Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Diabetologia ; 53(12): 2641-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20835917

ABSTRACT

AIMS/HYPOTHESIS: Glucagon-like peptide-1 (GLP-1) receptors are highly overexpressed in benign insulinomas, permitting in vivo tumour visualisation with GLP-1 receptor scanning. The present study sought to evaluate the GLP-1 receptor status in vitro in other pancreatic disorders leading to hyperinsulinaemic hypoglycaemia, specifically after gastric bypass surgery. METHODS: Fresh frozen pancreatic tissue samples (n=7) from six gastric bypass surgery patients suffering from hyperinsulinaemic hypoglycaemia were evaluated for GLP-1 receptor content using in vitro receptor autoradiography, and compared with normal pancreas and with pancreatic insulinoma tissues. RESULTS: GLP-1 receptor analysis of the pancreatic tissues, which histopathologically were compatible with nesidioblastosis and originated from post-bypass hypoglycaemic patients, revealed a mean density value of GLP-1 receptors in the islets of 1,483 ± 183 dpm/mg tissue. Pharmacological characterisation indicated the presence of specific GLP-1 receptors. The density of islet GLP-1 receptor in post-gastric bypass patients did not differ from that of normal pancreas (1,563 ± 104 dpm/mg tissue, n = 10). Receptor density in pancreatic acini was low in post-bypass and control conditions. In contrast, benign insulinomas showed a high density of GLP-1 receptors, with a mean value of 8,302 ± 1,073 dpm/mg tissue (n = 6). CONCLUSIONS/INTERPRETATION: In contrast to insulinoma, hyperinsulinaemic hypoglycaemia after gastric bypass surgery is not accompanied by overexpression of GLP-1 receptor in individual islets. Thus, patients with post-gastric bypass hyperinsulinaemic hypoglycaemia are not candidates for GLP-1 receptor imaging in vivo using radiolabelled exendin. These GLP-1 receptor data support the notion that the islet pathobiology of post-gastric bypass hypoglycaemia is distinctly different from that of benign insulinomas.


Subject(s)
Gastric Bypass , Hyperinsulinism/metabolism , Hypoglycemia/metabolism , Islets of Langerhans/metabolism , Obesity, Morbid/surgery , Receptors, Glucagon/metabolism , Adult , Aged , Autoradiography , Female , Gastric Bypass/adverse effects , Gastric Bypass/rehabilitation , Glucagon-Like Peptide-1 Receptor , Humans , Hyperinsulinism/complications , Hyperinsulinism/pathology , Hypoglycemia/etiology , Hypoglycemia/pathology , Insulinoma/metabolism , Insulinoma/pathology , Islets of Langerhans/pathology , Male , Middle Aged , Obesity, Morbid/metabolism , Obesity, Morbid/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Severity of Illness Index , Up-Regulation
2.
Mol Cell Endocrinol ; 314(1): 101-9, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19699258

ABSTRACT

Neuropeptide Y (NPY) is abundantly expressed in the nervous system and acts on target cells through NPY receptors. The human adrenal cortex and adrenal tumors express NPY receptor subtype Y1, but its function is unknown. We studied Y1-mediated signaling, steroidogenesis and cell proliferation in human adrenal NCI-H295R cells. Radioactive ligand binding studies showed that H295R cells express Y1 receptor specifically. NPY treatment of H295R cells stimulated the MEK/ERK1/2 pathway, confirming that H295R cells express functional Y1 receptors. Studies of the effect of NPY and related peptide PYY on adrenal steroidogenesis revealed a decrease in 11-deoxycortisol production. RIA measurements of cortisol from cell culture medium confirmed this finding. Co-treatment with the Y1 antagonist BIBP2336 reversed the inhibitory effect of NPY on cortisol production proving specificity of this effect. At mRNA level, NPY decreased HSD3B2 and CYP21A2 expression. However NPY revealed no effect on cell proliferation. Our data show that NPY can directly regulate human adrenal cortisol production.


