WO1995021380A1 - Assay for screening of receptor antagonists - Google Patents

Assay for screening of receptor antagonists Download PDF

Info

Publication number
WO1995021380A1
WO1995021380A1 PCT/US1995/001375 US9501375W WO9521380A1 WO 1995021380 A1 WO1995021380 A1 WO 1995021380A1 US 9501375 W US9501375 W US 9501375W WO 9521380 A1 WO9521380 A1 WO 9521380A1
Authority
WO
WIPO (PCT)
Prior art keywords
gastrin
binding
cck
compound
gbp
Prior art date
Application number
PCT/US1995/001375
Other languages
French (fr)
Inventor
Graham Sherard Baldwin
Original Assignee
Ludwig Institute For Cancer Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AUPM3650A external-priority patent/AUPM365094A0/en
Priority claimed from AUPM6638A external-priority patent/AUPM663894A0/en
Priority claimed from AUPM7779A external-priority patent/AUPM777994A0/en
Application filed by Ludwig Institute For Cancer Research filed Critical Ludwig Institute For Cancer Research
Priority to AU17405/95A priority Critical patent/AU1740595A/en
Publication of WO1995021380A1 publication Critical patent/WO1995021380A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/595Gastrins; Cholecystokinins [CCK]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it

Definitions

  • This invention relates to a method for screening of putative antagonists of binding of gastrin to its receptor, in particular the low-affinity gastrin-cholecystokinin C receptor, more particularly the gastrin-binding protein (GBP), and also of inhibitors of the enzyme activities associated with the GBP.
  • the low-affinity gastrin-cholecystokinin C receptor more particularly the gastrin-binding protein (GBP)
  • GBP gastrin-binding protein
  • Compounds having the ability to block binding of gastrin to GBP or to block the enzyme activities of the GBP are useful in control of cellular proliferation, especially in treatment of neoplastic disease, and are also useful in the control of acid production in the stomach.
  • gastrin has been known for over 80 years to be a stimulant of acid secretion by the parietal cells of the stomach.
  • Gastrin is a small polypeptide hormone which participates in a feedback system for regulating the environment in the lumen of the alimentary tract, and is thought to affect other target organs, including the brain and pancreas. More recently, it has been recognised that gastrin also acts as a growth factor for the gastric mucosa. Both of these functions are presumably mediated by the binding of gastrin to specific receptors on the target cells in the gastrointestinal mucosa. A variety of methods for detection of such
  • Gastrin/CCK-C receptors have been identified on cell lines of a variety of tumour cell lines of epithelial and non-epithelial origin, and on several cell lines of lymphocytic origin.
  • significant levels of gastrin/CCK-C receptors have been observed with cell lines established from carcinomas of the stomach, colon, breast, and vulva, and from malignant melanoma: in addition, lower levels of binding were observed with T- and B-cell lymphoma cell lines, while still smaller levels of binding were observed with promyelocyti ⁇ and myeloid leukaemia cell lines (3).
  • CCK cholecystokinin
  • Progastrin but not mature amidated gastrin, was also produced by all (5/5) colonic carcinoma cell lines tested. Exogenous gastrin 17 or its derivatives enhanced the growth of xenografts of 60% of colon
  • colon carcinoma cell lines do not increase either DNA or protein synthesis in vitro in response to exogenous gastrin, perhaps because they are already maximally stimulated by an autocrine gastrin-like peptide. In some cases previously
  • the antagonist proglumide which does not discriminate clearly between the three known classes of gastrin/CCK receptors, increases the survival time of mice bearing tumours derived from the mouse colon carcinoma line MC26 (1). Proglumide also reverses the increase in tumour volume observed in tumour-bearing mice after treatment with pentagastrin. Proliferation of 6 colon carcinoma cell lines in vitro is inhibited by the non-selective antagonists proglumide and benzotript, with IC 50 values in the mM range (2). Furthermore, in the case of the HCT 116 cell line proliferation is also inhibited by an anti-gastrin antiserum (2).
  • the high concentrations of gastrin required to reverse inhibition by both antagonists and antibodies suggest that neither pancreatic CCK-A nor gastric gastrin/CCK-B receptors are involved, but are consistent with the involvement of the low affinity
  • gastrin/CCK-C receptor which we have identified on the surface of several gastric and colonic carcinoma cell lines (2).
  • the failure of the selective antagonists 1.364,718 and L365,260 to inhibit proliferation of HCT 116 cells at concentrations as high as 1 ⁇ M confirms that neither CCK-A nor gastrin/CCK-B receptors participate in the autocrine loop (4).
  • GBP gastrin-binding protein
  • sequences of the naturall.v occurring protein (Baldwin et al: Int. J. Biochem 26 529-538). These sequences have been used to generate probes which have enabled cloning of the cDNA encoding the porcine GBP from both c-DNA and genomic libraries.
  • porcine GBP The sequence of porcine GBP, deduced from the cDNA sequence, is related to those of enzymes having enoyl CoA hydratase activity and to enzymes having 3-hydroxy-acyl-CoA dehydrogenase activity. These enzymes are
  • the N-terminal half of the GBP is related to the enoyl-CoA hydratase family, while the C-terminal half is related to the 3-hydroxy-acyl-CoA dehydrogenase family (6).
  • the sequence of the porcine GBP is closely related to the sequence of the ⁇ -subunit of a rat
  • MTP mitochondrial trifunctional protein
  • affinities of antagonists for the GBP correlate well with their affinities for the gastrin/CCK-C receptor, and with their potencies for inhibition of colon carcinoma cell growth.
  • gastrin/CCK receptor antagonists we have found that other classes of compounds inhibit the cross-linking.
  • non-steroidal anti-inflammatory drugs inhibit not only the cross-linking, but also proliferation of a human colon carcinoma cell line.
  • gastrin/CCK receptor antagonists and compounds such as non-steroidal anti- inflammatory drugs;
  • the present invention provides a method of identifying a compound having the ability to block the low-affinity gastrin-cholecystokinin type C receptor, comprising the step of measuring the ability of said compound to block the binding to gastrin-binding protein of a compound selected from the group consisting of a gastrin-related peptide, a cholecystokinin-related peptide, an antagonist of gastrin or of cholecystokinin, an acyl CoA, an enoyl CoA, an antibody to gastrin, and an antibody to cholecystokinin.
  • the binding may be reversible or irreversible, but is preferably irreversible.
  • inhibition of irreversible binding is assessed by measuring the ability of the compound to block cross-linking of a gastrin
  • Cross-linking may be effected by an agent which is homobifunctional, heterobifunctional, or photoreactive.
  • agent which is homobifunctional, heterobifunctional, or photoreactive.
  • disuccinimidyl suberate disulfosuccinimidyl suberate, and ethylene glycol bis-(succinimidyl)-succinate.
  • Heterobi-functional cross-linking reagents include succinimidyl-4-(p-maleimidophenyl) butyrate, which has been found to be equally effective in cross-linking the gastrin peptide to the gastrin binding protein.
  • Suitable photoreactive reagents include N-hydroxysuccinimidyl-4-azidobenzoate.
  • the gastrin peptide is labelled with a detectable marker, such as a radioactive label, a fluorescent label, or biotin.
  • a detectable marker such as a radioactive label, a fluorescent label, or biotin.
  • the gastrin peptide may be
  • iodinated or may be synthesised using one or more amino acids comprising a radioactive marker such as 14 C or 3 H
  • Native gastrin gastrin 17 , contains no free amino groups, and is therefore incapable of reacting with the cross-linking reagent.
  • the minimum gastrin sequence required for binding to the gastrin/CCK-C receptor is known to be the C-terminal tetrapeptide, so N-truncated peptides are expected to be suitable.
  • [Leu 15 ] gastrin 17 and [Nle 15 ] gastrin 2,17 have equal ability to bind to isolated canine parietal cells or parietal cell plasma membranes: substitution of methionine by leucine isomers does not interfere with the binding or biological activity of gastrin.
  • a particularly preferred gastrin peptide is [Nle 15 ] gastrin 2,17 , a derivative which lacks the amino-terminal pyroglutamate residue. Substitution of nor-leucine for the naturally occurring methionine at position 15 prevents formation of oxidised methionine derivatives during iodination. Although [Met 15 ]-gastrin 2,17 could be used, there would be some loss of activity for this reason. It is contemplated that gastrin peptides extended at the C-terminus could also be used.
  • a displacement-type assay using reversible binding is employed.
  • the ability of the putative antagonist to prevent binding of a known agonist or antagonist of very high affinity is measured, said known agonist or antagonist being labelled with a detectable marker.
  • the known agonist or antagonist has at least the binding affinity of benzotript.
  • the known antagonist is suitably labelled with 3 H.
  • the method of the invention provides a general screening method for identifying antagonists of the
  • compounds are useful for treatment of diseases, especially neoplastic diseases, involving rapidly proliferating cells. Such antagonists are also useful for controlling gastric acid secretion.
  • Non- steroidal anti-inflammatory drugs which are especially useful for treatment of neoplastic diseases of the gastrointestinal tract, in particular colorectal cancer.
  • NSAIDS non- steroidal anti-inflammatory drugs
  • GBP is widespread in the body, and consequently treatment of cancers of non-gastrointestinal origin is also within the scope of this invention.
  • the NSAID is an inhibitor of prostaglandin activity.
  • inhibition of the enoyl CoA hydratase and/or 3-hydroxyacyl CoA is another preferred embodiment, inhibition of the enoyl CoA hydratase and/or 3-hydroxyacyl CoA
  • dehydrogenase activities intrinsic to the GBP is assayed. Either activity can be measured in either direction, but is conventionally assayed spectrophotometrically by the decrease in enoyl CoA absorption on hydration or by the decrease in NADH absorption on oxidation. Alternative assays could employ radiochemically labelled substrates.
  • Gastrin-binding protein suitable for use in the method of the invention may be partially purified or purified naturally occurring GBP, or may be recombinant GBP.
  • a high-affinity labelled antagonist in a reversible binding assay, a high-affinity antibody, such as a monoclonal antibody, labelled with a detectable marker may be used.
  • a detectable marker for example, it is contemplated that an antibody capable of blocking binding of an acyl CoA or of gastrin to GBP may be used.
  • the invention in another aspect therefore provides a method of treating neoplastic disease
  • the neoplastic disease is selected from the group consisting of gastrointestinal cancers such as colon carcinoma and gastric carcinoma, mammary
  • the invention provides a pharmaceutical composition of a compound with the ability to inhibit the binding of a gastrin analogue to the low-affinity gastrin-cholecystokinin type C receptor,
  • the compound is a non-selective antagonist of the gastrin-cholecystokinin receptor.
  • the antagonist will have a IC 50 value" in the millimolar range or lower.
  • compositions of the invention may comprise an antagonist either alone, or in combination with other pharmaceutically-active molecules such as antacid compounds or anti-cancer agents.
  • Anti-cancer agents may include cytokines.
  • the invention provides a method of treatment of a neoplastic disease, comprising the step of inducing production of antisense gastrin mRNA in neoplastic tissue of an animal in need of such treatment.
  • Tissue-specific production of anti-sense mRNA can be achieved by targeting of corresponding DNA to the desired tissue, using tissue-specific promoters.
  • Figure 1 shows gastrin analogues and antagonists inhibit covalent cross-linking of 125 I-[Nle 15 ]-gastrin 2,17 to the 78kDa GBP.
  • Figure 2 shows binding of fatty acyl CoA derivatives to the 78 kDa GBP.
  • Cross-linking of 125 I-[Nle 15 ]-gastrin 2,17 to the 78 kDa GBP was measured by phosphorimager (insets) in the presence of increasing concentrations of crotonyl CoA, a substrate of enoyl CoA hydratase, or of acetoacetyl CoA, a product of 3-hydroxyacyl CoA dehydrogenase, and expressed as a percentage of the value obtained in the absence of competitor. Lines of best fit for inhibition by
  • acetoacetyl CoA and crotonyl CoA were obtained with the program LIGAND. Values for IC 50 and for the predicted ordinate intercept were as follows: Crotonyl CoA (•), 43 ⁇ M, 76%; acetoacetyl CoA ( ⁇ ), 74 ⁇ M, 70%. No inhibition was observed with NADH ( ⁇ ), a cofactor for 3-hydroxyacyl CoA dehydrogenase.
  • Figure 3 shows that anti-proliferative gastrin/CCK receptor antagonists target the 78 kDa GBP.
  • the mean IC 50 values (Table 1) for the inhibition of cross-linking of 125 I-[Nle 15 ]-gastrin 2,17 to the 78 kDa GBP by gastrin analogues (closed symbols as in Figure 2) and antagonists (open symbols as in Figure 2) were determined as described in the legend to Figure 2, and compared with the IC 50 values for (A) inhibition of binding of 125 I-CCK 8 labelled using Bolton-Hunter reagent to the cloned human gastrin/CCK-B receptor expressed in COS cells (analogues) (8) and of 125 I-CCK 33 to the gastrin/CCK-B receptor on guinea pig gastric glands (antagonists) (9), or (B) inhibition of binding of 125 I-gastrin 17 to the gastrin/CCK-C receptor on HCT 116 colon carcinoma cells (2). Lines
  • Figure 4 illustrates processing and sequences of gastrin-related peptides.
  • the C-terminal sequence of human progastrin is shown using the one letter code.
  • the corresponding region of porcine progastrin is only shown where it differs from human progastrin.
  • the indicated peptides (numbered from the N-terminus of gastrin 1-17 ) were synthesised according to the human sequence, with the N-terminal glutamic acid of gastrin 1-4 , gastrin 1-11 , gastrin 1-17 and gastrin 1-17 G cyclized to pyroglutamate.
  • Figure 5 shows binding of gastrin-related peptides to the 78 kDa GBP.
  • Figure 6 illustrates how expression of antisense gastrin inhibits cell proliferation.
  • Figure 7 shows the inhibition of YAMC cell proliferation by gastrin/CCK receptor antagonists.
  • Proliferation of YAMC cells in the presence of increasing concentrations of the non-selective gastrin/CCK receptor antagonists proglumide ( ⁇ ) or benzotript ( ⁇ ) was measured with the MTT assay at two days after seeding.
  • Antagonists were added one day after seeding.
  • the means of triplicate absorbance readings are shown as a percentage of the mean absorbance reading obtained on day 1 for the cells grown in 1% fetal calf serum; error bars represent the standard error of the mean. Lines of best fit were
