast (I)

SUBSTITUTE 9

were negative when stained with SM-3, wherβaβ both tissues stained positively with HM7G-2 with lactating brβaβt (J) much stronger than normal resting breast (H) .

SUBSTITUTE SHEET

Figure Legends

Figure 7. Periodic acid-silver stained milk mucin after antibody affinity column and gel filtration column. Milk mucin was purified on an HMFG-1 antibody affinity column (lane 1) followed by passage through a G75 Sephade∑ column (lane 2), subjected to KaDodSO./polyacrylamide gel electrophoresis, and silver stained following treatment of gels with 0.2% periodic acid.

Figure p, . Silver stain of partially and totally stripped core protein from milk mucin. The purified milk mucin was deglycoBylated by treatment with anhydrous hydrogen fluoride for 1 hr at O'C (lane 1) and 3 hr at room temperature (lane 2) , separated by electrophoresis through a NaDodSO /polyacrylamide gel (10%) and silver stained.

Figure q. Immunoprecipitation with MAbs of in vitro translated protein products from HCF-7 poly(A) + RNA. Poly(A) + RNA from MCF-7 cells was translated in a rabbit reticulocyte lysate system (Amersham) in the presence of [ 35 S]methioninβ (1000 Ci/mmole; 1Ci = 37 GBq) following the manu acturer's conditions. Samples containing 5 x 10 acid precipitable cpm were precipitated with MAbs SM- (lane a) , SM-3 (lane b), HMFG-2 (lane c), HMFG-1 (lane d) and an irrelevant MAb to interferon (lane e, 24), separated on a KaDodSO./polyacrylamide gel (10%), impregnated with Amplify and exposed to ΣAR-5 film at -70"C for 20 days.

j UBSTlTUTE SHEET

Figure 10. Immunoblot analysis of fusion proteins from the λmuc clones. The phage clones λMUC 3,4,6,7,8,9 and 10 were used to lysogenize bacterial strain I 1089. Lysogens were grown at 32"C, βhifted to 42 * C, and then induced with IPTG. Lysogen proteins were fractionated by electrophoresis through a KaDodSO /polyacrylamide gel

(7.5%), transferred to nitrocellulose, and reached with HM7G-2. The binding was detected with an ELISA method using 4-chloro-1-naphthol as the substrate. The numbers are those of the λ clones.

Figure11. .Hybridization of pMJCIO to cDNA insertB of pMJC clones. DNA from the plasmid clones waε digested with restriction enzyme EcoRI to excise the cDNA inserts, separated by electrophoresis on 1.4% agarose and transferred to Biodyne nylon membrane. The filter was hybridized using standard conditions (34) to the insert from pMTJCIO which was labelled with [α- p]dCTP by the method of random priming (41). Lanes: plasmid clones 3,4,6,7,8,9,10.

Figure 12. RNA blot hybridization analysis of mammary breaβt mucin mRNA. 10 μg of total RNA from human breast cancer cells MCF-7 (lane 1) and T47D (lane 2), normal human mammary epithelial cells Hu E (lane 3), human embryonic fibroblaβts ICRF 23 (lane 4), Daudi cells (lane 5) and careinosarcoma HS578T cells (lane 6) were separated in a 1.3% agarose/glyoxal gel, blotted on to nitrocellulose and hybridised to the pMTJCIO EcoRI insert which was labelled with [o- p]dCTP by the method of random priming (41). Ihe size markers were 28S (5.4 kb) and 18S (2.1kb) rRNAs.

. 2 .

Figure 13. Polymorphic human DNA fragments detected by hybridization with pMTJCIO probe. Genomic DNA samples prepared from the white blood cells from ten individuals (six unrelated) and from three cell Xines were digested to completion with HinfI and EcoRI, fractionated by electrphoresis through 0.7% and 0.6% agarose, respectively, and transferred to Biodyne nylon membranes. The filter was hybridized to the pMTJCIO DNA insert which was labelled with

32 [α pjdCTP by the method of random priming (41). X-ray film was exposed for 1 day at -70 * C with intensifying screens. Lanes 1-4 father, two daughters and mother, lanes 5-10 unrelated individuals, lane 11 is MCF-7, lane 12 is ZR75-1, lane 13 is ICRF-23. The DNA samples exhibit a wide distribution of sizes. Numbers indicate length of DNA in kb. The apparent bands at 23∑b are in lanes 12 and 13 are artefacts introduced in autoradiography

SUBS ι ' T1 τuτε saεeπ

Example 1

Purification of the milk mucin

The milk mucin was purified from human skimmed milk by passage through an HMFG-1 affinity column followed by aizβ exclusion chromatography. The HMFG-1 monoclonal antibody waβ purified from tiβBue culture supernatant using a protein A column (1). The purified antibody was coupled to cyanogen bromide activated eepharcee (Pharmacia) as described in the manufacturer's instructions. Human skimmed milk waε paBsed in batches of 100 ml throu£h the antibody co.1u.-_n followed by extensive washing with PBS. Bound tntigen was eluted from the column using 0.1 M glycine pH 2.5 and the fractions registering an optical density at 260nm were pooled, diεlyεgά against 0.25 M acetic acid and lyophilized. Batches of about 20 m B were dissolved in 0.25 M acetic acid and passed through a G75 Sephade∑ column (1 x 100 cm) which had been previously equiligrated with acetic acid. The column was washed with 0.25 M acetic acid and 1ml fractions collected. The peak fractions which were eluted in the void volume were pooled, lyophilized and the dry powder stored at 4"C. Amino acid analysis was performed using a Beekm n 6300 amino acid analyser.

Deglycosylation of the milk mucin

To remove the 0-1inked carbohydrate from the milk mucin the molecule was treated with anhydrous hydrogen fluoride aa described by Mort and Lamport (21), for either 1 hour at 4 * C which produced a partially stripped preparation, or 3 hours at room temperature which produced the extensively stripped mucin.

SUBSTITUTE SHEET

_ 22 _

lodination of the milk mucin

Iodinations of the purified mucin, the partially or extensively ■tripped mucin were carried out using the Bolton and Hunter method (51). Briefly, the aucin, 2.5 μg in 20 μl 0.1M borate buffer pH 8.5, was added to the dried Bolton and Hunter reagent (1 »Ci, .Amersham International pic) and incubated at room temperature for 15 minutes. Zhe reaction was stopped by the addition of 0.5 ml of 0.2M glycine in borate buffer and after a further 15 minutes incubation, free Bolton and Hunter reagent was removed by passage through a G25 Sephadex column (PD10 columnβ, Pharmacia) previously equilibriated in PBS.

lodination of ectins

Wheat germ agglutinin (WGA), peanut agglutinin (PKA) (Vector labs) and Helix pomatia agglutinin (HPA) (Boehringer) were iodinated as described by ∑arlsson et al. (52) using the chloramine T method.

Polyacrylamide eels and Western blots

Polyacrylamide gel electrophoresis and immunoblotting was performed as described previously (1). Briefly, samples were run on 5-15% polyacrylamide gels and then electrophoretically transferred to nitrocellulose paper (Schleichβr and Schuell) at 50 volts overnight at 4 * C (36). In the immunoblotting •xperisents the paper was reacted with monoclonal antibodies and binding detected with, an ZLISA method using 4-chloro-1-naphthol aa the substrate. For Xectin binding studies the Western blots were reacted with the iodinated lectins as described by Swallow et al. &8).