Subject(s)
Adrenal Glands/cytology , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/metabolism , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Aldosterone/biosynthesis , Cell Line , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Hydrocortisone/biosynthesis , Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , MAP Kinase Kinase Kinases/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/pharmacology , Peptide YY/metabolism , Radioimmunoassay , Receptors, Neuropeptide Y/genetics , Signal Transduction/physiology , Testosterone/biosynthesis
3.
Eur J Nucl Med Mol Imaging ; 34(8): 1228-38, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17287960

ABSTRACT

PURPOSE: It has been shown that some primary human tumours and their metastases, including prostate and breast tumours, overexpress gastrin-releasing peptide (GRP) receptors. Bombesin (BN) is a neuropeptide with a high affinity for these GRP receptors. We demonstrated successful scintigraphic visualisation of BN receptor-positive tumours in preclinical studies using the radiolabelled BN analogue [(111)In-DTPA-Pro(1),Tyr(4)]BN. However, the receptor affinity as well as the serum stability of this analogue leave room for improvement. Therefore new (111)In-labelled BN analogues were synthesised and evaluated in vitro and in vivo. METHODS AND RESULTS: The receptor affinity of the new BN analogues was tested on human GRP receptor-expressing prostate tumour xenografts and rat colon sections. Analogues with high receptor affinity (low nM range) were selected for further evaluation. Incubation in vitro of GRP receptor-expressing rat CA20948 and human PC3 tumour cells with the (111)In-labelled analogues resulted in rapid receptor-mediated uptake and internalisation. The BN analogue with the best receptor affinity and in vitro internalisation characteristics, Cmp 3 ([(111)In-DTPA-ACMpip(5),Tha(6),betaAla(11),Tha(13),Nle(14)]BN(5-14)), was tested in vivo in biodistribution studies using rats bearing GRP receptor-expressing CA20948 tumours, and nude mice bearing human PC3 xenografts. Injection of (111)In-labelled Cmp 3 in these animals showed high, receptor-mediated uptake in receptor-positive organs and tumours which could be visualised using planar gamma camera and microSPECT/CT imaging. CONCLUSION: With their enhanced receptor affinity and their rapid receptor-mediated internalisation in vitro and in vivo, the new BN analogues, and especially Cmp 3, are promising candidates for use in diagnostic molecular imaging and targeted radionuclide therapy of GRP receptor-expressing cancers.


Subject(s)
Bombesin/analogs & derivatives , Indium Radioisotopes/therapeutic use , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/diagnosis , Animals , Cell Line, Tumor , Humans , Male , Mice , Models, Chemical , Neoplasm Metastasis , Neoplasm Transplantation , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/diagnostic imaging , Protein Binding , Radionuclide Imaging , Rats , Rats, Inbred Lew
4.
Eur J Nucl Med Mol Imaging ; 34(7): 982-93, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17225119

ABSTRACT

PURPOSE: Gallium-68 is a metallic positron emitter with a half-life of 68 min that is ideal for the in vivo use of small molecules, such as [68Ga-DOTA,Tyr3]octreotide, in the diagnostic imaging of somatostatin receptor-positive tumours. In preclinical studies it has shown a striking superiority over its 111In-labelled congener. The purpose of this study was to evaluate whether third-generation somatostatin-based, radiogallium-labelled peptides show the same superiority. METHODS: Peptides were synthesised on solid phase. The receptor affinity was determined by in vitro receptor autoradiography. The internalisation rate was studied in AR4-2J and hsst-HEK-transfected cell lines. The pharmacokinetics was studied in a rat xenograft tumour model, AR4-2J. RESULTS: All peptides showed high affinities on hsst2, with the highest affinity for the Ga(III)-complexed peptides. On hsst3 the situation was reversed, with a trend towards lower affinity of the Ga(III) peptides. A significantly increased internalisation rate was found in sst2-expressing cells for all 67Ga-labelled peptides. Internalisation into HEK-sst3 was usually faster for the 111In-labelled peptides. No internalisation was found into sst5. Biodistribution studies employing [67Ga-DOTA,1-Nal3]octreotide in comparison to [111In-DOTA,1-Nal3]octreotide and [67Ga-DOTA,Tyr3]octreotide showed a significantly higher and receptor-mediated uptake of the two 67Ga-labelled peptides in the tumour and somatostatin receptor-positive tissues. A patient study illustrated the potential advantage of a broad receptor subtype profile radiopeptide over a high-affinity sst2-selective radiopeptide. CONCLUSION: This study demonstrates that 67/68Ga-DOTA-octapeptides show distinctly better preclinical, pharmacological performances than the 111In-labelled peptides, especially on sst2-expressing cells and the corresponding animal models. They may be excellent candidates for further development for clinical studies.