  • FIG. 8 shows the inhibition of growth of
  • Points are the mean values of 5 samples, and lines of best fit were obtained with the program LIGAND. Bars represent 1 SD.
  • FIG. 9 illustrates proliferation of LIM 1215 colon carcinoma cells in the presence of increasing
  • NSAIDS concentrations of NSAIDS, measured with the MTT assay as described for Figure 7.
  • Cells were grown in a medium containing 10% foetal calf serum, and NSAIDS were added one day after seeding. The means of triplicate absorbance readings were expressed as a percentage of the mean
  • Figure 10 shows that the inhibition of proliferation of LIM 1215 cells by NSAIDS is not correlated with the ability of the NSAIDS to inhibit either
  • cyclooxygenase I Panel A
  • cyclooxygenase II Panel B
  • the IC 50 values for inhibition of proliferation of LIM 1215 cells were measured as described for Figure 8, and are summarized in Table 4.
  • IC 50 values for inhibition of cyclooxygenase I and cyclooxygenase II were taken from
  • Figure 11 shows the ability of four different NSAIDS to block cross-linking of gastrin 2,17 to GBP in the assay described above.
  • Figure 12 shows the correlation between the ability of NSAIDS to inhibit proliferation of LIM 1215 cells and their ability to inhibit cross-linking of gastrin 2/17 to GBP.
  • GBP gastrin-binding protein
  • Na 125 I was from NEN, North Ryde, Australia.
  • the 78 kDa GBP was partially purified from detergent extracts of porcine gastric mucosal membranes (5), and crosslinked to 125 I-[Nle] 15 -gastrin 2,17 with
  • protease inhibitors were included in all buffers to prevent proteolysis: pepstatin, 1 ⁇ M; benzamidine, 1 mM; hexamethylphosphoramide, 0.1% (w/v); aprotinin, 500 units/ml.
  • pepstatin 1 ⁇ M
  • benzamidine 1 mM
  • hexamethylphosphoramide 0.1% (w/v)
  • aprotinin 500 units/ml.
  • the products of the cross-linking reaction were separated by polyacrylamide gel electrophoresis, and radioactivity associated with the 78 kDa GBP was detected and quantitated with a phosphorimager (Molecular Dynamics, Sunnyvale, CA).
  • a phosphorimager Molecular Dynamics, Sunnyvale, CA
  • nonpolyposis colorectal cancer (20), were obtained from Dr. R.H. Whitehead, Ludwig Institute for Cancer Research, Melbourne.
  • a colorimetric assay (21) was used to measure cell proliferation. Briefly, 10 4 cells were seeded in a 96 well plate in medium containing 1% fetal calf serum. The medium was replaced the next day with fresh medium
  • LIM 2412 cells were grown in RPM1 1640 medium, and LIM 1215 cells in the same medium containing 1 mM thioglycerol, 25 units/ml insulin, 1 mg/ml hydrocortisone, 5 mg/ml bovine serum albumin, 10 ⁇ g/ml iron-saturated transferrin and 1.5 mg/ml glutamine. Both cell lines were cultured in the absence of serum.
  • Proglumide 200 mM, Sigma, St. Louis, MO was made up in 50 mM Na + HEPES, pH 7.6, readjusted to pH 7.6 with 10 M NaOH, and added at concentrations ranging from 1 to 50 mM.
  • the gastrin concentrations in conditioned media and cell extracts were determined by radioimmunoassay against an amidated gastrin 17 standard curve with 125 I-amidated gastrin 17 as label (22).
  • Levels of amidated gastrin were assayed with antiserum 1296, which detects all amidated carboxyl terminal fragments greater than the pentapeptide, and does not cross-react with glycine-extended forms.
  • Levels of unprocessed, partially processed and mature amidated forms of gastrin i.e. all progastrin-derived peptides
  • were measured with antiserum 8017 which detects the amino terminal portion of
  • Gastrin/CCK-B receptors were measured by the method of Kopin et al (24). 10 4 cells/well were seeded in a 24 well plate and grown for 2 days at 33°C in RPMI medium containing 10 mM thioglycerol, 100 units/ml insulin, 50 mg/ml hydrocortisone and 10% fetal calf serum. Cells were washed in phosphate-buffered saline, and incubated for 80 min at 37°C in Hank's balanced salt solution containing 125 I-CCK 8 (10,000 cpm, 2.9 fmol, Amersham, Bucks., UK), 16 ⁇ M PMSF and 0.1% BSA. Cells were then washed twice with PBS and lysed with NaOH. Lysates were counted in a ⁇ -counter (Packard, Downer's Grove, IL) at 77% efficiency.
  • ⁇ -counter Packard, Downer's Grove, IL
  • Gastrin/CCK-C receptors were measured by the phthalate oil centrifugation method (3) with 125 l-gastrin 17 (1.5 ⁇ 10 5 cpm, 44 fmol (Amersham, Bucks., UK)) as ligand.
  • 125 l-gastrin 17 1.5 ⁇ 10 5 cpm, 44 fmol (Amersham, Bucks., UK)
  • IC 50 values (mean ⁇ SD, calculated from at least 3 sets of data) for inhibition of cross-linking of
  • Antagonists are .
  • the non-selective antagonists proglumide and benzotript both inhibited cross-linking of 125 I-gastrin 2,17 to the GBP, as shown in Figure IB, with IC 50 values of 5.1 mM and 195 ⁇ M respectively (Table 1).
  • IC 50 values 5.1 mM and 195 ⁇ M respectively
  • gastrin and its derivatives a good correlation is observed between the IC 50 values for the 78 kDa GBP and the IC 50 values for inhibition of 125 I-gastrin 17 binding to the gastrin/CCK-C receptor, while the correlation between the IC 50 values for the 78 kDa GBP and the IC 50 values for the gastrin/CCK-B receptor is poor.
  • gastrin 17 (gastrin 1-17 G), which is transamidated by
  • the IC 50 value for glycine-extended gastrin 17 was slightly lower than the IC 50 value for gastrin 17 , as shown in Figure 5 and Table 2. Addition of a further 5 residues to the C-terminus of glycine-extended gastrin 5-17 did not alter the affinity of the resultant peptide for the GBP, while addition of 5 residues to the C-terminus of glycine-extended gastrin 12-17 actually resulted in a 25-fold
  • benzotript which we have shown in Example 1 to be the best available antagonist for the GBP, is an acylated tryptophan derivative (N-4-chlorobenzoyl-L-tryptophan), it seems likely that the tryptophan residue of either the N-terminal or C-terminal tetrapeptide, or both, makes a significant contribution to binding.
  • a cDNA fragment encoding human progastrin (18) was cut from pBR322 with Pstl and cloned into pGEM-3Z
  • pCDNA1amp (Invitrogen, San Diego, CA).
  • the corresponding antisense plasmid was constructed by cloning an XbaI/SphI fragment from pGEM-3Z into pCDNAlamp, and a XbaI/BstXI fragment thence into pCDNA1neo (Invitrogen).
  • Half-confluent YAMC cells were incubated with 20 ⁇ g/ml plasmid DNA for 4-6 hrs at 33°C in serum-free RPMI medium containing 20 ⁇ g/ml lipofectin (Gibco/BRL), 10 mM thioglycerol, 100 units/ml insulin and 50 mg/ml hydrocortisone.
  • Transfected cells were grown at 33°C in RPMI medium containing 10 mM
  • thioglycerol 100 units/ml insulin, 50 mg/ml hydrocortisone and 10% fetal calf serum for 2 days, and selected for 14 days in the same medium containing 100 ⁇ g/ml neomycin.
  • YAMC or LIM 1215 cell extracts and conditioned media were readily detected in YAMC cell extract (20 fmol/10 7 cells) and conditioned medium (66 fmol/10 6 cells) following transfection with a sense gastrin construct; however, amidated gastrin was not detected in these cells.
  • No progastrin-derived peptides were detected in YAMC or LIM 1215 cells transfected with an antisense gastrin construct.
  • COS cells were transiently transfected with a plasmid encoding sense gastrin mRNA or with plasmids encoding both sense and antisense ga ⁇ crin mRNA.
  • the levels of progastrin-derived peptides in cell extracts (760 pmol/10 7 cells) and conditioned media (1540 pmol/10 7 cells) were reduced by 37 and 48% respectively in the doubly transfected cells. Amidated gastrin was not detected in either case. Similar results were obtained in 3 separate experiments with YAMC, and two experiments with LIM 1215.
  • the total amount of 125 I-CCK 8 (10,000 cpm, 2.9 fmol) bound to gastrin/CCK-A + -B receptors on YAMC, LIM 1215 and AR4-2J cells was measured by the method of Kopin et al (24).
  • the AR4-2J cell line was derived from a rat pancreatic carcinoma, and expresses both gastrin/CCK-A + -B receptors.
  • the amount of 125 I-gastrin 17 (150,000 cpm, 44 fmol) bound to gastrin/CCK-C receptors on YAMC and LIM 1215 cells was measured by the phthalate oil centrifugation assay described by Weinstock and Baldwin (3). Background binding was measured in the presence of 1 ⁇ M (A + B) or 10 ⁇ M (C) gastrin 17 .
  • Table 3 The results are summarised in Table 3.
  • AR4-2J 10 4 10567 ⁇ 1341 1866 ⁇ 252
  • Gastrin/CCK-C receptors were readily detected on both YAMC and LIM 1215 cells using the phthalate oil centrifugation assay and 125 I-gastrin 17 as ligand.
  • benzotript 13.7 mM, 113%. Concentrations of benzotript higher than 3 mM were toxic to YAMC cells. No inhibition of proliferation was observed with the CCK-A receptor-selective antagonist L364,718 or the gastrin/CCK-B
  • LIM 2412 cells is inhibited by proglumide, with IC 50 values of 2.2 ⁇ 1.0 mM, and 2.9 ⁇ 1.3 mM respectively. No correlation was observed between the degree of inhibition by proglumide and the presence or absence of binding sites for 125 I-[Nle 15 ]-gastrin 17 determined previously. Even though gastrin 17 breakdown did not exceed 30% during a 24 hour incubation in the absence of serum and a non-saturating concentration of inhibitor was chosen to facilitate ready reversal of inhibition, no reversal of the inhibition of LIM 1215 or LIM 2412 cells by proglumide was detected with gastrin 17 concentrations as high as 50 ⁇ M. These results are shown in Figure 8.
  • NSAIDS have been shown to inhibit colorectal tumour growth in several experimental models, and have been shown to reduce the size and number of colorectal polyps in patients with familial adenomatous polyposis (26), and to reduce the death rate from cancer of the eosophagus, stomach, colon, and rectum (27). It has been widely accepted that the effect of NSAIDS on colon cancers has resulted from the ability of these agents to inhibit cyclooxygenases, which are key enzymes in the synthesis of prostaglandins, and in particular it has been proposed that the anti-tumour activity is correlated with inhibition of cyclooxygenase-2 (28).
  • cyclooxygenase I and cyclooxygenase II were measured using the colorimetric assay described above.
  • the medium was RPMI 1640 containing 10 ⁇ M thioglycerol, 25 units/ml insulin, 1 mg/ml
  • FIG. 9 shows results for indomethacin, sulindac sulphoxide and aspirin. All of these agents inhibited proliferation of LIM 1215 cells. In fact, all of the NSAIDS tested inhibited proliferation of LIM 1215 cells, with IC 50 values obtained by computer curve fitting with the programmes EBDA and LIGAND ranging from 120 ⁇ M for meclofenamic acid and sulindac sulphide to 21 mM for acetaminophen. These results are summarized in Table 4, and are also compared for some agents with IC 50 values for cyclooxygenase I and cyclooxygenase II, which have been reported in the literature.
  • Tolmetin 1900 ⁇ 170 1400 ⁇ 100
  • Figure 10 compares IC 50 values for LIM 1215 cell proliferation and for inhibition of cyclooxygenases I and II in intact bovine aortic endothelial cells and murine macrophages respectively, and shows that there is no correlation between inhibition of cell proliferation and enzyme inhibition for either enzyme.
  • the potent cyclooxygenase inhibitor indomethacin which has an IC 50 value of 30 nM for cyclooxygenase I and 1.7 ⁇ M for cyclooxygenase II, was only able to inhibit LIM 1215 cell proliferation to a moderate extent, having an IC 50 value of 200 ⁇ M.
  • acetaminophen which does not decrease the risk of gastrointestinal tract cancer (27), was even less effective in inhibiting LIM 1215 cell proliferation, having an IC 50 of 21 mM.
  • This agent is more potent than aspirin as an inhibitor of cyclooxygenase II, with an IC 50 value of 130 ⁇ M (28).
  • Our results provide further evidence that the anti-proliferative affects of NSAIDS on colon cancers are not primarily mediated by cyclooxygenase II.
  • gastrin-binding protein cross-linking assay is useful, either alone or in
  • non-selective gastrin/CCK receptor antagonists proglumide and benzotript, and the IC 50 values for inhibition of HCT 116 colon carcinoma cell proliferation by the same
  • L365,260 has any effect on cross-linking of
  • CyclooxygenaseS are involved in prostaglandin synthesis, and utilize fatty acid derivatives as
  • NSAIDS The rat mitochondrial trifunctional protein (MTP), the ⁇ -subunit of which has strong amino acid sequence homology to the porcine GBP, also utilizes fatty acid derivatives as substrates.
  • MTP mitochondrial trifunctional protein
  • NSAIDS with the exception of acetaminophen, inhibit cross-linking of iodinated gastrin to the porcine GBP, and conclude that GBP may be the target of the anti-proliferative effect of these agents on colon cancer.
  • Hereditary defects in the MTP can cause death in early childhood (29). A wide range of diagnostic symptoms has been reported, including diarrhoea, vomiting,
  • hypoketotic hypoglycaemic coma myopathy and
  • conditionally immortalised mouse colon cell line YAMC also expresses cell surface gastrin/CCK-C receptors, but not CCK-A or
  • screening may be carried out using antibody to GBP, labelled with a detectable marker, and measuring the binding of the labelled antibody to dispersed tumour cells.
  • screening may be carried out as described by Weinstock and Baldwin (3).
  • immunochemical assays of inhibition of binding of labelled antibody, or assays of inhibition of binding of labelled high-affinity antagonists provide the opportunity for automated assays which can be used for large scale screening of putative antagonists.
  • Assays of the enzyme activities intrinsic to the GBP may also be used for large-scale screening of GBP antagonists.
  • Covalent cross-linking of 125 I-gastrin 2,17 to the purified 78 kDa GBP provides a convenient assay with which to isolate more potent and selective GBP antagonists capable of blocking cell growth.
  • a colon cancer cell line (LIM 1215) derived from a patient with inherited nonpolyposis colorectal cancer

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biochemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Biotechnology (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Analytical Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides a method of identifying a compound having the ability to block the low-affinity gastrin-cholecystokinin type C receptor, comprising the step of measuring the ability of said compound to block the binding to gastrin-binding protein of a compound selected from the group consisting of a gastrin-related peptide, a cholecystokinin-related peptide, an antagonist of gastrin or of cholecystokinin, an acyl CoA, an enoyl CoA, an antibody to gastrin, and an antibody to cholecystokinin. Several ways in which the method of the invention can be carried out are described. Thus, the invention provides a general screening method for identifying antagonists of the gastrin-binding protein interaction, which are useful for treatment of diseases involving rapidly proliferating cells, and for controlling gastric acid secretion. Compositions and methods of treatment are also claimed.