SUBSTITUTE SHEET

Production of monoclonal antibodies

A female EALB/c mouse was immunized with 5 μg of the partially stripped milk mucin in Frβund'β complete adjuvant and S months later boosted with a further 5 μg of the same preparation in Frβund's incomplete adjuvant. After a further 20 days, 5 μg of the mucin extensively stripped of its carbohydrate was given intravenously in saline solution. The spleen was removed 4 days later, and fused with the NΞ2 mouse myeloma cell line (53) .

Screening of hybridoma supernatant and immunoprecipitations

The screening assay was a modification-of that described by Melero and Gonzalez-Rodriguez (54">. Multiwell plates were coated with 50 μl of 0.1 mg/ml protein A (Pharmacia Pine Chemicals) in PBS and allowed to dry overnight at 37 * C. The plates were blocked with 5% BSA for 1 hour at 57"C followed by the addition of 50 μl of rabbit anti-mouse immunoglobulin (DAEO, diluted 1:10 in PBS BSA = PBS/BSA). After incubating for 2 hours at S7 * C the plates were washed twice with PBS containing 1% BSA and 50 μl of hybridoma supernatant added. The plates were incubated overnight at 4 * C, washed twice with PBS/BSA and 50 μl of iodinated partially stripped mucin containing 100,000 cpm added to each well. Zhe plates were then incubated at room temperature for 4 hourβ, washed 4 times with PBS/BSA »n*l the individual wells counted in a gamma counter. For immunoprecipitation experiments 50 μl of SDS sample buffer containing dithiothrβitol was added to each of the wells which were then boiled for S minutes and the buffer loaded onto 5-15% polyacrylamide gradient gels.

Staining of tissue sections

Tissues from primary mammary carcinomas, benign breast biopsies, normal breast, and pregnant lactating breast tissue were fixed in ■ethacam (sethanol chloroform and acetic acid 60:30:10) and embedded into paraffin wax. Sections were stained with the antibodies using the indirect peroxidase anti peroxidase method as previously described (47).

Results

Purification of the milk mucin

The milk mucin was purified from human skimmed milk on an HMFG-1 antibody affinity column. lodination of the eluted material revealed the presence of a large molecular weight component and a 68ΣD band. Precipitation of the affinity purified material with antibodieβ HMFG-1 and HMFG-2 (tracks 2 and 5) followed by gel electrophoresis showed that both the high molecular weight components and the 68ED component were precipitated by both antibodies (less effectively by HMFG-2). Since the 68ED component was also precipitated by two unrelated antibodies (figure 1, tracks 3 and 4) and this component was not evident on an iamunoblot of the purified material reacted with HMFG-1 ( igure 2A), the 68K component was removed by molecular sieve chromatography on a G75 column. Zhe final purified product showed a Bajor high molecular weight band, with only a traceof the 68£ component and a minor contaminant around 4Σ (figure 2B).

SUBSTITUTE SHEET

A high molecular weight glycoprotein (PAS-0) containing more than 50% carbohydrate in O-linkage has been purified from the human milk fat globule by Shi izu and lamauchi (8). To see whether this component was similar to the mucin isolated from silk by affinity chromatography on an HMFG-1 affinity column, the amino acid composition of the purified HMFG-1 reactive mucin was determined and compared to the amino acid composition of the purified PAS-0 component. Table 1 shows that there is good correspondence between the two sets of data, indicating that the core proteinβ of PAS-0 and the mucin purified here are the same.

Isolation of the core protein of the milk mucin

As there are no enzymes easily available that are efficient at removing 0-1inked sugars, and β elimination often results in damage to the protein core, the oligosaccharides were removed by treatment of the mucin with anhydrous hydrogen fluoride. This treatment has been shown by Mort and Lamport (21) to be effective in removing sugars from pig submaxillary mucin without damaging the protein core. Amino acid analysis of the material produced after HF treatment of the milk mucin suggested that the protein core was also in this case undamaged, since the composition was the same as that seen in the intact mucin (Table

1).

Initially the silk mucin was exposed to HF for only 1 hour at 4*, but analysis of the product showed that there was only partial removal of the sugars with such treatment, and it was necessary to treat the sucin at room temperature for 3 hours to obtain a molecule which

SUBSTITUTE SHEET

ahowed no lectin binding ability. Pigure 3 shows an autoradiograph of the iodinated productβ after treatment for 1 hour at 4 * (track 2) or 3 hours at RT (track 1). It can be seen from Pigure 3 that the milder treatment results in a mixture of products made up of high molecular weight material which is slightly smaller than the intact mucin and a number of smaller bands. After longer exposure to HF at room temperature, the high molecular weight bands disappeared resulting in polypeptide bandβ of about 68ΣD and 72ED.

To test for the presence of sugars on the intact =:ucin and on the products produced after the two different KF treatments each preparation was subjected to acryla.-.ide gel eleεtrophsreEiε,

12 ς transferred to nitrocellulose pεper and reacted ati b "I-labelled lectins. The lβctinε used were peanut lectin (PNA) hich reacts with galactose linked to -acetyl galactose-nine , wheat germ (WGA) reactive with H-acetyl glucosamine and Helix pomatia agglutinin (HPA) which reacts with the linkage sugar in 0-1inked glycoεylεtion, N-acetylgalactoβamine. Figure 4 shows autoradiographs of the reacted blots, and it can be seen that while treatment τilh E? for 1 hr at 4 * (track 2) alters the lectin reactivity of the mucin, carbohydrate is still present. Interestingly, however, there is a much lower level of binding of PNA to the high molecular weight material of the partially stripped mucin than is seen with the intact sucin (track 1). Moreover, this loss in FKA binding ability is accompanied by binding of the linkage sugar specific lectin HPA. This lectin shows so binding at all to the intact mucin, and the changed pattern of lectin binding after limited treatment with HF indicates that sugars masking the 0-linked N-acetylgslactosamine have been stripped off. The

SUBSTITUTE SHEET

smaller component seen in both the intact mucin (track 1) and in the partially stripped preparation (track 2) is a glycoprotein which reacts with WGA, although not with PNA. This may correspond to the component of similar molecular weight (around 68Σ) seen after affinity chromatography of the mucin and may represent an intermediate precursor molecule.

Figure 4 showβ clearly that the 6BE and 72Σ components produced after extensive treatment with KF (S fcr at RT) , show no reactivity with the lectins (track 3), including the K-acetylgalactosamine specific lectin HPA. Th ε observation constitutes strong evidence that the sugars have been recoveαfroin at least the majority of the molecules, and we will refer to this preparation as the extensively stripped mucin.

Generation of monoclonal antibodies to the milk mucin core protein

A fusion was carried out using the spleen of a mouse that had been immunized with two injections of the partially stripped milk mucin follo»βd by a boost with the extensively εtripped mucin. The clones were initially screened against the 125I partially stripped material using protein A plates (see Methods). Four hybridomas were selected and cloned, and table 2 shows their spectrum of reactivity with the intact, partially and extensively atripped mucin. As can be aeen from this table three of the hybridomas which were isolated showed a strong reaction with the partially and extensively atripped mucin and did sot react with the intact mucin. These appeared to be good candidates for monoclonal antibodies to the protein core and two, SM-3 and SM-4, were selected to be characterised further.