Subject(s)
Gallium Radioisotopes/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/metabolism , Somatostatin/pharmacokinetics , Animals , Cell Line, Tumor , Gallium Radioisotopes/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Isotope Labeling/methods , Male , Metabolic Clearance Rate , Organ Specificity , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemistry , Rats , Rats, Inbred Lew , Sensitivity and Specificity , Somatostatin/chemistry , Tissue Distribution
5.
Gut ; 53(10): 1431-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15361490

ABSTRACT

BACKGROUND AND AIMS: Many neuroendocrine gastrointestinal tumours express receptors for the regulatory peptide somatostatin. Among the five existing somatostatin receptor (sst) subtypes, sst2A is the most frequently expressed in these tumours. However, little information is available about the cellular location of sst2A in corresponding non-neoplastic epithelial tissues. METHODS: We searched for sst2A immunoreactive cells in non-neoplastic gastrointestinal tissues, and evaluated their number and immunohistochemical characteristics with neuroendocrine markers. RESULTS: The gastric antrum showed numerous sst2A cells, situated in the epithelium, corresponding to gastrin containing neuroendocrine cells, while the gastric corpus was largely devoid of sst2A cells, including enterochromaffin-like cells. The remaining foregut, namely the duodenum and proximal jejunum, also contained a large number of sst2A cells, all being neuroendocrine cells and many of them characterised as gastrin cells. Sst2A cells were also detected in the midgut, in low numbers in the epithelium of the distal jejunum and ileum, but not in the appendix vermiformis, the caecum, or the hindgut, despite the large number of neuroendocrine cells present in this area. In addition, sst2A cells were found in the whole gastrointestinal tract in the myenteric and submucosal plexus. CONCLUSIONS: While sst2A receptors on antral gastrin cells presumably mediate somatostatin inhibition of gastrin secretion, the effects of somatostatin on motility and ion transport in the lower gastrointestinal tract may be mediated by sst2A receptors in the neural plexus. These data provide a molecular basis for the physiological actions of somatostatin in human gastrointestinal tissue.


Subject(s)
Enteroendocrine Cells/metabolism , Receptors, Somatostatin/metabolism , Gastric Mucosa/metabolism , Humans , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Pyloric Antrum/metabolism , Somatostatin/physiology
6.
Ann N Y Acad Sci ; 1014: 132-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15153427

ABSTRACT

Many neuroendocrine gastrointestinal and lung tumors express sst2A somatostatin receptors. Because the cellular location of sst2A in the corresponding non-neoplastic tissue is unknown, we searched for sst2A immuno-reactive cells and characterized their type in these tissues using a highly specific sst2A antibody (R2-88). Epithelial sst2A cells, identified as neuroendocrine, gastrin-producing cells, were found in large numbers in the antrum and the duodenum, but not in the gastric corpus. They were also present in the proximal jejunum, rarely noted in the distal jejunum and ileum, and absent in the large intestine and the appendix vermiformis. Moreover, sst2A cells were found abundantly in the neural plexus. sst2A receptors on antral gastrin cells could mediate somatostatin inhibition on gastrin secretion, whereas those in the neural plexus could mediate somatostatin effects on motility and ion transport in the lower gastrointestinal tract. Rare sst2A cells in bronchi and bronchioles located basally and parabasally in the gastrointestinal epithelium were detected that could represent stem/progenitor cells. It is currently not clear whether and which of the identified sst2A cells are at the origin of sst2A-positive neuroendocrine gut or lung tumors.