Description

ASSAY FOR SCREENING OF RECEPTOR ANTAGONISTS
This invention relates to a method for screening of putative antagonists of binding of gastrin to its receptor, in particular the low-affinity gastrin-cholecystokinin C receptor, more particularly the gastrin-binding protein (GBP), and also of inhibitors of the enzyme activities associated with the GBP.
Compounds having the ability to block binding of gastrin to GBP or to block the enzyme activities of the GBP are useful in control of cellular proliferation, especially in treatment of neoplastic disease, and are also useful in the control of acid production in the stomach.
BACKGROUND OF THE INVENTION
The hormone gastrin has been known for over 80 years to be a stimulant of acid secretion by the parietal cells of the stomach. Gastrin is a small polypeptide hormone which participates in a feedback system for regulating the environment in the lumen of the alimentary tract, and is thought to affect other target organs, including the brain and pancreas. More recently, it has been recognised that gastrin also acts as a growth factor for the gastric mucosa. Both of these functions are presumably mediated by the binding of gastrin to specific receptors on the target cells in the gastrointestinal mucosa. A variety of methods for detection of such
receptors in tissue, cell, and membrane preparations from various tissues has been demonstrated.
Gastrin/CCK-C receptors have been identified on cell lines of a variety of tumour cell lines of epithelial and non-epithelial origin, and on several cell lines of lymphocytic origin. In particular, significant levels of gastrin/CCK-C receptors have been observed with cell lines established from carcinomas of the stomach, colon, breast, and vulva, and from malignant melanoma: in addition, lower levels of binding were observed with T- and B-cell lymphoma cell lines, while still smaller levels of binding were observed with promyelocytiσ and myeloid leukaemia cell lines (3).
Three classes of receptors for gastrin and for the related hormone cholecystokinin (CCK) have been described. The CCK-A receptor on the pancreatic acinar cell, and the gastrin/CCK-B receptor in the gastric mucosa and brain, both belong to the family of receptors with 7 transmembrane segments. These two receptors can be readily distinguished with the CCK-A receptor-selective antagonist 1.364, 718, and the gastrin/CCK-B receptor-selective
antagonist 1.365,260. Characteristics of the three types of receptors are summarised in Table 1 below.
Evidence that a gastrin-like peptide acts as an autocrine growth factor in colorectal carcinoma has been accumulating steadily. Gastrin mRNA has been demonstrated in both normal and neoplastic colorectal mucosa by Northern blotting, and in colon carcinoma cell lines by the
polymerase chain reaction. An increase in progastrin production in colorectal tumour tissue compared with normal mucosa has been demonstrated in tumour sections by
immunohistochemistry, and in tissue extracts by
radioimmunoassay. Progastrin, but not mature amidated gastrin, was also produced by all (5/5) colonic carcinoma cell lines tested. Exogenous gastrin17 or its derivatives enhanced the growth of xenografts of 60% of colon
carcinomas tested. However, many colon carcinoma cell lines do not increase either DNA or protein synthesis in vitro in response to exogenous gastrin, perhaps because they are already maximally stimulated by an autocrine gastrin-like peptide. In some cases previously
unresponsive cells have been rendered responsive by
synchronisation.
The most convincing evidence for an autocrine role for a gastrin-like peptide has been provided by studies with gastrin/cholecystokinin (CCK) receptor
agonists and antagonists. The antagonist proglumide, which does not discriminate clearly between the three known classes of gastrin/CCK receptors, increases the survival time of mice bearing tumours derived from the mouse colon carcinoma line MC26 (1). Proglumide also reverses the increase in tumour volume observed in tumour-bearing mice after treatment with pentagastrin. Proliferation of 6 colon carcinoma cell lines in vitro is inhibited by the non-selective antagonists proglumide and benzotript, with IC50 values in the mM range (2). Furthermore, in the case of the HCT 116 cell line proliferation is also inhibited by an anti-gastrin antiserum (2). The high concentrations of gastrin required to reverse inhibition by both antagonists and antibodies suggest that neither pancreatic CCK-A nor gastric gastrin/CCK-B receptors are involved, but are consistent with the involvement of the low affinity
gastrin/CCK-C receptor which we have identified on the surface of several gastric and colonic carcinoma cell lines (2). The failure of the selective antagonists 1.364,718 and L365,260 to inhibit proliferation of HCT 116 cells at concentrations as high as 1 μM confirms that neither CCK-A nor gastrin/CCK-B receptors participate in the autocrine loop (4).
We have identified a gastrin-binding protein (GBP) in detergent extracts of porcine gastric mucosal membranes. The GBP has a molecular weight of 78kDa, and does not appear to have any other disulphide bonded subunits (5). Purification of the porcine GBP by a
combination of lectin and ion exchange chromatography followed by preparative gel electrophoresis has permitted determination of N-terminal and internal amino acid
sequences of the naturall.v occurring protein (Baldwin et al: Int. J. Biochem 26 529-538). These sequences have been used to generate probes which have enabled cloning of the cDNA encoding the porcine GBP from both c-DNA and genomic libraries.
The sequence of porcine GBP, deduced from the cDNA sequence, is related to those of enzymes having enoyl CoA hydratase activity and to enzymes having 3-hydroxy-acyl-CoA dehydrogenase activity. These enzymes are
involved in two consecutive steps in the synthesis and degradation of fatty acids during metabolism located in the α-subunit of a mitochondrial trifunctional protein (MTP). The N-terminal half of the GBP is related to the enoyl-CoA hydratase family, while the C-terminal half is related to the 3-hydroxy-acyl-CoA dehydrogenase family (6). In particular, the sequence of the porcine GBP is closely related to the sequence of the α-subunit of a rat
mitochondrial trifunctional protein (MTP), which catalyses both activities. We have found that the sequence of mature human GBP is 90% identical to that of the porcine GBP
(Zhang and Baldwin, submitted for publication).
To investigate the possibility that the GBP might be involved in the autocrine effects of gastrin-like peptides on colorectal carcinoma cells, we have now
examined the effect of agonists and antagonists on the interaction between the GBP and gastrin. Since colon carcinoma cell lines synthesise progastrin, but fail to process the prohormone to mature C-terminally amidated gastrin17, a candidate receptor for autocrine gastrin should not require an amidated C-terminus for binding. We have therefore also examined the structural requirements for binding of C-terminally extended gastrins to the GBP.
We have now found that covalent cross-linking of 125I-[Nle15]-gastrin2,17 to the 78 kDa GBP is inhibited by crotonyl CoA and by acetoacetyl CoA. The finding that enoyl CoA and acyl CoA derivatives inhibit binding of gastrin to GBP is surprising. According to the autocrine hypothesis, the reverse effect would be expected. Gastrin, CCK, and their analogues also inhibit cross-linking, and the spectrum of analogue affinities correlates well with the values previously reported for binding to the
gastrin/CCK-C receptor, but poorly with the values reported for binding to either the CCK-A or gastrin/CCK-B receptors. The good correlation observed between the IC50 values for antagonists in the GBP cross-linking assay and the IC50 values for inhibition of either binding of gastrin to colon carcinoma cells or of cell proliferation suggests that the GBP is the target for the inhibitory effects of gastrin/CCK receptor antagonists on cell growth. Cross-linking is also inhibited by proglumide and benzotript, but no inhibition is seen with either the CCK-A receptor-selective antagonist L364,718 or the gastrin/CCK-B receptor-selective antagonist L365,260. The affinities of antagonists for the GBP correlate well with their affinities for the gastrin/CCK-C receptor, and with their potencies for inhibition of colon carcinoma cell growth. In addition to gastrin/CCK receptor antagonists, we have found that other classes of compounds inhibit the cross-linking. For example, non-steroidal anti-inflammatory drugs inhibit not only the cross-linking, but also proliferation of a human colon carcinoma cell line.
To investigate further the postulated autocrine loop for gastrin we have transfected two colon cell lines with a plasmid encoding a gastrin cDNA in the antisense orientation. We have observed inhibition of cell
proliferation, providing clear evidence for the presence of an autocrine loop. Moreover the failure to reverse
inhibition with exogenous gastrin17 suggests that the loop in these cells is intracellular.
We conclude that the 78 kDa gastrin-binding protein is
(i) a member of the hydratase/dehydrogenase
family of fatty acid oxidation enzymes; (ii) the gastrin/CCK-C receptor;
(iii) the target for the anti-proliferative
action of gastrin/CCK receptor antagonists and compounds such as non-steroidal anti- inflammatory drugs;
(iv) a potential target for the autocrine
effects of progastrin; and (v) a target for anti-sense gastrin RNA regulation of proliferation of colon carcinoma cells.
SUMMARY OF THE INVENTION
In one aspect, the present invention provides a method of identifying a compound having the ability to block the low-affinity gastrin-cholecystokinin type C receptor, comprising the step of measuring the ability of said compound to block the binding to gastrin-binding protein of a compound selected from the group consisting of a gastrin-related peptide, a cholecystokinin-related peptide, an antagonist of gastrin or of cholecystokinin, an acyl CoA, an enoyl CoA, an antibody to gastrin, and an antibody to cholecystokinin.
The binding may be reversible or irreversible, but is preferably irreversible.
In a preferred embodiment, inhibition of irreversible binding is assessed by measuring the ability of the compound to block cross-linking of a gastrin
analogue to gastrin binding protein.
Cross-linking may be effected by an agent which is homobifunctional, heterobifunctional, or photoreactive. The person skilled in the art will be aware of a number of suitable reagents. Examples of commercially-available homobifunctional cross-linking reagents include
disuccinimidyl suberate, disulfosuccinimidyl suberate, and ethylene glycol bis-(succinimidyl)-succinate. Heterobi-functional cross-linking reagents include succinimidyl-4-(p-maleimidophenyl) butyrate, which has been found to be equally effective in cross-linking the gastrin peptide to the gastrin binding protein. Suitable photoreactive reagents include N-hydroxysuccinimidyl-4-azidobenzoate.
The gastrin peptide is labelled with a detectable marker, such as a radioactive label, a fluorescent label, or biotin. For example, the gastrin peptide may be
iodinated, or may be synthesised using one or more amino acids comprising a radioactive marker such as 14C or 3H
Native gastrin, gastrin17, contains no free amino groups, and is therefore incapable of reacting with the cross-linking reagent. The minimum gastrin sequence required for binding to the gastrin/CCK-C receptor is known to be the C-terminal tetrapeptide, so N-truncated peptides are expected to be suitable. For example, [Leu15] gastrin17 and [Nle15] gastrin2,17 have equal ability to bind to isolated canine parietal cells or parietal cell plasma membranes: substitution of methionine by leucine isomers does not interfere with the binding or biological activity of gastrin. A particularly preferred gastrin peptide is [Nle15] gastrin2,17, a derivative which lacks the amino-terminal pyroglutamate residue. Substitution of nor-leucine for the naturally occurring methionine at position 15 prevents formation of oxidised methionine derivatives during iodination. Although [Met15]-gastrin2,17 could be used, there would be some loss of activity for this reason. It is contemplated that gastrin peptides extended at the C-terminus could also be used.
In another preferred embodiment, a displacement-type assay using reversible binding is employed. The ability of the putative antagonist to prevent binding of a known agonist or antagonist of very high affinity is measured, said known agonist or antagonist being labelled with a detectable marker. Preferably the known agonist or antagonist has at least the binding affinity of benzotript. The known antagonist is suitably labelled with 3H.
The method of the invention provides a general screening method for identifying antagonists of the
gastrin-gastrin binding protein interaction, which
compounds are useful for treatment of diseases, especially neoplastic diseases, involving rapidly proliferating cells. Such antagonists are also useful for controlling gastric acid secretion.
One preferred group of compounds useful for prevention and treatment of neoplastic diseases is non- steroidal anti-inflammatory drugs (NSAIDS), which are especially useful for treatment of neoplastic diseases of the gastrointestinal tract, in particular colorectal cancer. However, we have shown that GBP is widespread in the body, and consequently treatment of cancers of non-gastrointestinal origin is also within the scope of this invention. It is preferable, but not essential, that the NSAID is an inhibitor of prostaglandin activity.