SUBSTITUTE SHEET

- 2 Q ■

It can also be seen from table 2 that the HMFG-1 and HMFG-2 antibodies reacted very strongly with the mucin stripped of its carbohydrate. Theβe two antibodies were, in fact, developed using the intact mucin (from the silk fat globule) as immunogen and, in the case of HMFG-2, growing mammary epithelial cells 0.4). Their reaction with the stripped mucin was unexpected, as circumstantial evidence had previously led to the belief that carbohydrate sight form at least part of their antigenic epitopes.

Molecular Weight of molecules carrying antigenic determinants

The antibody SM-3 was shown to be of the IgG1 subclass, while the SM-4 antibody was found to be IgM. We therefore chose to use the SM-3 antibody in subsequent experiments since antibodies of the IgM class can present problems in some appliction. Immunoprecipitation of the extensively stripped material with SM-3 showed a reaction with the lectin unreactive 68£ component (Figure 5A, track 3). The monoclonal antibody HMFG-2 can also be seen to immune precipitate the lectin-unreactive 58Σ component (track * 2). The antibodies were reactive with antigen on ismunoblots and Figure 5B shows the reaction of antibody SM-3 with the dominant 68Σ band of the extensively stripped mucin (track 2).

We have previously shown that the molecular weight of the eosposentβ is breast cancer cells carrying determinants found on the silk mucin is lower than 400Σ and can vary from one tumour to another (1). Reaction ox * antibody SM-3 with Western blots of gel separated extracts of breast tumour cells shows that this antibody reacts with components of similar molecular weight to those reactive with antibody

SUBSTITUTE HEET

HMFG-2 (data not shown). Because the antibody SM-3 differs from the antibodies HMFG-1 and 2 in that it does not react with the intact suein processed by the lactating gland and yet reacts with molecules processed by breast cancer cells, it was appropriate to examine the reaction of SM-3 with a range of breast cancers.

Reactivity of SM-3 with breaBt tisBueB and tumours

The antibody SM-3 reacted with paraffin embedded tissues provided these were fixed in mβthacarn (not formal saline), using this method for preparation of tisβue sections, the reaction of the antibody was compared to that of HMFG-2 on breast tissues and tumours with an indirect immunoperoxid se staining method. Thiβ analysis showed a dramatic difference in the staining pattern of SM-3 compared to that seen with HMFG-2. Thus, although a strong positive reaction was seen in 20/22 breast cancers stained with SM-3 (as compared to 22/22 stained with HMFG-2), normal resting breast, pregnant or lactating tissues and most benign lesions were largely unstained with SM-3 but were stained with HMFG-2. Some examples of staining patterns of breast tissues and tumours are illustrted in Figure 6.

Twenty-two primary carcinomas and fourteen benign lesions were examined and the reaction of SM-3 compared to the staining with HMFG-2 is each case. In the primary carcinomas, staining with SM-3 was heterogeneous but generally quite strong asd always confined to tumour cells; connective tissue and βtroma showed so reaction (see figures 6A,B). In the four fibroa enomas examined, staining of the epithelium with HMFG-2 was strong although heterogeneous. In comparison, staining with SM-3 was negative in one case and in the three others

SUBSTITUTE SHEET

βtaining was confined to only one or two glandular elements. HMFG-2 showed strong positivity on the five papillomas and five cases of cystic disease studied while the staining observed with SM-5 was very such weaker and sore heterogeneous (figureB 6G,H). Zhe papillomas as a group showed the strongest staining with SM-3, and it can be seen that the staining was membranous or extracellular.

In contrast to HMFG-1 and HMFG-2 which strongly stain lactating and pregnant breast, SM-3 was totally negative with three out of aix caBβs of pregnant or lactating breaεt (see figure 6C and D). Two positive cases showed only very weak staining of an occasional cell and in the third, staining was confined to two areas of one lobule. Again, in contrast to HMFG-1 and HMFG-2 which do react with some terminal ductal lobular units of normal, resting breast (albeit weakly) , SM-3 was totally negative on eight out of the thirteen cases teβted and in the other five cases staining was extremely weak and often confined to one or two acini in the tissue section (see figure 6E and F). It should perhaps be noted that the intensity of staining with HMFG-2 seen with normal breast tissueβ and benign lesions fixed in methacarn was somewhat higher than that reported previously using formalin fixed material (50,4 * 3 *

SM-3 was also shown to be negative on sections of normal liver, lung, thymus, sweat gland, epididymus, prostate, bladder, small istestise, large istestise, appendix, thyroid and skin. The antibody showed weak positive.staining only with the distal tubules of the kidney, the occasional chief cell of the stomach, the occaaional duct cell of the salivary gland and the sebaceous gland.

SUBSTITUTE SHEET

Pi B CUE si on

Large molecular weight mucin molecules are expressed by many carcinomas and carry many of the tumour associated antigenic determinants recognised by monoclonal antibodies. These epitopes may also be expressed by some normal epithelium, and some monoclonal antibodies like HMFG-1 react particularly well with a mucin found in normal human milk (1,17). As long as the study of the sucinε is restricted to their detection with antibodies reactive with undefined epitopes, the knowledge of their structure, expression and procbssing will alεo be restricted. We have begun to investigate the structure and expression of the can-nary mucin by isolating the core protein and developing antibodies which have allowed us to select partial cDNA clonβB for the gene coding for the core protein . This Example describes the production nd characterization of these antibodies.

Treatment of the HL-FG-1 affinity purified milk mucin with hydrogen fluoride resulted in the appearance of a dominant band of about 68Σ daltons and a minor species of about 72ΣD on SDS acrylamide gels. These bands showed no reactivity with lectins, including Helix pomatia agglutinin which is specific for N-acetyl galactosamine, the first sugar in 0-1inked glyco'sylation (55). i therefore seems probable that this 6BΣ dalton polypeptide represents the core protein of the suein. Supportive evidence for this comes from the observation that the antibodies described here, which are reactive with the stripped 681 component, can precipitate a molecule of this size from the in vitro translation products of sRNA isolated from breast cancer cells expressing the suein .

SUBSTITUTE SHEET

As the silk mucin contains at least 50% carbohydrate (16) ,a protein core of only 68ΣD appears too small if the intact molecule has an observed solecular weight greater than 400ED. However, sucins can be composed of small subunits which aggregate and are held together by some form of non-covalent interactions, as yet sot understood. For example, although the solecular weight of the ovine submaxillary mucin has been reported to be greater than 1 x 0 daltons ( -5 it has a protein core of only 650 amino acids with a solecular weight of 56,300 daltons & ).

An unexpected finding was that the antibodies HMFG-1 and HMFG-2 which react with the milk mucin, also show a positive reaction with the extensively stripped material which showed no lectin binding capability. Previous indirect evidence, including the resistance to fixation, boiling and reduction, the repetitive nature of their epitopes and the appearance of several bands on immunoblots, had led to the belief that carbohydrate present on the silk sucin was involved in these epitopes. This idea was reinforced by the observation that lectins could block the binding of HMFG-1 and 2 (1 ). While it is sot possible to exclude the possibility that some sugars, undetected by the lectin binding experiments, remain on the extensively stripped sucin described here, this is unlikely to be the explasatios for the

SUBSTITUTE SHEET

reactivity of the antibodies HMFG-1 and 2. This can be said since both antibodies have recently been shown to react positively with β-galactosidaβe fusion proteins expressed by phage carrying DMA coding for the core protein of the mammary sucin . It appears therefore that at least part of each of the epitopes recognised by HMFG-1 and HMFG-2 contain amino acids but it must be assumed that some of these epitopes on the core protein are exposed, i.e. sot masked is the fully glycosylated molecule. The HMFG-2 epitope is however lees abundant on the silk sucin than the HMFG-1 epitope, while it is readily detectable on the mucin molecules expressed by tumours (1). These molecules have a smaller molecular weight and say be less heavily glycosylated or polymerised.