Subject(s)
Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Lung Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Respiratory Mucosa/metabolism , Gastrins/metabolism , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , Lung Neoplasms/pathology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Respiratory Mucosa/pathology
7.
Eur J Nucl Med Mol Imaging ; 30(8): 1134-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12768332

ABSTRACT

Neurotensin (NT) receptors are overexpressed in exocrine pancreatic cancer and Ewing's sarcoma. The potential utility of native NT in cancer diagnosis and therapy is, however, limited by its rapid degradation in vivo. Therefore, NT analogues were synthesised with modified lysine and arginine derivatives to enhance stability and coupled either to DTPA, to enable high specific activity labelling with indium-111 for imaging, or to DOTA, to enable high specific activity labelling with beta-emitting radionuclides, such as lutetium-177 and yttrium-90. Based on serum stability (4 h incubation at 37 degrees C in human serum) and receptor binding affinity, the five most promising analogues were selected and further evaluated in in vitro internalisation studies in human colorectal adenocarcinoma HT29 cells, which overexpress NT receptors. All five NT analogues bound with high affinity to NT receptors on human exocrine pancreatic tumour sections. The analogues could be labelled with (111)In to a high specific activity. The (111)In-labelled compounds were found to be very stable in serum. Incubation of HT29 cells with the (111)In-labelled analogues at 37 degrees C showed rapid receptor-mediated uptake and internalisation. The most promising analogue, peptide 2530 [DTPA-(Pip)Gly-Pro-(PipAm)Gly-Arg-Pro-Tyr-tBuGly-Leu-OH] was further tested in vivo in a biodistribution study using HT29 tumour-bearing nude mice. The results of this study showed low percentages of injected dose per gram tissue of this (111)In-labelled 2530 analogue in receptor-negative organs like blood, spleen, pancreas, liver, muscle and femur. Good uptake was found in the receptor-positive HT29 tumour and high uptake was present in the kidneys. Co-injection of excess unlabelled NT significantly reduced tumour uptake, showing that tumour uptake is a receptor-mediated process. With their enhanced stability, maintained high receptor affinity and rapid receptor-mediated internalisation, the (111)In-labelled DTPA- and DOTA-conjugated NT analogues are excellent candidates for imaging and therapy of exocrine pancreatic cancer, peptide 2530 being the most promising analogue.


Subject(s)
Heterocyclic Compounds, 1-Ring/pharmacokinetics , Indium Radioisotopes/pharmacokinetics , Neurotensin/analogs & derivatives , Neurotensin/pharmacokinetics , Pancreatic Neoplasms/metabolism , Pentetic Acid/pharmacokinetics , Receptors, Neurotensin/metabolism , Animals , Drug Evaluation, Preclinical , Drug Stability , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/therapeutic use , Indium Radioisotopes/chemistry , Indium Radioisotopes/therapeutic use , Isotope Labeling/methods , Metabolic Clearance Rate , Mice , Mice, Nude , Neurotensin/therapeutic use , Organ Specificity , Pancreas/diagnostic imaging , Pancreas/metabolism , Pancreas/radiation effects , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/radiotherapy , Pentetic Acid/chemistry , Pentetic Acid/therapeutic use , Radionuclide Imaging , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Tissue Distribution , Whole-Body Counting
8.
J Med Chem ; 45(21): 4737-47, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12361401