In another preferred embodiment, inhibition of the enoyl CoA hydratase and/or 3-hydroxyacyl CoA
dehydrogenase activities intrinsic to the GBP is assayed. Either activity can be measured in either direction, but is conventionally assayed spectrophotometrically by the decrease in enoyl CoA absorption on hydration or by the decrease in NADH absorption on oxidation. Alternative assays could employ radiochemically labelled substrates.
Gastrin-binding protein suitable for use in the method of the invention may be partially purified or purified naturally occurring GBP, or may be recombinant GBP.
As an alternative to using a high-affinity labelled antagonist in a reversible binding assay, a high-affinity antibody, such as a monoclonal antibody, labelled with a detectable marker may be used. For example, it is contemplated that an antibody capable of blocking binding of an acyl CoA or of gastrin to GBP may be used.
The invention in another aspect therefore provides a method of treating neoplastic disease,
comprising the step of administering to a mammal in need of such treatment an effective amount of a compound having the ability to inhibit binding of a gastrin peptide to gastrin-binding protein.
Preferably the neoplastic disease is selected from the group consisting of gastrointestinal cancers such as colon carcinoma and gastric carcinoma, mammary
carcinoma, malignant melanoma, tumours of epithelial origin, and lymphomas. In yet another a act, the invention provides a pharmaceutical composition of a compound with the ability to inhibit the binding of a gastrin analogue to the low-affinity gastrin-cholecystokinin type C receptor,
comprising a pharmaσeutically-effective amount of said compound and a pharmaceutically-acceptable carrier.
In one preferred embodiment the compound is a non-selective antagonist of the gastrin-cholecystokinin receptor. Preferably the antagonist will have a IC50 value" in the millimolar range or lower.
The compositions of the invention may comprise an antagonist either alone, or in combination with other pharmaceutically-active molecules such as antacid compounds or anti-cancer agents. Anti-cancer agents may include cytokines. Methods and pharmaceutical carriers for
preparation of pharmaceutical compositions are well known in the art, as set out in text books such as Remington's Pharmaceutical Sciences, 17th Edition, Mack Publishing Company, Easton, Pennsylvania, USA.
In a fourth aspect, the invention provides a method of treatment of a neoplastic disease, comprising the step of inducing production of antisense gastrin mRNA in neoplastic tissue of an animal in need of such treatment. Tissue-specific production of anti-sense mRNA can be achieved by targeting of corresponding DNA to the desired tissue, using tissue-specific promoters.
DETAILED DESCRIPTION OF THE INVENTION
The invention is illustrated by reference to the following non-limiting examples, and to the figures, in which:
Figure 1 shows gastrin analogues and antagonists inhibit covalent cross-linking of 125 I-[Nle15]-gastrin2,17 to the 78kDa GBP.
Cross-linking of 125I- [Nle15]-gastrin2,17 to the 78 kDa GBP in the presence of increasing concentrations of (A) gastrin analogues (closed symbols) and (B) gastrin/CCK receptor antagonists (open symbols) was measured by phosphorimager (insets), and expressed as a percentage of the value obtained in the absence of competitor. The following values for IC50 and for the predicted ordinate intercept were obtained with the program LIGAND: gastrin17 (♦), 0.11 μM, 102%; CCK8 (■), 5.6 μM, 98%; CCKeSO4 (•), 3.55M, 125%; pentagastrin (▲), 200 μM, 96%; gastrin4 (▼), 320 μM, 98%; proglumide (□), 4.1 mM, 94%; benzotript (◯), 250 μM, 114%. Lines of best fit are not shown for CCK8 or gastrin4 for clarity. No inhibition was observed with the CCK-A receptor-selective antagonist L364,718 (data not shown), or with the gastrin/CCK-B receptor-selective antagonist L365,260 (◊).
Figure 2 shows binding of fatty acyl CoA derivatives to the 78 kDa GBP.
Cross-linking of 125I-[Nle15]-gastrin2,17 to the 78 kDa GBP was measured by phosphorimager (insets) in the presence of increasing concentrations of crotonyl CoA, a substrate of enoyl CoA hydratase, or of acetoacetyl CoA, a product of 3-hydroxyacyl CoA dehydrogenase, and expressed as a percentage of the value obtained in the absence of competitor. Lines of best fit for inhibition by
acetoacetyl CoA and crotonyl CoA were obtained with the program LIGAND. Values for IC50 and for the predicted ordinate intercept were as follows: Crotonyl CoA (•), 43 μM, 76%; acetoacetyl CoA (■), 74 μM, 70%. No inhibition was observed with NADH (▲), a cofactor for 3-hydroxyacyl CoA dehydrogenase.
Figure 3 shows that anti-proliferative gastrin/CCK receptor antagonists target the 78 kDa GBP. The mean IC50 values (Table 1) for the inhibition of cross-linking of 125I-[Nle15]-gastrin2,17 to the 78 kDa GBP by gastrin analogues (closed symbols as in Figure 2) and antagonists (open symbols as in Figure 2) were determined as described in the legend to Figure 2, and compared with the IC50 values for (A) inhibition of binding of 125I-CCK8 labelled using Bolton-Hunter reagent to the cloned human gastrin/CCK-B receptor expressed in COS cells (analogues) (8) and of 125I-CCK33 to the gastrin/CCK-B receptor on guinea pig gastric glands (antagonists) (9), or (B) inhibition of binding of 125I-gastrin17 to the gastrin/CCK-C receptor on HCT 116 colon carcinoma cells (2). Lines of best fit were obtained by linear regression.
Figure 4 illustrates processing and sequences of gastrin-related peptides.
The C-terminal sequence of human progastrin is shown using the one letter code. The corresponding region of porcine progastrin is only shown where it differs from human progastrin. The indicated peptides (numbered from the N-terminus of gastrin1-17) were synthesised according to the human sequence, with the N-terminal glutamic acid of gastrin1-4, gastrin1-11, gastrin1-17 and gastrin1-17G cyclized to pyroglutamate. The C-termini of gastrin1-17 and
gastrin14-17 were amidated.
Figure 5 shows binding of gastrin-related peptides to the 78 kDa GBP.
Cross-linking of gastrin-related peptides was measured as described above, and the cross-linking
quantitated by phosphorimager, as shown in the inset panels, and lines of best fit calculated and plotted against peptide concentration.
Figure 6 illustrates how expression of antisense gastrin inhibits cell proliferation.
Proliferation of YAMC (A) or LIM 1215 (B) cells transfected with plasmid alone in the absence (black bars) or presence (light grey bars) of 1 μM gastrin17, or with a plasmid expressing antisense gastrin mRNA in the absence
(dark grey bars) or presence (white bars) of 1 μM gastrin17 was measured with the MTT assay at the indicated times after seeding in media containing 1% fetal calf serum. The means of triplicate absorbance readings were expressed as a percentage of the mean absorbance reading obtained on day 1 for the YAMC cells grown in the absence of gastrin17; error bars represent the standard error of the mean. Statistical significance was assessed by Student's t-test: p < 0.05, *; p < 0.02, **.
Figure 7 shows the inhibition of YAMC cell proliferation by gastrin/CCK receptor antagonists.
Proliferation of YAMC cells in the presence of increasing concentrations of the non-selective gastrin/CCK receptor antagonists proglumide (●) or benzotript (■) was measured with the MTT assay at two days after seeding.
Antagonists were added one day after seeding. The means of triplicate absorbance readings are shown as a percentage of the mean absorbance reading obtained on day 1 for the cells grown in 1% fetal calf serum; error bars represent the standard error of the mean. Lines of best fit were
obtained with the programs EBDA and LIGAND as described previously (6).
Figure 8 shows the inhibition of growth of
LIM 1215 and LIM 2412 cells by proglumide.
Uptake of 3H-thymidine (○) by, and viability (□) of, cells was measured after 17 hours treatment with the concentration of proglumide indicated. LIM 1215 (A) and LIM 2412 (B) cells were grown in the absence of serum.
Points are the mean values of 5 samples, and lines of best fit were obtained with the program LIGAND. Bars represent 1 SD.
Figure 9 illustrates proliferation of LIM 1215 colon carcinoma cells in the presence of increasing
concentrations of NSAIDS, measured with the MTT assay as described for Figure 7. Cells were grown in a medium containing 10% foetal calf serum, and NSAIDS were added one day after seeding. The means of triplicate absorbance readings were expressed as a percentage of the mean
absorbance reading obtained for cells grown in the absence of NSAIDS; error bars represent the standard error of the mean. Lines of best fit were obtained with the programs EBDA and LIGAND as described previously (6). Values for IC50 and for the predicted ordinate intercept were as follows: indomethacin, 91 μM, 137% (t); sulindac sulphoxide, 358 μM, 132% (■); aspirin, 3.22 mM, 134% (▲). Similar results were obtained in 3 separate experiments.
Figure 10 shows that the inhibition of proliferation of LIM 1215 cells by NSAIDS is not correlated with the ability of the NSAIDS to inhibit either
cyclooxygenase I (Panel A) or cyclooxygenase II (Panel B). The IC50 values for inhibition of proliferation of LIM 1215 cells were measured as described for Figure 8, and are summarized in Table 4. IC50 values for inhibition of cyclooxygenase I and cyclooxygenase II were taken from
Mitchell et al (Proc. Natl. Acad. Sci. USA., 1994 90. 11693-11697).
Figure 11 shows the ability of four different NSAIDS to block cross-linking of gastrin2,17 to GBP in the assay described above.
Figure 12 shows the correlation between the ability of NSAIDS to inhibit proliferation of LIM 1215 cells and their ability to inhibit cross-linking of gastrin2/17 to GBP.
Abbreviations used herein are as follows:
ASP - aspirin
CCK - cholecystokinin
CHO - Chinese hamster ovary
DIC - diclofenac
DIF - diflunisal
FCS - foetal calf serum
GBP - gastrin-binding protein
IBU - ibuprofen
IC50 - concentration required for 50%
inhibition of cross-linking
IFN - niflumic acid
IND - indomethacin
MEC - meclofenamic acid
MEF - mefenamic acid
MTT - 3-(4,5-dimethylthiazol-2-yl)-2-,5- diphenyltetrazolium bromide
NAP - naproxen PHE - phenylbutazone
PIR - piroxicam
SAL - salicylic acid
SUL - sulindac sulphoxide
SUS - sulindac sulphide
TIA - tiaprofenic acid
TOL - tolmetin
YAMC - young adult mouse colon.
MATERIALS AND METHODS
Gastrin17 , CCK8, and CCK8SO4 were from Research
Plus, Bayonne, NJ. Pentagastrin, gastrin4, proglumide (4-(benzoylamino)-5-(dipropylamino)-5-oxopentanoic acid, acetoacetyl CoA, crotonyl CoA, pepstatin, benzamidine, hexamethylphosphoramide, aprotinin and NSAIDS were from Sigma, St.Louis, MO. Benzotript (N-(p-chlorobenzoyl)-L-tryptophan) was from Calbiochem, San Diego, CA. The antagonists L364,718 and L365,260 were generous gifts from Dr. V.J.Lotti, Merck, Sharp and Dohme, West Point, PA.
Na125I was from NEN, North Ryde, Australia.
1251- [Nle15]-gastrin2,17 was prepared and iodinated as described in reference 5, the whole contents of which publication are herein incorporated by reference.
The 78 kDa GBP was partially purified from detergent extracts of porcine gastric mucosal membranes (5), and crosslinked to 125I-[Nle]15-gastrin2,17 with
disuccinimidylsuberate as previously described (5). The following protease inhibitors were included in all buffers to prevent proteolysis: pepstatin, 1 μM; benzamidine, 1 mM; hexamethylphosphoramide, 0.1% (w/v); aprotinin, 500 units/ml. The products of the cross-linking reaction were separated by polyacrylamide gel electrophoresis, and radioactivity associated with the 78 kDa GBP was detected and quantitated with a phosphorimager (Molecular Dynamics, Sunnyvale, CA). Initial estimates of IC50 values, and of the levels of 125I-gastrin2,17 in the absence of competitor, were obtained by fitting the data with the program EBDA, and were refined with the program LIGAND (3).
The purity and composition of gastrin-related peptides was assessed by the manufacturer (Chiron
Mimotopes, Clayton, Australia) by reversed phase high pressure liquid chromatography and by ion spray mass spectrometry.
Human gastrin cDNA in the plasmid pBR322 (18) was kindly provided by Dr. E. Boel, University of Aarhus, Denmark. The conditionally immortalised young adult mouse colon cell line (YAMC), which was derived from a mouse transgenic for a temperature sensitive SV40 large T antigen (19), and the human colon carcinoma cell line LIM 1215, which was derived from a patient with inherited
nonpolyposis colorectal cancer (20), were obtained from Dr. R.H. Whitehead, Ludwig Institute for Cancer Research, Melbourne.
Cell Proliferation Assay.
A colorimetric assay (21) was used to measure cell proliferation. Briefly, 104 cells were seeded in a 96 well plate in medium containing 1% fetal calf serum. The medium was replaced the next day with fresh medium
containing 1% fetal calf serum and the substance under investigation. For each time point 10 μl of 5 mg/ml MTT
(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, Sigma, St.Louis, MO) was added to each well, and the plate was incubated for 4 hrs at 37°C before the medium was discarded. 200 μl 0.04M HCl in isopropanol was added to lyse the cells, and the absorbance at 560 nm was read on a Titertek Multiscan MCC 1340 (Labsystems, Helsinki,
Finland). Student's t-test was used for statistical analysis.
In one set of experiments with the human colon carcinoma cell lines LIM 1215 and LIM 2412, cell
proliferation was measured using a 3H-thymidine uptake assay, using a standard method. LIM 2412 cells were grown in RPM1 1640 medium, and LIM 1215 cells in the same medium containing 1 mM thioglycerol, 25 units/ml insulin, 1 mg/ml hydrocortisone, 5 mg/ml bovine serum albumin, 10 μg/ml iron-saturated transferrin and 1.5 mg/ml glutamine. Both cell lines were cultured in the absence of serum.