Here we have reported the development of sew antibodies which are reactive with the protein core of the mucin and with the partially deglycosylated molecule, but which are unreactive with the fully processed sucin produced by the lactating mammary gland. One of these antibodies SM-3, which is an IgG1, has been studied in sore detail. It has been shown to react with the sucin molecules which are produced by breast cancer cells and are recognised by sany antibodies developed against the intact silk sucin. It should be emphasised however that the epitope recognised by SM-5 which is on the core protein and is exposed in the sucin aa processed by tumour cells, is not exposed on the normally processed silk sucin. This feature offers the possibility of enhanced tumour specificity, and a pilot immunohistochemical study of breast tumours and tiaaues has shown that indeed the SM-3 antibody reacts strongly with the majority of primary

SUBSTITUTE SHEET

breast cancers (91%) but ahows little or no reaction with benign breaεt tumours, resting or lactating breast, and most normal tissues.

There are several implications of the work described here which say be important for both basic and clinical studies is breast cancer. The observation that parts of the core protein (detectable by antibodieβ) are exposed on the sucins as processed by breast cancer but sasked on the sucin as processed by cells in normal breast and benign tumours implies that there is an alteration in the processing of the sucin in malignancy. A sore detailed study of the processing of the sucin in normal and malignant cells say then give basic information for defining the malignant cell. Moreover, since the specificity of the reaction of the antibody SM-3 for tumours is better than that of antibodies developed against the intact sucin, this antibody say prove to be a sore effective diagnostic tool for the detection of breast cancer cells in tissue sections, tissue fluids and cells. The reactive components are membrane associated as well as intracellular and in vivo localisation of tumours say also be possible.

Abbreviations

Zhe abbreviations used are: HMFG, human silk fat globule; PBS, phosphate-buffered saline (153 sM KaCl, 3 sM ΣCL, 10 sM Na.HPO , 2 sM ΣM P0 pH 7.4); WGA, wheat germ agglutinin; PNA, peanut agglutinin; HPA, Helix pomatia agglutinin; BSA, bovine serum albumin; SDS, sodium dodecyl sulfate.

SUBSTITUTE SHEET

Example 2

Purification and deglycosylation-.of hu.nan milk mucin was conducted as in Example 1 mucin was purified on an HMFG-1 antibody.

The stripped sucin preparations were separated by electrophoresis through HaDodSO./polyacrylamide gels (10%) and silver atained by two methods, one of which can be used to stain highly glycosylated proteins (22,23).

Preparation of polyclonal rabbit antiserum to Btri-pped core protein

One New Zealand White rabbit was immunized with 100 μg of the partially stripped core protein in complete Freund's adjuvant (Gibco). Booster injections of 500 μg of the totally stripped core protein were administered in incomplete Freund's adjuvant (Gibco) 3 and 4 weeks after the initial injection and the rabbit was bled one week later. Ten microliters of immune serum (75 μg/ml protein) precipitated 200 ng of fully stripped core protein in a Protein A assay (24) and detected it on immunoblots. The immunoglobulin fractions of rabbit preimmune and rabbit anti-mucin core protein were prepared by adding ammonium βulfate to 50% saturation. The resulting pellet was resuspended in one-half the original serum volume of PBS and dialyzed against the same buffer. After dialyβiβ, only residual precipitate was removed by centrifugation. Immunoglobulin fractions were stored is aliquots at -20 * C.

SUBSTITUTE SHEET

Descri-ption of MAbs used

In addition to the polyclonal antiserum used for ini ial screening , a cocktail of two MAbs, SM-3 «d »*-« <•* « * "P i * *> ** ich « w iβe the ucin core protein (20) and HMFG-1 and BM7G-2 (1 , 14) were used to screen the purified plaques , the β-galactosidasβ fusion proteins and tor immunopreeipitationβ from in vitro translated proteins . Other MAbs used were a monoclonal anti-β-«alactosidase antibody (25) which was a gift from H. Durbin (ICRF, London) , an anti-interferon antibody , ST254 (24) , LE61 , a keratin antibody (26) and M1 8 which recognizes a carbohydrate structure on the milk mucin (27) .

In Vitro translation of proteins

RNA was isolated from the human breast cancer cell line MCF-7 using the guanidium isothiocyanate method of Chirgwin et al . (28) and poly(A) RNA wae purified by chromatography using oligo (dT)-cellulose (New England Bio Labs ) . The poly(A) RNA was translated in a reticulocyte lysate system (Amersham) in the presence of [ S] methionine (1 000 Ci/mmole ; 1 Ci = 37 GBq, Amersham) . Samples containing 5 x 1 0 acid insoluble cpm were precipitated in a protein A assay (24) using MAbs SM-3, SM-4, HMFG-1 , HMFG-2 and a control antibody to human interferon. The antibody-aelected proteins were then separated on a 10% HaDodSO /polyacrylamide gel , impregnated with Amplify (Amersham) and exposed to IAR-5 film (Kodak) at -70 *C.

The MAbs SM-3 and SM-4 (SM refers to stripped mucin) show strong reactivity with the partially and fully stripped core protein but no reactivity with the fully glycosylated mucin (20) .

Ϊ UBSTΓΓUTE SHEET

Antibody screening of λgtπ library and protein blotting

The λgt11 library used in this study was constructed from mRNA isolated from the human breast cancer cell line MCF-7 and was generously provided by Philippe Walter and Pierre Chambon (Strasbourg, France) . The poly (A) EHA used for the preparation of the randomly primed library was prepared from mRNA that aedimβnted faster than 8S rRNA and was enriched in estrogen receptor (29). The library was made essentially as described by Huynh et al. and Young and Davis (30-32) g and contained approximately 1 x 10 rβcombinants per μg of RNA. Between B5% and 95% of the plaques contained inserts.

The phage library was plated onto bacterial strain T1090 and grown for 3 hr at 42"C. After isopropyl β-D-thiogalactoside (IPTG) induction and 3 hr of growth at 37"C, filters were prepared from each plate and screened with anti-mucin core protein antibody by the method of loung and Davis (32). The first antibody used in screening was the rabbit antiserum raised against the stripped core protein prepared as described above. Prior to use in screening, the antiserum was diluted 1:200 in PBS containing Λ % bovine serum albumin (PBS/BSA). Preabsorption with T1090 bacterial lysate was not found to be necessary. The nitrocellulose filters (Schleicher and Schuell) were blocked by incubation in PBS containing 5% BSA for 1 hr at room temperature with gentle agitation. The filtera were incubated at room temperature overnight with a 1:200 dilution of antiserum in heat sealed plastic bags. The filters were washed 5 x 5 min in PBS/BSA, and bound antibody was detected by using horseradish peroxidase-conjugated sheep anti-rabbit antiserum (Dako) diluted 1:500 with PBS/BSA and incubated for 2 hr with the filters. The filters

SUBS

were washed 5 x 5 min in PBS/BSA and 1 x 10 min in PBS before color detection using 4-chloro-1-naphthol (1). Immunoreactive bacteriophage were picked and purified through two additional rounds of screening.