ABSTRACT

We present evidence that members of the corticotropin releasing factor (CRF) family assume distinct structures when interacting with the CRF(1) and CRF(2) receptors. Predictive methods, physicochemical measurements, and structure-activity relationship studies have suggested that CRF, its family members, and competitive antagonists such as astressin [cyclo(30-33)[DPhe(12),Nle(21),Glu(30),Lys(33),Nle(38)]hCRF((12-41))] assume an alpha-helical conformation when interacting with their receptors. We had shown that alpha-helical CRF((9-41)) and sauvagine showed some selectivity for CRF receptors other than that responsible for ACTH secretion(1) and later for CRF2.(2) More recently, we suggested the possibility of a helix-turn-helix motif around a turn encompassing residues 30-33(3) that would confer high affinity for both CRF(1) and CRF(2)(2,4) in agonists and antagonists of all members of the CRF family.(3) On the other hand, the substitutions that conferred ca. 100-fold CRF(2) selectivity to the antagonist antisauvagine-30 [[DPhe(11),His(12)]sauvagine((11-40))] did not confer such property to the corresponding N-terminally extended agonists. We find here that a Glu(32)-Lys(35) side chain to side chain covalent lactam constraint in hCRF and the corresponding Glu(31)-Lys(34) side chain to side chain covalent lactam constraint in sauvagine yield potent ligands that are selective for CRF(2). Additionally, we introduced deletions and substitutions known to increase duration of action to yield antagonists such as cyclo(31-34)[DPhe(11),His(12),C(alpha)MeLeu(13,39),Nle(17),Glu(31),Lys(34)]Ac-sauvagine((8-40)) (astressin(2)-B) with CRF(2) selectivities greater than 100-fold. CRF receptor autoradiography was performed in rat tissue known to express CRF(2) and CRF(1) in order to confirm that astressin(2)-B could indeed bind to established CRF(2) but not CRF(1) receptor-expressing tissues. Extended duration of action of astressin(2)-B vs that of antisauvagine-30 is demonstrated in the CRF(2)-mediated animal model whereby the inhibition of gastric emptying of a solid meal in mice by urocortin administered intraperitoneally at time zero is antagonized by the administration of astressin(2)-B but not by antisauvagine-30 at times -3 and -6 h while both peptides are effective when given 10 min before urocortin.


Subject(s)
Corticotropin-Releasing Hormone/chemistry , Peptide Fragments/chemical synthesis , Peptides, Cyclic/chemical synthesis , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Autoradiography , Binding, Competitive , Brain/anatomy & histology , Brain/metabolism , CHO Cells , Cricetinae , Eating/drug effects , Gastric Emptying/drug effects , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Radioligand Assay , Rats , Structure-Activity Relationship
9.
Eur J Nucl Med ; 28(7): 836-46, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11504080

ABSTRACT

Somatostatin receptors are known to be expressed in a large number of human tumours and represent the basis for in vivo tumour targeting. Stable somatostatin derivatives such as octreotide or lanreotide are the most frequently used radiopharmaceuticals acting through specific binding to somatostatin receptors; however, they do not bind with high affinity to all five receptor subtypes. Whereas the mRNAs for most receptor subtypes have been detected in tumours, it is in most cases unclear which of the receptor subtype proteins are expressed. Since in vitro receptor binding methods are close correlates and predictors of in vivo peptide receptor targeting, we took advantage of the recently developed subtype-selective analogues and evaluated approximately 200 tumours for their receptor subtype protein expression in specific binding assays using autoradiography with 125I-[Leu8, D-Trp22, Tyr25]-somatostatin-28 and displacement by subtype-selective analogues. The majority of the tested neuroblastomas, meningiomas, medulloblastomas, breast carcinomas, lymphomas, renal cell carcinomas, paragangliomas, small cell lung carcinomas and hepatocellular carcinomas predominantly expressed sst2. The prostate carcinomas and sarcomas preferentially expressed sstl, while a majority of inactive pituitary adenomas displayed sst3 and, to a lesser extent, sst2. Growth hormone-secreting pituitary adenomas preferentially expressed sst2 and sst5; gastroenteropancreatic tumours and phaeochromocytomas frequently displayed sst2 and/or sstl. Non-neoplastic human tissues such as vessels, nerve plexus, pancreatic islets, prostatic stroma, adrenal medulla, spleen and germinal centres of the lymphoid tissues preferentially expressed sst2. However, the human gastric mucosa predominantly expressed sst1 while colonic mucosa displayed sst2. Interestingly, a minority of tumours showed a strong 125I-[Leu8, D-Trp22, Tyr25]-somatostatin-28 binding, of which less than 50% could be displaced by the sum of the five subtype-selective analogues. This observation suggests the existence of an as yet unknown subtype in selected tumours. This study is the first report to analyse the somatostatin receptor subtype expression in tumours with binding methods. We conclude that sst2, with high affinity for current radiopharmaceuticals such as Octreoscan, is predominantly expressed in a majority of tumours. Fewer tumour types (sarcomas, prostate cancers, inactive pituitary adenomas) preferentially express another subtype. This information is of importance with regard to the clinical applications and development of somatostatin analogues with distinct receptor subtype selectivities.