Proglumide (200 mM, Sigma, St. Louis, MO) was made up in 50 mM Na+ HEPES, pH 7.6, readjusted to pH 7.6 with 10 M NaOH, and added at concentrations ranging from 1 to 50 mM.
Control wells received the same volume of 50 mM Na+ HEPES, pH 7.6, as this buffer was sometimes found to inhibit [3H]-thymidine uptake. The cells were then incubated in a total volume of 100 μl at 37°C in a humidified atmosphere of 10% CO2. After 17 hours the cells were pulsed with [3H]-thymidine (0.5 μCi per ml, NEN-DuPont, Boston, MA) for four hours. The medium was removed, and the cells were lysed with a mixture of 30 μl lysis buffer (30 mM Tris HCl, pH 7.6, 100 mM EDTA, 1% w/v Sarcosyl (Sigma, St. Louis, MO)) and 70 μl PBS, harvested and counted using a Beta-Plate harvester and liquid scintillation counter (LKB-Wallace, Turku, Finland). Cell viability in a duplicate plate was measured by Trypan Blue exclusion.
Radioimmunoassay
The gastrin concentrations in conditioned media and cell extracts were determined by radioimmunoassay against an amidated gastrin17 standard curve with 125I-amidated gastrin17 as label (22). The calculated 50% inhibitory dose for antiserum 8017 was 1.6 ± 0.1 fmol/tube (n=13). The intraassay variation was <6% (n=6) and the interassay variation was less than 20% (n=13). Levels of amidated gastrin were assayed with antiserum 1296, which detects all amidated carboxyl terminal fragments greater than the pentapeptide, and does not cross-react with glycine-extended forms. Levels of unprocessed, partially processed and mature amidated forms of gastrin (i.e. all progastrin-derived peptides) were measured with antiserum 8017, which detects the amino terminal portion of
gastrin17, after digestion of samples with trypsin (23). Samples were treated with an equal volume of 50 μg/ml trypsin in 20 mM veronal, pH 8.2, containing 0.005% sodium azide and 2% charcoal-stripped plasma. After incubation at 37°C for 2 hr, the samples were boiled for 2 min, cooled on ice for 2 min, and frozen until assay.
Gastrin Binding Assays
Gastrin/CCK-B receptors were measured by the method of Kopin et al (24). 104 cells/well were seeded in a 24 well plate and grown for 2 days at 33°C in RPMI medium containing 10 mM thioglycerol, 100 units/ml insulin, 50 mg/ml hydrocortisone and 10% fetal calf serum. Cells were washed in phosphate-buffered saline, and incubated for 80 min at 37°C in Hank's balanced salt solution containing 125I-CCK8 (10,000 cpm, 2.9 fmol, Amersham, Bucks., UK), 16 μM PMSF and 0.1% BSA. Cells were then washed twice with PBS and lysed with NaOH. Lysates were counted in a γ-counter (Packard, Downer's Grove, IL) at 77% efficiency.
Gastrin/CCK-C receptors were measured by the phthalate oil centrifugation method (3) with 125l-gastrin17 (1.5 × 105 cpm, 44 fmol (Amersham, Bucks., UK)) as ligand. Example 1 Definition of the Gastrin Binding Site
The ability of gastrin peptides and of non-specific and specific CCK receptor antagonists to inhibit cross-linking of GBP to a labelled gastrin peptide was tested as described above. The results, expressed as IC50 values, are set out in the far right hand column of Table 1, and are compared with IC50 values previously reported in the literature for binding of gastrin agonists or
antagonists to tissues or tumour cells, or for inhibition of proliferation of tumour cell lines. Values are for the human receptors unless indicated as follows: a guinea pig; b rat. ND, not determined. Reference numbers are given in brackets.
IC50 values (mean ± SD, calculated from at least 3 sets of data) for inhibition of cross-linking of
125I-[Nle15]-gastrin2,17 to the 78 kDa GBP in detergent extracts of porcine gastric mucosal membranes were
determined from the data presented in Figure 1 and from similar experiments by least squares fitting with the programs EBDA and LIGAND.
Table 1
Comparison of the 78 kDa GBP with Human Gastrin and Cholecystokinin Receptors.
IC50 for Binding IC50 for Cell Proliferation IC50 for Cross
(μM) (μM) linking (μM)
Receptor CCK-A Gastrin/CCK-B Gastrin/CCK- 78 kDa GBP
Tissue Pancreas, Stomach, C HCT 116 Cells HT 29 Cells Stomach
Gall Bladder Brain HCT 116
Cells
Agonists
Gastrin17 0.08 6.4 × 10-3 1.6 - - 0.23 ± 0.15
1.8 9.4 × 10-4
2.6 × 10-3
CCk8 0.14a 1.9 × 10-3 13.2 - - 4.5 ± 1.0
4.7 × 10-3
I
CCK8SO4 2 × 10-5 3 × 10-3 ND - - 5.8 ± 3.2
3 × 10-3 1.4 × 10-4
1.1 × 10-4
Pentagastrin ND ND 100 - - 210 ± 90
Gastrin4 ND 3.2 × 10-2 ND - - 370 ± 280
3.0 × 10-2
Antagonists
Proglumide 600b 900a 8600 5000 14000 5100 ± 3600
Benzotript 102b 59a 400 500 3400 200 ± 120
L364,718 1 × 10-4 0.5 ND 4 >100 >200
8 × 10-4 0.15
L365,260 9 1.0 × 10-2 ND 15 22 >200
0.02 3.8 × 10-3
Data in first 5 columns summarised from reports in the literature.
Gastrin17 and its derivatives pentagastrin and gastrin4 all inhibited cross-linking of 125I-[Nle15]-gastrin2,17 to the 78 kDa GBP, as illustrated in Figure IA. The observed IC50 values, which are dependent on chain length, are in good agreement with the IC50 values reported previously for inhibition of binding of 125I-gastrin17 to the gastrin/CCK-C receptor on the HCT 116 colon carcinoma cell line (see Table 1). In contrast, the IC50 values for the 78 kDa GBP differ markedly from the IC50 values for the CCK-A and gastrin/CCK-B receptors shown in Table 1. The correlation between the IC50 values for the 78 kDa GBP and the IC50 values for the gastrin/CCK-C receptor, and the lack of correlation between the IC50 values for the 78 kDa GBP and the IC50 values for the gastrin/CCK-B receptor, is particularly apparent when the data are presented
graphically (Figures 3A and 3B).
Example 2 Inhibition by Gastrin/CCK Receptor
Antagonists .
The non-selective antagonists proglumide and benzotript both inhibited cross-linking of 125I-gastrin2,17 to the GBP, as shown in Figure IB, with IC50 values of 5.1 mM and 195 μM respectively (Table 1). As with gastrin and its derivatives, a good correlation is observed between the IC50 values for the 78 kDa GBP and the IC50 values for inhibition of 125I-gastrin17 binding to the gastrin/CCK-C receptor, while the correlation between the IC50 values for the 78 kDa GBP and the IC50 values for the gastrin/CCK-B receptor is poor. These results are summarised in Figure 3. Furthermore, neither the CCK-A receptor antagonist L364,718 nor the gastrin/CCK-B receptor antagonist L365,260 had any effect on cross-linking at concentrations as high as 200 μM (Figure 1B). A good correlation was also observed between the IC50 values for the 78 kDa GBP and the IC50 values for inhibition of HCT 116 and HT 29 colon carcinoma cell proliferation by proglumide and benzotript (Table 1). Example 3 Inhibition by Fatty Acid Derivatives.
A substrate for enoyl CoA hydratase, crotonyl CoA, and a product of 3-hydroxyacyl CoA dehydrogenase, acetoacetyl CoA, both inhibited cross-linking of
125I-gastrin2,17 to the GBP, as summarised in Figure 2. The mean IC50 values from 3 separate experiments were 45 ± 26 μM for crotonyl CoA and 180 ± 150 μM for acetoacetyl CoA. No inhibition was observed with NADH, the cofactor of the dehydrogenase reaction, at concentrations as high as
1.5 mM.
Example 4 Amidation of Gastrin17 is not Required for
GBP Binding
The sequences of gastrin related peptide and glycine-extended peptides are shown in Figure 4.
Processing of progastrin to form gastrin begins by cleavage by a dibasic-specific endopeptidase at the sites marked by arrowheads. Removal of the basic amino acids by carboxypeptidase E yields glycine-extended
gastrin17 (gastrin1-17G), which is transamidated by
peptidyl-glycine α-amidating monooxygenase to form mature amidated gastrin1-17.
The effect of extending the C-terminus of
gastrin17 on binding to the GBP was investigated by
comparing the potencies of glycine-extended gastrin17 and gastrin17 as inhibitors of cross-linking of [125I]-Nle15-gastrin2-17 to the GBP.
IC50 values (mean ± SEM of at least 3 separate determinations) for the inhibition of cross-linking of
[125I] -Nle15-gastrin2-17 to the 78 kDa gastrin-binding protein by the indicated peptides were determined as described above, and are summarised in Table 2. IC50 values for gastrin12-17GRRSAE, gastrin1-4 and YFGRRSAEEGD represent estimates only, because the limiting amounts of peptide available precluded assay at concentrations greater than 100, 400 and 100 μM respectively. IC50 values for the gastrin/CCK-B receptor on canine parietal cells, and for the binding of gastrin1-17 and gastrin14-17 to the GBP, are taken from reference 24 and Example 1 respectively. The amino acid sequences of C-terminal extensions are shown in the one letter code; the peptide sequences are shown in full in Figure 4.
Table 2
Affinities of Gastrin-Related Peptides
for Gastrin Receptors
IC50 (μM)
Peptide Purity 78 kDa Gastrin- Gastrin/CCK-B *
(%) Binding Protein Receptor
Gastrin1-17 82 0.23 ± 0.15 5 × 10-4
Gastrin1-17G 78 0.19 ± 0.10 ND
Gastrin5-17G 93 26 ± 15 ND
Gastrin5-17GRRSAE 87 20 ± 11 ND
Gastrin12-17G 81 31 ± 17 14 ± 71
Gastrin12-17GRR ND ND 44 ± 21
Gastrin12-17GRRSAE 91 760 ± 550 ND
Gastrln14-17 95 370 ± 280 0.1
Gastrin1-11 97 21 + 9 ND
Gastrin1-4 98 >1000 ND
YFGRRSAEEGD 62 105 ± 21 ND
ND : not determined * Values taken from Reference 11
The IC50 value for glycine-extended gastrin17 was slightly lower than the IC50 value for gastrin17, as shown in Figure 5 and Table 2. Addition of a further 5 residues to the C-terminus of glycine-extended gastrin5-17 did not alter the affinity of the resultant peptide for the GBP, while addition of 5 residues to the C-terminus of glycine-extended gastrin12-17 actually resulted in a 25-fold
reduction in affinity. Example 5 Gastrin17 Binding Determinants
The regions of gastrin17 contributing to GBP binding were investigated by testing the effect of N- and C-terminal deletion on the potency of glycine-extended gastrin17 as an inhibitor of cross-linking of [125I]-Nle15-gastrin2-17 to the GBP. Results are shown in Figure 5.
Cross-linking of 125I-[Nle15]-gastrin2-17 to the 78 kDa GBP was measured as described above in the presence of increasing concentrations of gastrin-related peptides .
Duplicate samples were subjected to electrophoresis on NaDodS04-polyacrylamide gels, and the radioactivity
associated with the 78 kDa GBP was quantitated by
phosphorimager scanning and expressed as a percentage of the value obtained in the absence of competitor. The following values for IC50 and for the predicted ordinate intercept were obtained by computer fitting as described herein, and used to construct the indicated lines of best fit: (A) gastrin1-17 (138 nM, 115.5%) or glycine-extended gastrin1-17 (98 nM, 90.0%), (B) glycine-extended gastrin5-17 (11 μM, 93.8%) or glycine-extended gastrin12-17 (40 μM,
99.6%), or (C) gastrin1-4 (1.96 mM, 101.7%) or gastrin1-11 (31 μM, 101.3%).
Comparison of the affinities of gastrin analogues for the 78 kDa GBP revealed that both ends of gastrin17 contribute to binding. Thus removal of 4 residues
(pyroglutamylGPW) from the N-terminus of glycine-extended gastrin17 resulted in a 140-fold decrease in affinity, while removal of 7 residues (YGWMDFG) from the C-terminus of glycine-extended gastrin17 resulted in a 110-fold decrease in affinity (Table 2). Removal of a further 7 residues from the C-terminus of glycine-extended gastrin17 resulted in almost complete loss of binding affinity. In reciprocal experiments the binding of the deleted peptides to the GBP was also measured. As expected, gastrin14-17 bound weakly to the GBP, but binding of the N-terminal tetrapeptide pyroglutamylGPW was barely detectable. Since benzotript, which we have shown in Example 1 to be the best available antagonist for the GBP, is an acylated tryptophan derivative (N-4-chlorobenzoyl-L-tryptophan), it seems likely that the tryptophan residue of either the N-terminal or C-terminal tetrapeptide, or both, makes a significant contribution to binding.
The role of the sequence of five glutamic acid residues of gastrin17 in GBP binding is not clear at present. The similar affinities of glycine-extended gastrin5-17 and glycine-extended gastrin12-17 for the GBP (Table 2) suggest that this polyglutamate sequence makes no contribution. However, deletion of the five glutamate residues from gastrin1-11 to yield gastrin1-4 resulted in a 60-fold decrease in affinity.
In contrast to the present results with the GBP, two lines of evidence demonstrate that the gastrin/CCK-B receptor does not recognise the N-terminus of gastrin17. Firstly, no binding of the N-terminal tridecapeptide to the gastrin/CCK-B receptor on canine parietal cells was
observed even at concentrations as high as 100 μM (11). Secondly, removal of 4 residues from the N-terminus of gastrin17 had no effect on binding to rabbit parietal cells (12). The observation that removal of a further 5
residues, including 4 of the 5 glutamic acid residues, reduced binding to rabbit parietal cells by 100-fold additionally implied that the pentaglutamic acid sequence contributed to gastrin/CCK-B receptor binding (16). The gastrin binding sites of the GBP and the gastrin/CCK-B receptor are thus clearly distinct.
Example 6 Anti-Sense Progastrin RNA Inhibits
Expression of Progastrin and Gastrin
A cDNA fragment encoding human progastrin (18) was cut from pBR322 with Pstl and cloned into pGEM-3Z
(Promega, Madison, WI). An expression plasmid encoding gastrin mRNA in the sense orientation was then constructed by cloning a Bam HI/SphI fragment from pGEM-3Z into
pCDNA1amp (Invitrogen, San Diego, CA). The corresponding antisense plasmid was constructed by cloning an XbaI/SphI fragment from pGEM-3Z into pCDNAlamp, and a XbaI/BstXI fragment thence into pCDNA1neo (Invitrogen).