Subsequently, bacteriophage inserts were aubcloned into the EcoRI sites of pUC8 (33) producing the plasmid used most extensively, pMTJC

10. The plasmids were maintained in DH1 cells.

To examine the β-galactosidase-cDNA fusion proteins for immunoreactivity, cell lysates were derived. Lysogens were prepared as described in Young and Davis (34). Cells were pelleted, suspended in Laemmli sample buffer (35) and separated by electrophoresis through

NaDodSO /polyacrylamide gels (10%) and transferred onto nitrocellulose 4 filters as described (1,36). The filters were treated as above for antibody screening.

Northern Analysis

RNA was isolated from tissue culture cells and frozen tissues by the guanidinium isothiocyanate method of Chirgwin et al. (28). Total RNA (10 μg per lane) was denatured by heating at 55 * C for 1 hr in deionized glyoxal and fractionated by electrophoresis through a 1.3% glyoxal gel (38). The RNA was transferred to nitrocellulose (Schleicher and Schuell), prehybridized and hybridized aa described by Thomas (54). Filters were washed down to 0.1X SSC with 0.1% SDS at 65"C and exposed to IAR-5 film (∑odak) at -70 * C with intensifying acrβens.

WBSTΓΠJTE SH EET

Southern analysis

High molecular weight genomic DNA was prepared from white blood cells and cell lines (39,40). These genomic DNAs (10μg) were cleaved with restriction βnzymeβ following the manufacturer's recommended conditions and fractioned through 0.6% and 0.7% agarose gels. Cloned plasmid DNA was cleaved and fractionated on 1.3% agarose. The gels were denatured, neutralized and transferred to nylon membranes (Biodyne) according to the manufacturer's instructions. The EcoRI insert from pMUC10 was separated on a 1% low melting point agarose (Biorad) gel and labelled with [α- P]dCTP by the method of random priming (41) and hybridized to filters at 42 * C. Filters were washed down to 0.1X SSC with 0.1% SDS at 55 * C and exposed to IAR-5 film (Kodak) at -70"C with intensifying βcreens.

Results

Purification and deglycosylation of mucin glycoprotein

Mucin glycoprotein reactive with the monoclonal antibody HMFG-1 was prepared from pooled human breast milk by using an HMFG-1 antibody affinity column, followed by molecular sieve chromatography on Sephadex G-75 in order to remove lower molecular weight components (Figure 7. lane 1). In order to demonstrate the homogeneity of the purified molecule, amino acid analyses of four separate preparations were performed and revealed a fairly consistent composition with serine, threonine, proline, alanine and glycine accounting for 58% of

s ^r,τu Tε SHεετ

the amino acids ( Periodic acid silver stained gels revealed a diffuse band of greater than 400,000 daltons visible only when the gel was treated with periodic acid before the silver stain (Fig. 7, lane 2). No other lower molecular weight bands were visualized on the gel using the silver stain without prior treatment with periodic acid.

The purified material was subjected to treatment with hydrogen fluoride to remove the 0-linked sugars that are characteristic of mucin glycoproteins. Two different reaction conditions were used which resulted in a partially deglycosylated core protein (treated at 0*C for 1 hr) and a fully deglycosylated core protein (treated at room temperature for 3 hr) as determined by iodinated lectin binding following separation by gel electrophoresis and transfer to nitrocellulose paper (20). The partially deglycosylated core protein was reactive with wheat germ agglutinin, peanut agglutinin and helix pommatia lectin (which recognizes the linkage sugar N-acetylgalactoβamine) whereas the fully stripped protein showed no reactivity with any of these three lectins.

The hydrogen fluoride treated core protein was separated by electrophoresis through NaDodSO./polyacrylamide gels (10%) "d silver stained. Silver staining revealed that the predominant component of the partially stripped mucin was a high solecular weight band of about 400 kd, although faint bands of lower molecular weight could also be observed (Fig.B , lane 1). Since the high molecular weight material ahowed a somewhat increased mobility in the gel and reacted with the lectin recognising the linkage sugar, it can be assumed that some sugars had been removed. The fully stripped mucin consisted of two bands of about 68 kd and 72 kd (Fig. 8 , lane 2).

Si irs:

Antibody reactive proteins produced by MCF-7 cells

The MCF-7 breast cancer cell line expresses large amounts of HMFG-1 and -2 reactive material on its cell surface (14) and was thus judged to be a suitable source of mRNA for a cDNA library. Before proceeding to acreen the MCF-7 library with the monoclonal antibodies, they were teβted for their ability to precipitate a component from in vitro translation products produced from MCF-7 mRNA. Poly (A) RNA from MCF-7 was prepared and translated in vitro. Proteins from the translation reaction were immunoprecipitated using the monoclonal antibodies HMFG-1, HMFG-2, SM-3 and SM-4 and displayed by polyacrylamide gel electrophoresis and fluorography (Fig. 9). Two proteins of about 68 kd and 92 kd were immunoprecipitated by SM-3 (lane 2) and SM-4 (lane 1). It was also found that HMFG-1 (lane 4) and HMFG-2 (lane 3) immunoprecipitated these proteins ; however, no bands in these areas were precipitated by an irrelevant monoclonal antibody to human interferon (lane 5). The fact that HMFG-1 and -2 immunoprecipitated these proteins was an unexpected finding as it was previously thought that these MAbs recognize carbohydrate determinants (1). However, we also found that HMFG-1 and -2 react very strongly with the fully atripped, iodinated core protein (20). These results together' with the MAb reactions on the β-galactosidase fusion proteins (see below) confirm that the epitopes for HMFG-1 and - are, at least in part, protein in nature.

The abundance of the core protein aRNA in total cellular poly (A) +

35 RNA was 4% as estimated by comparing the amount of ( S)methionine present as immunoprecipitated protein to the amount of methionine

incorporated into total protein during in vitro translation.

Screening of the cDNA library

The λgt11 cDNA library made from size selected MCF-7 mRNA (see

Methods) was screened initially with the polyclonal antiserum made to the mucin core protein which had been stripped of its carbohydrate.

Screening of 2 i 10 plaques resulted in 11 positive clones, 7 of which were taken successfully through two further rounds of plaque purification.

To demonstrate that the reactivity of the phage donee with the antibody probes was due to antigenic determinants on the cDNA translation product, β-galactosidase fusion proteins were made from all 7 clones. The proteins were separated by electrophoresis, transferred to nitrocellulose paper and probed with a variety of antibodies to the stripped mucin, including the polyclonal antiserum which was used initially to select the clones and a cocktail or SM-3 and SM-4. In addition, HMFG-1 and HMFG-2, the two monoclonal antibodies which originally detected this differentiation and tumour-associated epithelial mucin (1,14) were tested. All 7 clones yielded fusion proteins which were specifically recognized by the polyclonal antiserum, the monoclonal cocktail, and HMFG-2. HMFG-1 antibody reacted with 6 of the 7 fusion proteins and failed to recognize the protein from clone 9 which contains the smallest insert. In every case the strongest signal was given by the HMFG-2 antibody and this reaction is shown in Figure 1Q. Monoclonal antibodiee to keratins and to a carbohydrate epitope on thiβ fully glycosylated mucin were used as controls and showed no reactivity

SUBSTITUTE SHEET

A monoclonal antibody to β-galactosidase was a positive control end the band recognized correlated in every caee with the band recognized by the specific antibodies. The sizes of the fusion proteins varied in proportion to the sizes of the cDNA inserts found in the bacteriophage.