For transient transfections 107 COS cells were incubated with 2.5 μg/ml plasmid DNA, 100 μM chloroquine and 200 μg/ml DEAE-dextran in RPMI medium containing 2 mM glutamine and 10% heat-inactivated NuSerum (Gibco/BRL, Gaithersburg, MD) for 4 hr. at 37°C, grown for 3 days at 37°C in RPMI medium containing 2 mM glutamine and 10% fetal calf serum, and harvested. The lipofection method (25) was used to generate stable cell lines. Half-confluent YAMC cells were incubated with 20 μg/ml plasmid DNA for 4-6 hrs at 33°C in serum-free RPMI medium containing 20 μg/ml lipofectin (Gibco/BRL), 10 mM thioglycerol, 100 units/ml insulin and 50 mg/ml hydrocortisone. Transfected cells were grown at 33°C in RPMI medium containing 10 mM
thioglycerol, 100 units/ml insulin, 50 mg/ml hydrocortisone and 10% fetal calf serum for 2 days, and selected for 14 days in the same medium containing 100 μg/ml neomycin.
No progastrin or amidated gastrin was detected in
YAMC or LIM 1215 cell extracts and conditioned media. In contrast, progastrin-derived peptides were readily detected in YAMC cell extract (20 fmol/107 cells) and conditioned medium (66 fmol/106 cells) following transfection with a sense gastrin construct; however, amidated gastrin was not detected in these cells. No progastrin-derived peptides were detected in YAMC or LIM 1215 cells transfected with an antisense gastrin construct.
To confirm that expression of antisense gastrin mRNA was able to reduce levels of progastrin-derived peptides, COS cells were transiently transfected with a plasmid encoding sense gastrin mRNA or with plasmids encoding both sense and antisense gaε crin mRNA. The levels of progastrin-derived peptides in cell extracts (760 pmol/107 cells) and conditioned media (1540 pmol/107 cells) were reduced by 37 and 48% respectively in the doubly transfected cells. Amidated gastrin was not detected in either case. Similar results were obtained in 3 separate experiments with YAMC, and two experiments with LIM 1215.
We have previously demonstrated the presence of very low levels (2.1 molecules/1000 cells) of gastrin mRNA in LIM 1215 cells by quantitative PCR (15). Unfortunately we have been prevented from quantitating the amount of gastrin mRNA in YAMC cells by PCR by absence of a published mouse gastrin cDNA sequence. However, we have been unable to detect any progastrin-derived peptides or mature amidated gastrin by radioimmunoassay in either cell extracts or media conditioned by YAMC or LIM 1215 cells. Based on the sensitivity of the assay (0.4 fmol), we should have detected levels of progastrin-derived peptides as low as 1 fmol/106 cells. Presumably the discrepancy between our results and the observation that progastrin-derived peptides were present in all 5 colon carcinoma cell lines tested at levels between 17 and 54 fmol/106 cells (13) is a reflection of the fact that LIM 1215 cells had the lowest levels of gastrin mRNA of 7 colon carcinoma cell lines tested (15). Because of the absence of detectable levels of progastrin-derived peptides in YAMC and LIM 1215 cells, we were unable to demonstrate any reduction in levels following expression of antisense gastrin mRNA. However, we were able to demonstrate such a reduction in COS cells transiently transfected with plasmids expressing both sense and antisense gastrin mRNA, compared with COS cells transiently transfected with plasmids expressing sense gastrin mRNA only. We therefore postulate that the observed inhibition of proliferation of YAMC and LIM 1215 cells is a consequence of a reduction in the level of progastrin-derived peptides.
Example 7 Expression of Antisense Gastrin Reduces
Proliferation
Expression of antisense gastrin mRNA
significantly reduced proliferation of both YAMC cells and the colon carcinoma cell line LIM 1215 (Figure 6). In neither case was the inhibition reversed by the addition of gastrin17 to the medium.
Example 8 Gastrin Receptors in YAMC and LIM 1215
cells
The total amount of 125I-CCK8 (10,000 cpm, 2.9 fmol) bound to gastrin/CCK-A + -B receptors on YAMC, LIM 1215 and AR4-2J cells was measured by the method of Kopin et al (24). The AR4-2J cell line was derived from a rat pancreatic carcinoma, and expresses both gastrin/CCK-A + -B receptors. The amount of 125I-gastrin17 (150,000 cpm, 44 fmol) bound to gastrin/CCK-C receptors on YAMC and LIM 1215 cells was measured by the phthalate oil centrifugation assay described by Weinstock and Baldwin (3). Background binding was measured in the presence of 1 μM (A + B) or 10 μM (C) gastrin17. The results are summarised in Table 3.
Table 3
Presence of Gastrin/CCK-C, but not Gastrin/CCK-A or -B
Receptors on YAMC and LIM 1215 Cells
Cell line Cell No. 125I-CCK8 Bound Background
(cpm) (cpm)
Gastrin/CCK-A + -B Receptors
YAMC 104 201 ± 22 171 ± 12
LIM 1215 104 1663 ± 89 1604 ± 106
AR4-2J 104 10567 ± 1341 1866 ± 252
Gastrin/CCK-C Receptor;
YAMC 4.2 × 105 1002 ± 72 420 ± 88
LIM 1215 106 5407 ± 800 700 ± 89 No CCK-A or gastrin/CCK-B receptors were detected on YAMC or LIM 1215 cells by binding of 125I-CCK8, even though gastrin/CCK-A and -B receptors were readily detected on AR4-2J cells. The lack of involvement of either CCK-A or gastrin/CCK-B receptors in the autocrine growth of YAMC cells was confirmed by demonstrating that neither the CCK- A-receptor selective antagonist L364,718, nor the
gastrin/CCK-B receptor-selective antagonist L365,260, inhibited YAMC cell proliferation at concentrations
(100 nM) sufficient to saturate their respective receptors (data not shown).
Gastrin/CCK-C receptors were readily detected on both YAMC and LIM 1215 cells using the phthalate oil centrifugation assay and 125I-gastrin17 as ligand.
Example 9 Inhibition of Cell Proliferation by
Receptor Antagonists
The effect of non-selective gastrin/CCK receptor antagonists on the proliferation of YAMC cells was
measured. The non-selective gastrin/CCK receptor
antagonists proglumide and benzotript were both able to inhibit proliferation of YAMC cells, as assessed using the MTT assay at two days after seeding of the cells. The results of one experiment are shown in Figure 7.
Values for IC50 and for the predicted ordinate intercept were as follows: proglumide, 7.4 mM, 119% ;
benzotript, 13.7 mM, 113%. Concentrations of benzotript higher than 3 mM were toxic to YAMC cells. No inhibition of proliferation was observed with the CCK-A receptor-selective antagonist L364,718 or the gastrin/CCK-B
receptor-selective antagonist L365,260 at 100 nM. Similar results were obtained in 2 further experiments.
Our overall results of inhibition of
proliferation of YAMC by proglumide (IC50 value of 7.2 ± 0.4 mM) and by benzotript (IC50 value of 8.0 ± 8.1 mM) were consistent with the involvement of the gastrin/CCK-C receptor in the autocrine growth of YAMC. Similarly, we have found that proliferation of LIM 1215 cells and
LIM 2412 cells is inhibited by proglumide, with IC50 values of 2.2 ± 1.0 mM, and 2.9 ± 1.3 mM respectively. No correlation was observed between the degree of inhibition by proglumide and the presence or absence of binding sites for 125I-[Nle15]-gastrin17 determined previously. Even though gastrin17 breakdown did not exceed 30% during a 24 hour incubation in the absence of serum and a non-saturating concentration of inhibitor was chosen to facilitate ready reversal of inhibition, no reversal of the inhibition of LIM 1215 or LIM 2412 cells by proglumide was detected with gastrin17 concentrations as high as 50 μM. These results are shown in Figure 8.
Example 10 Inhibition of Proliferation of LIM 1215
Cells by Non-Steroidal Anti-Inflammatory Drugs
NSAIDS have been shown to inhibit colorectal tumour growth in several experimental models, and have been shown to reduce the size and number of colorectal polyps in patients with familial adenomatous polyposis (26), and to reduce the death rate from cancer of the eosophagus, stomach, colon, and rectum (27). It has been widely accepted that the effect of NSAIDS on colon cancers has resulted from the ability of these agents to inhibit cyclooxygenases, which are key enzymes in the synthesis of prostaglandins, and in particular it has been proposed that the anti-tumour activity is correlated with inhibition of cyclooxygenase-2 (28).
We have compared the ability of a variety of NSAIDS to inhibit the proliferation of the colon carcinoma cell line LIM 1215 with their ability to inhibit
cyclooxygenase I and cyclooxygenase II. Cell proliferation was measured using the colorimetric assay described above. For these experiments, the medium was RPMI 1640 containing 10 μM thioglycerol, 25 units/ml insulin, 1 mg/ml
hydrocortisone and 10% foetal calf serum. After 24-hours, fresh medium containing the test drug was added. Otherwise the assay was as described above. Figure 9 shows results for indomethacin, sulindac sulphoxide and aspirin. All of these agents inhibited proliferation of LIM 1215 cells. In fact, all of the NSAIDS tested inhibited proliferation of LIM 1215 cells, with IC50 values obtained by computer curve fitting with the programmes EBDA and LIGAND ranging from 120 μM for meclofenamic acid and sulindac sulphide to 21 mM for acetaminophen. These results are summarized in Table 4, and are also compared for some agents with IC50 values for cyclooxygenase I and cyclooxygenase II, which have been reported in the literature.
Table 4
IC50 Values for Inhibition of Colon Carcinoma Cell Proliferation by NSAIDS
IC50 (μM)
NSAIDS
LIM 1215 GBP Cross-linking Cyclo-oxygenase I Cyclo-oxygenase II Cell Proliferation
Acetaminophen 21000 ± 14000 10000 17 .9* 132*
Aspirin 1700 ± 640 9300 ± 4700 1 .7 278
Diclofenac 120 ± 110 160 ± 90 1 .6 1.1
Ibuprofen 290 ± 160 2300 ± 600 4 .8 73
Indomethacin 200 ± 130 150 ± 40 0 .03 1.7
Naproxen 660 ± 460 1200 ± 400 9 .6 5.7
Salicylate 1750 ± 400 3300 ± 1400 219 625
Diflunisal 160 ± 80 80 ± 30
Meclofenamate 120 ± 70 90 ± 50
Mefenamate 200 ± 60 220 ± 60
Niflumate 130 ± 100 290 ± 180
Phenylbutazone 600 ± 180 2200 ± 400
Piroxicam 690 ± 390 2600 ± 500
Sulindac S 120 ± 120 40 ± 10
Sulindac 330 ± 170 580 ± 260
Tiaprofenate 440 ± 230 1400 ± 400
Tolmetin 1900 ± 170 1400 ± 100
* IC30 values are shown because 50% inhibition of cyclooxygenase II was not achieved by concentrations as high as 1 mg/ml (28) .
Figure 10 compares IC50 values for LIM 1215 cell proliferation and for inhibition of cyclooxygenases I and II in intact bovine aortic endothelial cells and murine macrophages respectively, and shows that there is no correlation between inhibition of cell proliferation and enzyme inhibition for either enzyme. In particular, the potent cyclooxygenase inhibitor indomethacin, which has an IC50 value of 30 nM for cyclooxygenase I and 1.7 μM for cyclooxygenase II, was only able to inhibit LIM 1215 cell proliferation to a moderate extent, having an IC50 value of 200 μM. Moreover, acetaminophen , which does not decrease the risk of gastrointestinal tract cancer (27), was even less effective in inhibiting LIM 1215 cell proliferation, having an IC50 of 21 mM. This agent is more potent than aspirin as an inhibitor of cyclooxygenase II, with an IC50 value of 130 μM (28). Our results provide further evidence that the anti-proliferative affects of NSAIDS on colon cancers are not primarily mediated by cyclooxygenase II.
Example 11 Non-Steroidal Anti-Inflammatory Drugs
Inhibit Cross-Linking of Gastrin Analogue to Gastrin-Binding Protein
Using the cross-linking assay described above, the ability of several NSAIDS to inhibit the cross-linking of 125I-gastrin2,17 to gastrin-binding protein was examined. The results for sulindac sulphide, indomethacin, ibuprofen and aspirin are presented in Figure 11. All four agents inhibited the cross-linking in a concentration-dependent manner, with the order of activity being sulindac
sulphide > indomethacin > ibuprofen > aspirin.
The correlation between ability to inhibit in the gastrin-gastrin binding protein cross-linking assay and the ability to inhibit proliferation of LIM 1215 cells was therefore investigated, using sixteen different NSAIDS. The results are shown in Figure 12. Anti-proliferative activity was strongly correlated with the IC50 value in the gastrin binding protein cross-linking assay, with a correlation coefficient of 0.87.
Again the results suggested that the strongest activity was shown by sulindac sulphide, and the weakest by aspirin. No inhibition was detected with acetaminophen at concentrations as high as 10 mM.
We conclude that the gastrin-binding protein cross-linking assay is useful, either alone or in
conjunction with the cell proliferation assay, to identify NSAIDS useful for treatment of diseases involving rapidly proliferating cells, especially neoplastic diseases. It is considered that use of both assays for a given candidate comopund gives a more accurate prediction of activity than the use of either assay alone.
CONCLUSIONS
Identity of the 78 kDa GBP and the Gastrin/CCK-C Receptor
The IC50 values determined for inhibition of cross-linking of 125I-[Nle15]-gastrin2,17 to the 78 kDa GBP by gastrin17, CCK8 and their analogues correlate well with the values previously reported for the gastrin/CCK-C receptor on gastrointestinal cell lines (2,3), as shown in Figure 3B. In contrast, the correlation with the values reported for binding to the CCK-A and gastrin/CCK-B
receptors (Figure 3A) is poor. Similarly, for the
antagonists proglumide and benzotript a good correlation is observed in the case of the gastrin/CCK-C receptor only (Figure 3B).
Anti-proliferative Gastrin/CCK Receptor Antagonists Target the Gastrin/CCK-C Receptor
A good correlation is also observed between the IC50 values for inhibition of cross-linking of
125I-[Nle15]-gastrin2,17 to the 78 kDa GBP by the
non-selective gastrin/CCK receptor antagonists proglumide and benzotript, and the IC50 values for inhibition of HCT 116 colon carcinoma cell proliferation by the same
antagonists (2; Table 1). We have previously reported that the CCK-A receptor-selective antagonist L364,718 and the gastrin/CCK-B receptor-selective antagonist L365,260 had no effect on colon carcinoma cell proliferation at
concentrations as high as 1 μM. Furthermore, the
inhibition observed at higher concentrations of L364,718 was not reversed by addition of gastrin17 or CCK8SO4, and hence was not caused by binding to cell surface gastrin or CCK receptors (4).
We have now found that neither L364,718 nor
L365,260 has any effect on cross-linking of
125I-[Nle15]-gastrin2,17 to the 78 kDa GBP at concentrations up to 200 μM. The effects of antagonists on cross-linking are consistent with the conclusion that binding of
antagonists to the gastrin/CCK-C receptor inhibits cell growth.
NSAIDS inhibit cell proliferation via the GBP
CyclooxygenaseS are involved in prostaglandin synthesis, and utilize fatty acid derivatives as
substrates; they are widely thought to be the targets for NSAIDS. The rat mitochondrial trifunctional protein (MTP), the α-subunit of which has strong amino acid sequence homology to the porcine GBP, also utilizes fatty acid derivatives as substrates. We have shown that NSAIDS, with the exception of acetaminophen, inhibit cross-linking of iodinated gastrin to the porcine GBP, and conclude that GBP may be the target of the anti-proliferative effect of these agents on colon cancer.
Hereditary defects in the MTP can cause death in early childhood (29). A wide range of diagnostic symptoms has been reported, including diarrhoea, vomiting,
hypoketotic hypoglycaemic coma, myopathy and
cardiomyopathy. Pathological examination reveals
widespread fat deposition in liver, heart and kidneys.
Similar symptoms and pathology have been described in
Reye's syndrome, which affects children treated with aspirin during viral infection (30). We suggest that the previously unsuspected link between these syndromes is the MTP. Limitation of caloric intake during illness would result in an increased reliance as a metabolic fuel on acetyl CoA, which is one product of the reaction catalysed by the β-subunit of the MTP. Blockade of fatty acid oxidation by the MTP, either by mutation or by inhibition with aspirin, would then result in hypoketotic
hypoglycemia. Hence the observation of a negative
correlation between acetaminophen usage and Reye's syndrome is consistent with the failure of acetaminophen to inhibit cross-linking of gastrin to the GBP at concentrations as high as 10 mM. This hypothesis predicts that NSAIDS will not inhibit the proliferation of cell lines derived from patients with MTP mutations, and enables rational design of more potent and selective MTP/GBP antagonists for use in novel therapies for colon cancer.
Expression of gastrin CCK-C receptors by YAMC and LIM 1215 cells
We have previously reported that LIM 1215 colon carcinoma cells express cell-surface gastrin/CCK-C
receptors (3). This observation was consistent with our observation that proliferation of LIM 1215 cells was inhibited by the non-selective gastrin/CCK receptor
antagonist proglumide (Example 9, and our unpublished results 10), and with the lack of inhibition by the CCK-A receptor-selective antagonist L364,718 or the gastrin/CCK-C receptor-selective antagonist L365,260 at concentrations as high as 1 μM (4).
We have now found that the conditionally immortalised mouse colon cell line YAMC also expresses cell surface gastrin/CCK-C receptors, but not CCK-A or
gastrin/CCK-B receptors. YAMC cell proliferation is unaffected by the selective antagonists L364,718 and
L365,260 at concentrations as high as 0.1 μM, but is inhibited by the non-selective gastrin/CCK receptor antagonists proglumide and benzotript. Moreover the IC50 values which we have observed for these antagonists are consistent with the hypothesis that the gastrin/CCK-C receptor is the target for the inhibitory effects (6).
Role of the Gastrin/CCK-C Receptor in Autocrine Growth
Without wishing to be bound by any proposed mechanism for the observed effect, our observations provide a possible explanation for the inhibitory effects of gastrin/CCK receptor antagonists on cell proliferation. Competition between antagonists and gastrin, and between acyl CoA derivatives and gastrin, in the GBP cross-linking assay implies that antagonists will compete for substrate binding to the 78 kDa GBP. If the GBP possesses enoyl CoA hydratase and 3-hydroxyacyl CoA dehydrogenase activities with longer chain acyl CoA derivatives as substrates, as suggested by the binding of short chain acyl CoA
derivatives (Figure 1) and by sequence comparisons with other members of the hydratase/dehydrogenase family (7), then presumably inhibition of fatty acid metabolism by antagonist will block cell growth, because of a reduction either in energy supply or in the availability of essential lipids. Although the competition observed between gastrin and acyl CoA derivatives suggests that gastrin might also inhibit one or both activities, gastrin is able to reverse the inhibition of HCT 116 cell proliferation by proglumide (2), implying that gastrin itself does not block any essential function of the 78 kDa GBP. Direct measurements of the effect of gastrin on the enzyme activities of the purified 78 kDa GBP are required to resolve this
inconsistency.
The inhibition of HCT 116 cell proliferation by proglumide and benzotript, and the reversal of inhibition by concentrations of gastrin in the mM range, is consistent with the 78 kDa GBP being a component of an extracellular autocrine loop involving gastrin. However, no such
reversal of proglumide inhibition by gastrin is observed with LIM 1215 or LIM 2412 cells (10), although the observed competition between antagonists and gastrin for the 78 kDa GBP implies that reversal should be observed if the GBP is accessible to exogenous gastrin. One possible explanation for this discrepancy is that the 78 kDa GBP, the mRNA for which is expressed in all colon carcinoma cell lines tested so far, may appear on the cell surface in only a subset of colon carcinoma cell lines. The observation that of 5 colon carcinoma cell lines tested only HCT 116 and LoVo cells secreted progastrin (1) is also consistent with the hypothesis that only a subset of cell lines utilise gastrin in an extracellular autocrine loop.
Inhibition of proliferation of both YAMC and LIM 1215 cells by expression of antisense gastrin mRNA provides clear evidence for the presence in both cell lines of an autocrine loop involving progastrin-derived peptides. Our evidence implicates the gastrin/CCK-C receptor in the autocrine loop in both YAMC and LIM 1215 cells. While binding studies clearly localise a population of the gastrin/CCK-C receptor to the cell surface in both YAMC and LIM 1215 cells (Table 3), subcellular fractionation has localised a second population of the gastrin/CCK-C receptor to mitochondrial membranes in rat liver (31). Furthermore the failure to reverse antisense inhibition by exogenous gastrin17 indicates that the autocrine loop is exclusively intracellular. We therefore postulate that a novel intracellular autocrine (i.e. intracrine) loop involving amidated gastrin and the gastrin/CCK-C receptor
contributes to proliferation of both YAMC and LIM 1215 cells.
It may therefore be necessary to screen biopsy samples for the presence of cell-surface GBP in order to determine the suitability of a cancer for treatment with an antagonist according to the method of the present
invention. Such screening may be carried out using antibody to GBP, labelled with a detectable marker, and measuring the binding of the labelled antibody to dispersed tumour cells. Alternatively screening may be carried out as described by Weinstock and Baldwin (3).
The use of immunochemical assays of inhibition of binding of labelled antibody, or assays of inhibition of binding of labelled high-affinity antagonists, provide the opportunity for automated assays which can be used for large scale screening of putative antagonists. Assays of the enzyme activities intrinsic to the GBP may also be used for large-scale screening of GBP antagonists. Covalent cross-linking of 125I-gastrin2,17 to the purified 78 kDa GBP provides a convenient assay with which to isolate more potent and selective GBP antagonists capable of blocking cell growth.
References cited herein are listed on the following pages. it will be clearly understood that the invention in its general aspects is not limited to the specific details referred to herein.
REFERENCES
1. Baldwin, G.S. and Whitehead, R.H.
Bailliere's Clin. Endocrinol. and Metab., 1994 8
185-214.
2. Hoosein, N.M., Kiener, P.A., Curry, R.C.,
Rovati, L.C., McGilbra, D.K. and Brattain, M.G.
Cancer Res., 1988 48 7179-7183.
3. Weinstock, J. and Baldwin, G.S.
Cancer Res., 1988 48 932-937.
4. Thumwood, C.M., Ji, H. and Baldwin, G.S.
Exp. Cell Res., 1991 192 189-192.
5. Baldwin, G.S., Chandler, R., Scanlon, D.B. and
Weinstock, J.
J. Biol. Chem., 1986 261 12252-12257.
6. Mantamadiotis, T., Sobieszczuk, P., Weinstock, J.
and Baldwin, G.S.
Biochim. Biophys. Acta, 1993 1170 211-215.
7. Baldwin, G.S.
Comp. Biochem. Physiol., 1993 104B 55-61
8. Lee, Y.M., Beinborn, M., McBride, E.W., Lu, M.,
Kolakowski, L.F. and Kopin, A.S.
J. Biol. Chem., 1993 268 8164-8169.
9 . Chang, R.S.L., Lotti, V.J., Monaghan, R.L.,
Birnbaum, J., Stapley, E.O., Goetz, M.A.,
Albers-Schonberg, G., Patchett, A.A., Liesch, J.M.,
Hensens, O.D. and Springer, J.P.
Science, 1985 230 177-179.
10. Murphy, V.J., McBride, K., Ji Hong and Baldwin, G.S.
Gastrointestinal Cancer, 1994 In press.
11. Matsumoto, M., Park, J., Sugano, K. and Yamada, T.
"Biological activity of progastrin posttranslational processing intermediates"
Am. J. Physiol., 1987 252 G315-G319.
12. Magous, R. and Bali, J.-P.
"High-affinity binding sites for gastrin on isolated rabbit gastric mucosal cells"
Eur. J. Pharmacol., 1982 82 47-54. 13. Van Solinge, W.W., Nielsen, F.C., Friis-Hansen, L., Falkmer, U.G. and Rehfeld, J.F.
"Expression but incomplete maturation of progastrin in colorectal carcinomas"
Gastroenterology, 1993 104 1099-1107.
14. Baldwin, G.S., Casey, A., Mantamadiotis, T.,
McBride, K., Sizeland, A. and Thumwood, C.M. "PCR cloning and sequence of gastrin mRNA from carcinoma cell lines"
Biochem. Biophys. Res. Commun., 1990 170 691-697.
15. Baldwin, G.S. and Zhang, Q.-X.
"Measurement of gastrin and transforming growth factor α messenger RNA levels in colonic carcinoma cell lines by quantitative polymerase chain
reaction"
Cancer Res., 1992 52 2261-2267.
16. Finley, G.G., Koski, R.A., Melhem, M.F., Pipas, J.M.
and Meisler, A.I.
"Expression of the gastrin gene in the normal human colon and colorectal adenocarcinoma"
Cancer Res., 1993 53 2919-2926.
17. Guo, Y.-S., Baijal, M., Jin, G.-F., Thompson, J.C,
Townsend, CM. and Singh, P.
"Growth-promoting effects of gastrin on mouse colon cancer cells in vitro: Absence of autocrine effects"
In Vitro Cell. Dev. Biol., 1990 26 871-877.
18. Boel, E., Vuust, J., Norris, F., Norris, K.,
Wind, A., Rehfeld, J.F. and Marcker, K.A.
"Molecular cloning of human gastrin cDNA: Evidence for evolution of gastrin by gene duplication"
Proc. Natl. Acad. Sci. USA., 1983 80 2866-2869.
19. Whitehead, R.H., VanEeden, P.E., Noble, M.D.,
Ataliotis, P. and Jat, P.S.
"Establishment of conditionally immortalized epithelial cell lines from both colon and small intestine of adult H-2Kb-tsA58 transgenic mice" Proc. Natl. Acad. Sci. USA, 1993 90 587-591. 20. Whitehead, R.H., Macrae, F.A., St. John, D.J.B. and Ma, J.
"A colon cancer cell line (LIM 1215) derived from a patient with inherited nonpolyposis colorectal cancer"
J. Natl. Cancer Inst., 1985 74 759-765.
21. Mossman, T.
"Rapid colorimetric assay for cellular growth and survival: Application to proliferation and
cytotoxicity assays"
J. Immunol. Methods, 1983 65 55-63.
22. Ciccotosto, G. and Shulkes, A.
"Pharmacokinetics and organ specific metabolism of glycine-extended and amidated gastrin in sheep" Am. J. Physiol., 1992 263 G802-G809.
23. Rehfeld, J.F. and Bardram, L.
"Processing-independent analysis (PIA) - A new diagnostic tool"
Scan J. Clin. Lab. Invest., 1991 204 9-16.
24. Kopin, A.S., Lee, Y.-M., McBride, E.W.,
Miller, L.J., Lu, M., Lin, H.Y., Kolakowski, L.F. and Beinborn, M.
"Expression cloning and characterization of the canine parietal cell gastrin receptor"
Proc. Natl. Acad. Sci. USA., 1992 89 3605-3609.
25. Feigner, P.L., Gadek, T.R., Holm, M., Roman, R.,
Chan, H.W., Wenz, M., Northrop, J.P., Ringold, G.M. and Danielsen, M.
"Lipofection: A highly efficient, lipid-mediated DNA-transfection procedure"
Proc. Natl. Acad. Sci. USA, 1987 84 7413-7417.
26. Giardiello, F.M., Hamilton, S.R., Krush, A.J.,
Piantadosi, S., Hylind, L.M., Celano,
P., Booker, S.V., Robinson, CR. and
Offerhaus, G.J.A.
New Eng. J. Med., 1993 328 1313-1316 27. Thun, M.J., Namboodiri, M.M., Calle, E.E. ,
Flanders, W.D. and Heath, C.W.
"Aspirin use and risk of fatal cancer"
Cancer Res., 1993 53 1322-1327
28. Mitchell, J.A., Akarasereenont, P., Thiemermann, C. ,
Flower, R.J. and Vane, J.R.
"Selectivity of nonsteroidal antiinflammatory drugs as inhibitors of constitutive and inducible
cyclooxygenase"
Proc. Natl. Acad. Sci. USA., 1994 90 11693-11697
29. R.J.A. Wanders et al
Lancet, 1989 ii 52
30. K.M. Starko, C.G. Ray, L.B. Dominguez,
W.L. Stromberg, D.F. Woodall
Pediatrics, 1980 66 859
31. Uchida, Y., Izai, K., Orii, T. and Hashimoto, T.
"Novel fatty acid β-oxidation enzymes in rat liver mitochondria. II. Purification and properties of enoyl-coenzyme A (CoA) hydratase/3-hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase trifunctional enzyme"
J. Biol. Chem., 1992 267 1034-1041.