Characterization of c NAs and RNA blot analysis

The inserts from the λ clones were designated pMUC3-10 (omitting pMUC5) and were subcloned into the rector pUC 8 for easier manipulation. The 7 clones were compared to each other for sequence homology. Each of the plasmids was digested with EcoRI and the insert separated on a 1.4% agarose gel. The largest cDNA insert from pMUC O was used to probe the inserts and found to hybridize to all 6 inserts (Fig.ϋ). pMIIC 7 was found to contain two ineerts following digestion with EcoRI; however, only 1 of the inserts hybridized to the pMUCIO probe. The insert bands were not derived from phage DNA since the pMUC10 probe did not hybridize to Hind Ill-digested λ phage DNA

As shown by agarose gel electrophoresis (Fig.11), the inserts vary in size from about 200 to up to about 1800 bp. The largest insert from pMUCIO has been used as the hybridization probe in all subsequent experiments.

Because the λMUC clones were identified only by antibody binding, we needed additional assurance that they were indeed coding for the breast epithelial mucin. To determine the authenticity of pMUCIO, we correlated the presence of mRNA hybridizing to the clone with mucin expression in various cell lines. As shown in figure 2,the cDNA hybridized to two transcripts of 4.7 kb and 6.4 kb in the RNA from the

SUBSTITUTE SHE!

breast cancer cell lines MCF-7 and T47D which were βhown previously to express the HMFG-2 antigen (1,14). Significantly, the pMUCIO probe hybridized to transcripts of approximately the βa e size in RNA extracted from normal mammary epithelial cells cultured from milk (42). A third band of 5.7kb can be seen in the RNA from these normal cells. In contrast, three human cell types that lack the mucin, breast fibroblasts, Daudi cells and HS578T, a carcinosarcoma line derived from breast tisβue (43), showed no detectable pMUC10-related mRNA. The 6.4 kb band appears to be the most adundantly expressed. The preβence of at least two sizes of mRNA from MCF-7 cells correlates with the immunoprecipitation of two proteins of (molecular weights 68 kd and 92 kd) from in vitro translated mRNA from MCF-7 cells. The normal mammary epithelial cells were derived from pooled milk samples and the additional transcript observed may be due to polymorphisms among individuals.

Genomic DNA blot hybridization and detection of a restriction fragment length polymorphism (RFLP)

Genomic DNA was prepared from a panel of ten individuals consisting of six unrelated individuals and a family of four, from three cell lines. The DNAs which were digested with Hinfl or EcoRI and blotted and hybridized to the radiolabelled pMUCIO insert, exhibit restriction fragment length polymorphisms. The restriction fragments from the ten individuals and three cell lines are ahown in figure . The pattern consists of either a single band or a doublet of sizeβ ranging from 3400bp to 6200bp in the Hinfl digest (with the exception of the ZR75-1 DNA in lane 12, figure 13Awhich shows three bands) or

SUBSTITUTE SHEET

from 8200bp to 9600bp in the EcoRI digest (Figure 13 " B).There appears to be a continuous distribution of the fragment sizes which implies a high in vivo instability at the locus. The pattern of fragments observed in the family of four (lanes 1-4) suggests that these fragments are allelic. Preliminary studies of the DNA made from white blood cells of normal, related individuals indicate the existence of a number of independent alleles with an autosomal codominant mode of inheritance These studies will be the subject of a separate investigation.

Discussion

The cDNA clones described here which were obtained from the MCF-7 λgt11 library were selected using polyclonal and monoclonal antibodies prepared against a normal cellular product, the silk mucin in its deglycosylated form. This was done because it was easier to obtain large quantities of the sucin for stripping than to prepare similar quantities of immunologically related glycoproteins expressed by breast cancer cells (44). The fact that the antibodies did select for cDNA coding for nonglycosylated core protein molecules in MCF-7 cells, strongly suggests that the glycoproteins in these cells, which were originally detected by their reaction with antibodies to the silk sucin, contain the same core protein aa this sucin. This is confirmed by the detection of sRNAs of approximately the aame aizes in the normal and malignant cells, using one of the probes isolated from the MCF-7 library. We will therefore refer to the antibody reactive glycoproteins on breast cancer cells as mucins, bearing in mind that

SUBSTITUTE SHEET

their processing may be different resulting in molecules of different molecular weights but with the same core protein as that of the milk mucin.

Seven clones were obtained from the MCF-7 library of which the largest was 1β00kb. This clone cross hybridized with the other 6 smaller clones. The β-galactosidase fusion proteins expressed by six of the cross-hybridizing lambda clones were reactive with the polyclonal antiserum directed against the mucin core protein as well as with four well-characterized monoclonal antibodies directed to various epitopes on the stripped core protein, SM-3, SM-4, HMFG-1 and

HMFG-2 (14,20). The smallest lambda clone, λMUC9, produced a β-galactosidase fusion protein which reacted with three of the four monoclonal antibodies and with the polyclonal antiserum.

The surprising result that the extensively characterized HMFG-1 and HMFG-2 monoclonal antibodies reacted strongly with the lambda plaques and the fusion proteins and could immunoprecipitate proteins from in vitro translated mRNA provides strong evidence that these clones do indeed code for a portion of the mucin core protein.

Although previous evidence βuch as resistance to fixation, boiling, treatment with dithiothreitol and NaDodSO. and the presence of

4 multiple epitopes on the molecule suggested that these were carbohydrate (1 ), it has now been established that the epitopes of the HMFG-1 and HMFG-2 monoclonal antibodies are definitely protein in nature. Carbohydrate may be required to obtain the strongest binding, either as part of the epitope or by conferring some conformational change on the protein portion, but part of the antigenic determinant sust consist of an amino acid sequence. Since these two MAbs are

SUBSTITUTE SHE, E ET

-48 -

reactive with the fully glycosylated milk mucin as well as the stripped core protein, this data means that the intact molecule contains areas of naked peptide which contribute to the antigenic sites for these two antibodies.

Confirmatory evidence that pMUCIO codes for the mammary sucin core protein is provided by RNA blots. The relative abundance of mRNA in the breast cancer cell lines MCF-7, T47D, ZR-75-1 and in normal mammary epithelial cells corresponds to the antigen expression by these cells as measured by the binding of the HMFG-1 and HMFG-2 monoclonal antibodies. Cell types which are negative for antigen expression such as human fibroblasts, Daudi cells and HS578T, a carcinosarcoma line derived from breast (14), are negative in RNA blot hybridizations. A fortuituous observation made with the ZR-75-1 cells yielded indirect strong evidence that pMUCIO does indeed code for the mucin glycoprotein core protein. This cell line, which routinely expresses large amounts both of mRNA and antigen, yielded one preparation of RNA which was unexpectedly negative by blot hybridization. It was subsequently found that those particular ZR-75-1 cells from which the RNA had been sade had lost the expression of the antigen as well at this time (as determined by reaction with HMFG-1 and 2). Different passage numbers of the ZR-75-1 cells were recovered and ahown once again to express both antigen and message. The sizes of the messages, 4.7 kb and 6.4 kb, are quite large, since a 68 kd or 92 ∑d protein would need only about S kb to code for the protein portions. This suggestβ that a large portion of the mRNA maybe untranslated. Efforts are underway to obtain a full-length clone.

SUBSTITUT E S

Thus, the cDNA clones presented here represent a portion of the gene coding for the human mammary mucin which is expressed by differentiated breast tissue as well as by most breast cancers. The major proteins precipitated from in vitro translation products of RNA from MCF-7 cells by antibodies to the silk sucin core protein (68Ed) have an apparent solecular weight of 68Ed and 92£d. These proteins, produced by the breast cancer cell therefore share epitopes with the 68∑d core protein of the silk sucin (20). Whether a similar 92∑d protein is also produced by normal mammary epithelial cells, and is truncated or destroyed by HF treatment is not yet clear. MCF-7 cells biosynthetically labelled with 14C amino acids yield upon immunoprecipitation with HMFG-1 and HMFG-2 antibodies, two glycosylated proteins of 320 kd and 430 kd, and it is possible that each of these glycoproteins utilizes only one core protein of either 68∑d or 92∑d. Alternatively, each of the glycoproteins could contain both the 92∑d and 68£d proteins either in different proportions or variably glycosylated. Further screening of the library may yield full length cDNAs coding for both sizes of the immunologically related core proteins. Since there appears to be only a single gene (based on Southern blot data obtained by using a partial cDNA probe) , it is probable that the multiple messages arise by alternative RNA splicing and this would explain the fact that they contain common sequences. Although a core protein of 68 kd appears to be smnTI to yield a fully glycosylated molecule of greater than 300 kd which contains 50% carbohydrate, there is evidence that such a structure for mucins is possible. Ovine submaxillary mucin has a reported molecular weight of 1 x 10 daltons (45), yet its protein

SUBSTITUTE SHEET

-50-

core consists of 650 amino acids resulting in a molecule of 58 kd (46).

The sucins which are detected with HMFG-1 and HMFG-2 MAbs on immunoblots of tumours and breast cancer cell lineβ show variations in size from 80 kd to 400 kd in the solecular weights of the tumour sucin molecules (1,47). Using theBe same antibodies which detect high molecular weight sucins present in normal urine, a polymorphism has indeed been shown to be genetically determined (48). Although the very low molecular weight components are likely to represent precursor forms of the mucin which appears to be incompletely processed in many tumour cells (20), the variations in the higher molecular weight components are likely to be due to this genetic polymorphism. It was unclear, however, whether the structural basis of the polymorphism was due to the genetically determined protein or to the carbohydrate portion of the sucin. The detection of restriction fragment length polymorphisms in the Southern blotting experiments using the mucin probe suggest that the sucin polymorphism occurs at the level of the DNA which codes for the protein. Preliminary sequence data /suggest that the basis for this polymorphism is a region of variable tandem repeats present in the protein coding sequences. This structural feature say be responsible for the generation of the sany allelic restriction fragments at the sucin locus. We are presently investigating the basis of the sucin polymorphism by a Southern blot survey of DNA from white blood cells of normal, related individuals whose Inheritance pattern of urinary mucins has been determined. In addition, we are examining DNA preparations sade from the white blood cells and tumours of individual breast cancer patients

SUBSTITUTE SHEET

to determine if there is any discordance between genotype in the paired samples, since tandemly repeated DNA may provide an unstable site where recombination or amplification could occur.

The presence of sucins in the majority of carcinomas and their association with the differentiation of mammary epithelial cells makes it particularly important to identify regions involved in the tissue specific and developmental regulation of the gene. Moreover, the introduction of a functional sucin gene into cells should provide insightβ into the role of this molecule in breast epithelial differentiation and possibly enable us to identify any alterations in the function or expression of the mucin which are related to malignant transformation in the human breast.

SUBSTITUTE SHEET

Abbreviations

The abbreviations are as follows: PBS, phosphate-buffered B aline ; MAb, monoclonal antibody; IPTG, isopropyl β-D-thiogalactoside; bp, base pair(s); ∑b, Jcilobase(s) .

SUBSTITUTE SHEET

- -

TABI£ 1

Amino acid composition of the human milk mucin - comparison with PAS-0

Amino acid HMFG-1 purified Extensively stripped PAS-0 silk sucin silk sucin (Shisizu & lamauchi

1982)

Asp 6.1 7.2 6.4

Thr 9.4 9.7 9.8

Ser 9.1 13. 0 13.1

Glx 6.3 9.6 8.3

Pro 14.8 14.4 12.0

Gly 8.1 1 0.1 12.2

Ala 12.3 11 .9 13.0

Cys Not analysed Not analysed 0.5

Tal 6.0 6.3 5.3

Met 0.5 0.4 0.6 lie 1 .6 1 .7 1 .9

Leu 4.5 4. 8 3.7

Tyr 2.0 0.9 1 .6

His 3.2 2.5 3.8

IflTB 2.8 3.3 2.2

Arg 4.0 4.0 3.9

Table 2 Reactivity of the antibodies on intact, partially and totally deglycosylated milk mucin

125

I cpm bound

Antibody Intact molecule Partially Totally stripped sucin stripped sucin

5.17 8,524 11 , 925 5,780

9.13 525 3,000 3,328

SM-3 465 15,414 9,200

SM-4 816 16,750 9,561

HMFG-1 32,000 33,768 9,494

HMFG-2 29,500 29,230 15,832

NS2 medium 397 845 650

The binding of the antibodies to iodinated intact, partially and totally deglycosylated silk sucin was assayed using the protein A plate sethod as described in Materials and Methods.

S

References

1. Burchell, J.M., Durbin, H. and Taylor-Papadimitriou, J. (1983) J. Immunol. 131, 508-513.

2. Bramwell, M.E., Bhavanandan, 7. P. , Wiseman, G. and Harris, H. (1983) Br. J. Cancer 48, 177-183.

3. McΣLhinney, R.A. , Patel, S. and Gore, M.E. (1985) Biochem. J. 227, 155-162.

4. Hilkens, J., Euijs, F. , Hilgers, J. , Hagemann, Ph., Calafat, J. , Sonnenberg, A. and Van der Valk, M. (1984) Int. J. Cancer 34, 197-206.

5. Tagliabue, E. , Porro, G. , Barbanti, P., Delia-Torre, G. , Menard, S., Rilke, F., Cerasoli, S. and Colnaghi, M. (1986) Hybridoma 5, 107-115.

6. Johnson, V.G. , Schlom, J. , Paterson, A.J., Bennett, J. , Magnani, J.L. and Colcher, D. (1986) Cancer Res. 46, 850-857.

7. Sekine, H. , Ohno, T. and ∑ufe, D.W. (1985) J. Immunol. 135, 3610-3615.

8. Ormerod, M.G. , Steele, Σ. , Edwards, P.A.W. and Taylor-Papadimitriou, J. (1984) J. Expt. Path. 1, 263-271.

9. Wilkinson, M.J., Howell, A., Harris, J. , Taylor-Papadimitriou, J. , Swindell, R. and Sellwood, R.A. (1984) Int. J. Cancer 33, 299-304.

10. ∑ufe, D., Inghirasi, G. , Abe, M. , Hayes, D. , Justi-Wheeler, H. and Schlom, J. (1984) Hybridoma 3, 223-232.

11. Price, M.R., Edwardβ, S. , Robins, R.A., Hilgens, J., Hilkens, J. and Baldwin, R. (1966) Bur. J. Can. EL in. Oncol. 22, 115-117.

12. Johnston, W.W. , Szpak, C.A., Lottick, S.C., Thor, A. and Schlom, J. (1985) Cancer Res. 45, 1894-1900.

_ 56 .