Claims

CLAIMS :
1. A method of identifying a compound having the ability to block the low-affinity gastrin-cholecystokinin type C receptor, comprising the step of measuring the ability of said compound to block the binding to gastrin-binding protein of a compound selected from the group consisting of a gastrin-related peptide, a cholecystokinin-related peptide, an antagonist of gastrin or of
cholecystokinin, an acyl CoA, an enoyl CoA, an antibody to gastrin, and an antibody to cholecystokinin.
2. A method according to Claim 1 in which the binding is irreversible.
3. A method according to Claim 2 in which inhibition of irreversible binding is assessed by measuring the ability of the compound to block cross-linking of a gastrin
analogue to gastrin binding protein.
4. A method according to Claim 3 in which the cross-linking is effected by a homobifunctional cross-linking agent.
5. A method according to Claim 4 in which the
homobifunctional cross-linking agents is selected from the group consisting of disuccinimidyl suberate,
disulfosuccinimidyl suberate, and ethylene glycol bis-(succinimidyl)-succinate.
6. A method according to Claim 3 in which the cross-linking is effected by a heterobifunctional cross-linking agent.
7. A method according to Claim 6 in which the
heterobifunctional cross-linking agent is succinimidyl-4-(p-maleimidophenyl) butyrate.
8. A method according to Claim 3 in which the cross-linking is effected by a photoreactive cross-linking agent.
9. A method according to Claim 8 in which the
photoreactive agent is N-hydroxysuccinimidyl-4-azidobenzoate.
10. A method according to any one of Claims 3 to 9 in which the detectable marker is biotin or is a radioactive or fluorescent label.
11. A method according to Claim 1 in which the binding is reversible.
12. A method according to Claim 11 in which the ability of the putative antagonist to prevent binding of a known agonist or antagonist of very high affinity is measured, said known agonist or antagonist being labelled with a detectable marker.
13. A method according to Claim 12 in which the known agonist or antagonist has at least the binding affinity of benzotript.
14. A method according to Claim 12 or Claim 13 in which the detectable marker is 3H.
15. A method according to Claim 11 in which the ability of the putative antagonist to prevent binding of a high affinity antibody is measured, said antibody being labelled with a detectable marker.
16. A method according to Claim 15, in which the
antibody is capable of blocking binding of an acyl CoA or of gastrin to gastrin-binding protein.
17. A method according to Claim 16 in which the
antibody is a monoclonal antibody.
18. A method according to Claim 1 in which the
inhibition of the enoyl CoA hydratase and/or 3-hydroxyacyl CoA dehydrogenase activity intrinsic to the GBP is assayed.
19. A method according to Claim 18 in which the
hydratase or dehydrogenase activity is measured
spectrophotometrically or using a substrate labelled with a radioactive marker.
20. A method according to Claim 19 in which the activity is measured spectrophotometrically by the decrease in enoyl CoA absorption on hydration or by the decrease in NADH absorption on oxidation.
21. A method according to any one of Claims 1 to 20 in which the gastrin-related peptide is [Leu15]gastrin17 or
[Nle15]gastrin2,17.
22. A method according to Claim 21 in which the gastrin-related peptide is [Nle15]gastrin2,17.
23. A pharmaceutical composition of a compound with the ability to inhibit the binding of a gastrin analogue to the low-affinity gastrin-cholecystokinin type C receptor, comprising a pharmaceutically-effective amount of said compound and a pharmaceutically-acceptable carrier, with the proviso that the compound is not idomethacin, sulindac, piroxicam, aspirin, proglumide or benzotript.
24. A composition according to Claim 23, in which the compound is a non-selective antagonist of the gastrin-cholecystokinin receptor.
25. A composition according to Claim 24 in which the antagonist has an 1C50 value in the millimolar range or lower.
26. A composition according to any one of Claims 23 to 25 which additionally comprises another pharmaceutically-active molecule.
27. A composition according to Claim 26 in which the other pharmaceutically-active molecule is an antacid.
28. A composition according to Claim 26 in which the other pharmaceutically-active molecule is an anti-cancer agent.
29. A composition according to Claim 28 in which the anti-cancer agent is a cytokine.
30. A method of controlling gastric acid secretion, comprising the step of administering to a mammal in need of such treatment an effective amount of a compound having the ability to inhibit binding of a gastrin peptide to gastrin-binding protein.
31. A method according to Claim 30, comprising the step of administering a composition according to Claim 27.
32. A method of treating neoplastic disease comprising the step of administering to a mammal in need of such treatment an effective amount of a compound having the ability to inhibit binding of a gastrin peptide to gastrin-binding protein, with the proviso that the compound is not indomethacin, sulindac, piroxicam, aspirin, proglumide or benzotript.
33. A method according to Claim 32, in which the neoplastic disease is a cancer of the gastrointestinal tract.
34. A method according to Claim 32, in which the neoplastic disease is selected from the group consisting of colon carcinoma, gastric carcinoma, mammary carcinoma, malignant melanoma, tumours of epithelial origin, and lymphomas .
35. A method according to any one of Claims 32 to 34, comprising the step of administering a composition
according to Claim 28 or Claim 29.
36. A method according to any one of Claims 32 to 35, in which the compound having the ability to inhibit binding of the gastrin peptide to gastrin-binding protein is a non-steroidal anti-inflammatory compound.
37. A method according to Claim 36 in which the non-steroidal anti-inflammatory compound is an inhibitor of prostaglandin activity.
38. A method of treatment of a neoplastic disease, comprising the step of inducing production of antisense gastrin mRNA in neoplastic tissue of an animal in need of such treatment.
39. A method according to Claim 38, in which tissue-specific production of anti-sense mRNA is achieved by targeting of corresponding DNA to the desired tissue, using tissue-specific promoters.
40. A method according to Claim 38 or Claim 39, in which the neoplastic disease is selected from the group
consisting of colon carcinoma, gastric carcinoma, other gastrointestinal cancers, mammary carcinoma, malignant melanoma, tumours of epithelial origin, and lymphomas.
PCT/US1995/001375 1994-02-02 1995-02-02 Assay for screening of receptor antagonists WO1995021380A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU17405/95A AU1740595A (en) 1994-02-02 1995-02-02 Assay for screening of receptor antagonists

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
AUPM3650A AUPM365094A0 (en) 1994-02-02 1994-02-02 Assay for screening of receptor antagonists
AUPM3650 1994-02-02
AUPM6638 1994-07-05
AUPM6638A AUPM663894A0 (en) 1994-07-05 1994-07-05 Assay for screening of receptor antagonists
AUPM7779 1994-08-31
AUPM7779A AUPM777994A0 (en) 1994-08-31 1994-08-31 Assay for screening of receptor antagonists

Publications (1)

Publication Number Publication Date
WO1995021380A1 true WO1995021380A1 (en) 1995-08-10

Family

ID=27157767

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/001375 WO1995021380A1 (en) 1994-02-02 1995-02-02 Assay for screening of receptor antagonists

Country Status (1)

Country Link
WO (1) WO1995021380A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1185303A1 (en) * 1999-05-24 2002-03-13 George Tachas Inhibition of gastric acid production and/or secretion
EP1409541A2 (en) * 2001-06-05 2004-04-21 Duke University A single-cell biosensor for the measurement of gpcr ligands in a test sample
WO2006008649A1 (en) * 2004-07-15 2006-01-26 Aphton Corporation Anti-sense polynucleotide therapy for gastrin-promoted tumors
US11583576B2 (en) 2017-06-15 2023-02-21 Cancer Advances Inc. Compositions and methods for inducing humoral and cellular immunities against tumors and cancer

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BIOCHEMICA ET BIOPHYSICA ACTA, Vol. 1170, issued 1993, T. MANTAMADIOTIS et al., "Nucleotide Sequence Encoding a Novel Member of the Hydratase/Dehydrogenase Family", pages 211-215. *
COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY-B COMPARATIVE BIOCHEMISTRY, Vol. 104B, No. 1, issued 1993, G.S. BALDWIN, "Comparison of Sequences of the 78 kDa Gastrin-binding Protein and Some Enzymes Involved in Fatty Acid Oxidation", pages 55-61. *
EMBO JOURNAL, Vol. 8, issued 1989, D. BECKER et al., "Proliferation of Human Malignant Melanomas is Inhibited by Antisense Oligodeoxynucleotides Targeted Against Basic Fibroblast Growth Factor", pages 3685-3691. *
PROC. NATL. ACAD. SCI. U.S.A., Vol. 84, issued May 1987, F-T. MU et al., "Monoclonal Antibody to the Gastrin Receptor on Parietal Cells Recognizes a 78-kDa Protein", pages 2698-2702. *
PROC. NATL. ACAD. SCI. U.S.A., Vol. 91, issued August 1994, G.S. BALDWIN, "Antiproliferative Gastrin/cholecystokinin Receptor Antagonists Target the 78-kDa Gastrin-binding Protein", pages 7593-7597. *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1185303A1 (en) * 1999-05-24 2002-03-13 George Tachas Inhibition of gastric acid production and/or secretion
EP1185303A4 (en) * 1999-05-24 2003-09-10 George Tachas Inhibition of gastric acid production and/or secretion
US7501400B1 (en) 1999-05-24 2009-03-10 George Tachas Inhibition of gastric acid production and/or secretion
EP1409541A2 (en) * 2001-06-05 2004-04-21 Duke University A single-cell biosensor for the measurement of gpcr ligands in a test sample
EP1409541A4 (en) * 2001-06-05 2006-09-20 Univ Duke A single-cell biosensor for the measurement of gpcr ligands in a test sample
WO2006008649A1 (en) * 2004-07-15 2006-01-26 Aphton Corporation Anti-sense polynucleotide therapy for gastrin-promoted tumors
US11583576B2 (en) 2017-06-15 2023-02-21 Cancer Advances Inc. Compositions and methods for inducing humoral and cellular immunities against tumors and cancer

Similar Documents

Publication Publication Date Title
Dufresne et al. Cholecystokinin and gastrin receptors
Probst-Kepper et al. An alternative open reading frame of the human macrophage colony-stimulating factor gene is independently translated and codes for an antigenic peptide of 14 amino acids recognized by tumor-infiltrating CD8 T lymphocytes
Rehfeld Accurate measurement of cholecystokinin in plasma
Garland et al. Agonist-induced internalization of the substance P (NK1) receptor expressed in epithelial cells
Rehfeld et al. Gastrin in human bronchogenic carcinomas: constant expression but variable processing of progastrin
US6297022B1 (en) Method of identifying agonists and antagonists for tumor necrosis related receptor TR1
EP1280552A2 (en) Compositions and methods of treating cancer using compositions comprising an inhibitor of endothelin receptor activity
HU228479B1 (en) Immunological process for detecting antibodies directed towards tissue transglutaminase (ttg), use of ttg for diagnostic purposes and therapy control, and oral pharmaceutical agent containing ttg
BR112015012238B1 (en) GLUCAGON/GLP-1 AGONIST PEPITIDES, PHARMACEUTICAL COMPOSITION AND KIT
Bardram Progastrin in serum from Zollinger-Ellison patients: an indicator of malignancy?
US6346388B1 (en) Method of identifying agonist and antagonists for tumor necrosis related receptors TR1 and TR2
Kleemann et al. Receptor binding and cellular uptake studies of macrophage migration inhibitory factor (MIF): use of biologically active labeled MIF derivatives
US20050250702A1 (en) Controlling availability or activity of proteins by use of protease inhibitors or receptor fragments
Wang et al. long-acting human growth hormone receptor antagonists produced in E. Coli and conjugated with polyethylene glycol
Varro et al. Gastrin: an analytical review
WO1995021380A1 (en) Assay for screening of receptor antagonists
JP2003521244A (en) DNA encoding a novel RG1 polypeptide
Law et al. Agonist binding to rat brain somatostatin receptors alters the interaction of the receptors with guanine nucleotide-binding regulatory proteins.
AUTHIER et al. Uptake and metabolic fate of [HisA8, HisB4, GluB10, HisB27] insulin in rat liver in vivo
CA2500665A1 (en) Methods of regulating body weight
EP1298439B1 (en) Method for screening mch receptor antagonist/agonist
US20020068036A1 (en) Novel methods of diagnosis of prostate cancer and/or breast cancer, compositions, and methods of screening for prostate cancer and /or breast cancer modulators
US20100279950A1 (en) Peptides of a melanoma antigen and their use in diagnostic, prophylactic, and therapeutic methods
Rehfeld Processing of precursors of gastroenteropancreatic hormones: diagnostic significance
US20050026224A1 (en) Methods and compositions for modulating G-protein coupled receptor 54

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA CN FI JP KR NO US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: CA

122 Ep: pct application non-entry in european phase