13. Rasmussen, B.B., Pedersβn, B.Y. , Thorpe, S.M., Hilkens, J., Hilgers, J. and Rose, C. (1986) Cancer Res. 45, 1424-1427.

14. Taylor-Papadimitriou, J., Peterson, J.A., Arklie, J., Burchell, J. , Ceriani, R.L. and Bodmer, W.F. (1981) Int. J. Cancer 28, 17-21.

15. Burchell, J. , Wang, D. and Taylor-Papadimitriou, -J. (1984) Int. J. Cancer 34, 763-768.

16. Shifflizu, M. and lamauchi, Σ. (1982) J. Biochem. 91, 515-519.

17. Ormerod, M.G. , Steele, Σ. , Westwood, J.H. and Mazzini, M.N. (1983) Br. J. Cancer 48, 533-541.

18. Taylor-Papadimitriou, J. , Lane, E.B. and Chang, S.E. (1983) In, Understanding Breast Cancer: Clinical and Lab oratory Concepts. (Rich, M.A. , Hager, J.C. and Furmanski, P. Eds.) Marcel Dekker, Inc. New lork and Basel, pp. 215-246. 19. Chang, S.E. and Taylor-Papadimitriou, J. (1983) Cell Diff. 12, 143-154.

20. Example l " above.

21. Mort, A. and Lamport, D. (1977) Anal. Biαchsm._ 82 *. 289-.309.

22. Wray, W., Boulikaβ, T. t Wray, Σ.P. and Hancock, R. (1981) Anal. Biochem. 118, 197-203.

23. Dubray, G. and Bezard, G. (1982) Anal. Biochem. 119, 325-329.

24. Shearer, M., Taylor-Papadimitriou, J., Griffin, D. and Balkwill, F. (1984) J. Ismunol. 133, 3096-3101.

25. Durbin, H. and Bodmer, W.F. (1986) J. Immunol. Meth., In Press.

26. Lane, E.B. (1982) J. Cell Biol. 92, 665-673.

SUBSTITUTE SHEET

27. Gooi, H.C., Jones, N.J., Hounsell, E.F., Scudder, P., Hilkens, J. , Hilgers, J. and Feizi, T. (19B5) Biochem. Biophyβ. Res. Commun. 131, 543-550.

28. Chirgwin, J.M. , Przybyla, A.E., MacDonald, R.J. and Rutter, W.J. (1979) Biochem. 18, 5194-5199.

29. Walter, P., Green, S., Greene, G. , ∑rust, A., Bornert, J.-M., Heltsch, J.-M., Staub, A., Jensen, E., Scrace, G., Waterfield, M. and Chambon, P. (1985) Proc. Natl. Acad. Sci. USA. 82, 7889-7893.

SO. Huynh, T.V. , loung, R.A. and Davis, R.W. (1985) In, DNA Cloning: A

\

Practical Approach, ed. Glover, D.M. (IRL, Oxford), Vol. 1, pp.

98-121.

31. ϊoung, R.A. and Davis, R.W. (1983) Proc. Natl. Acad. Sci. USA 80, 1194-1198.

32. ϊoung, R.A. and Davis, R.W. (1983) Science 222, 778-782.

33. Vieira, J. and Messing, J. (1982) Gene 19, 259-268.

34. oung, R.A. and Davis, R.W. (1985) In Genetic Engineering, eds. Setlow, J.Σ. and Hollaender, A. (Plenum, New ork) , Vol. 7.

35. Laemmli, U.K. (1970) Nature (Lond) 227, 680-685.

36. Towbin, H., Staehelin, ϊ. and Gordon, J. (1979) Proc. Natl. Acad. Sci. USA. 76, 4350-4354.

37. Maniatiβ, T., Fritsch, E.F. and Sambrook, J. (1982) Molecular Cloning: A Laboratory Manual (Coldspring Harbor Laboratory, Cold Spring Harbor, NT).

38. Thomas, P.A (1980) Proc. Natl. Acad. Sci. USA. 77, 5201-5205.

39. Woodhead, J.L., Fallon, R., Figueiredo, H., Langdale, J. and Malcolm, A.D.E. (1986) In Human Genetic Diseaβes, ed. Davies, Σ.E. (IRL, Oxford), p. 56.

40. Old, J.M. (1986) In Human Genetic Diseases, Ed. Davies, Σ.E., (IRL, Oxford), p.4.

41. Feinberg, A.P. and Vogelstein, B. (1984) Anal. Biochem. 137, 266-267.

42. Taylor-Papadimitriou, J., Purkis, P. and Fentiman, I.S. (1980) J. Cell. Physiol. 102, 317-321.

43. Hackett, A.J., Smith, H.S., Springer, Z.L., Owens, R.B., Hβlaon-Rβes, W.A., Riggs, J.L. and Gardner, M.B. (1977) J. Natl. Cancer Inst. 58, 1795-1800.

44. Griffiths, A.B. , Burchell, J. , Taylor-Papadimitriou, J. , Gendler, S.J., Lewis, A. and Tilly, R. (1987) Int. J. Cancer (submitted).

45. Gottschalk, A., Bhargava, S. and Mury, V. In Gottschalk, A. (ed) Glycoproteins their composition, structure and function, pp. 810-829, Eleevier, New ϊork.

46. Hill, M.D.Jr. Reynolds, J.A. and Hill, R. (1977) J. Biol. Chβm. 252, 3791-3798.

47. Taylor-Papadimitriou, J., Millis, R., Burchell, J., Nash, R. , Pang, L. and Gilbert, J. (1986) J. Expt. Path. 2, 247-260. '

48. Swallow, D., Griffiths, B. , Bramwell, M. , Wiseman, G. and Burchell, J. (1986) Disease Markers 3, In Press.

SUBSTITUTE SHEET

49. Abe, M. and ∑ufe, D. Effects of saturational agents on expression and secretion of two partially characterized high solecular weight silk-related glycoproteins in MCF-7 breast carcinoma cells. J. Cell. Fhysiol. 126:126-136, 1986.

50. Arklie, J., Taylor-Papadisitriu, J., Bodser, f.F., Egan, M. and Millie, R. Differentiation antigens •xpressed by epithelial cells in the lactating breast are also detectable is breast cancers. Int. J. Cancer 28:23-29, 981.

51. Bolton, A.E. and Hunter, W.M. The labelling of proteins to high

125 specific radioactivities by conjugation to a I containing acylating agent. Biochem. J. 133:529-536, 1973.

52. ∑arlsson, S., Swallow, D., Griffiths, B., Corney, G. and Hoplinaon, P. A genetic polymorphism of a human urinary sucin. Ann. Hum. Genet. 47:263-269, 1983.

53. ∑earnβy, J.F., Radbruch, A., Liesegang, B. and Rajβwsky, Σ. Anew mouse myeloma cell line that has lost immunoglobulin expression but permits the construction of antibody-secreting hybrid cell lines. J. Immunol. 123: 548-1550, 1979.

54. Melero, J. and Gonzalez-Rodriguez, J. Preparation of monoclonal antibodies against glycoproteins 111a of human platelets. Zur. J. Sioehes. 141:421-427, 1984.

55. Clamp, J.R., Allan, A., Gibbons, R.A. and Roberts, G. Chemical aspects of sucins. Br. Mβd. Bull. 34:25-41, 1978.