Subject(s)
Iodine Radioisotopes , Ligands , Neoplasms/chemistry , Receptors, Somatostatin/analysis , Autoradiography , Humans , In Vitro Techniques , Tissue Distribution
10.
Eur J Pharmacol ; 426(3): 165-73, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11527540

ABSTRACT

A possible cross-competition between vasoactive intestinal peptide (VIP) and somatostatin (somatotropin release inhibiting factor; SRIF) and their respective receptors, was investigated at native or recombinant SRIF and VIP/pituitary adenylate cyclase-activating polypeptide (PACAP) receptors. The activity of VIP was examined in radioligand binding assays at mouse sst(1-5), rat sst(1-2) and human sst(1-5) receptors; or at human tumours preferentially expressing each of the five SRIF receptors. Moreover, SRIF was investigated at human tumoral tissues known to exclusively express specific VIP/PACAP receptor(s). VIP had no significant effect on any of the radioligand binding sites of the SRIF receptor family of rat, mouse or human origin tested. Conversely, SRIF did not interfere with the human VIP/PACAP binding sites tested. Taken together, the results cast reservation on the claimed cross-competition between VIP and SRIF at, specifically human sst(3) receptors, or any of the cloned SRIF or VIP/PACAP receptors recognised to date.


Subject(s)
Receptors, Somatostatin/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Somatostatin/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Binding, Competitive/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , Iodine Radioisotopes , Male , Mice , Neoplasms/metabolism , Neuropeptides/metabolism , Peptides, Cyclic/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Receptors, Somatostatin/genetics , Recombinant Proteins/metabolism , Somatostatin/pharmacology , Vasoactive Intestinal Peptide/analogs & derivatives , Vasoactive Intestinal Peptide/pharmacology
11.
Cancer Res ; 61(11): 4636-41, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389101

ABSTRACT

Overexpression of selected peptide receptors in human tumors has been shown to represent clinically relevant targets for cancer diagnosis and therapy. Neuropeptide Y (NPY) is a peptide neurotransmitter mediating feeding behavior and vasoconstriction. It has never been shown to be involved in human cancer. We show here, using in vitro receptor autoradiography, a NPY receptor incidence of 85% in primary human breast carcinomas (n = 95) and of 100% in lymph node metastases of receptor-positive primaries (n = 27), predominantly as Y(1) subtype, whereas non-neoplastic human breast expressed Y(2) preferentially. Y(1) mRNA was detected in Y(1)-expressing tumors by in situ hybridization, whereas Y(2) mRNA was found in normal breast tissue. The strong predominance of Y(1) in breast carcinomas compared with Y(2) in normal breast suggests that neoplastic transformation can switch the NPY receptor expression from Y(2) to Y(1) subtype. Moreover, in Y(1)-expressing human SK-N-MC tumor cells, an NPY-induced, dose-dependent inhibition of tumor cell growth of >40% was observed, suggesting a functional role of NPY in cancer, mediated by Y(1). NPY should therefore be added to the list of small regulatory peptides related to cancer. The high incidence of Y(1) in in situ, invasive, and metastatic breast cancers allows for the possibility to target them for diagnosis and therapy with NPY analogues.


Subject(s)
Breast Neoplasms/metabolism , Neuropeptide Y/pharmacology , Receptors, Neuropeptide Y/physiology , Adult , Aged , Aged, 80 and over , Autoradiography , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Division/drug effects , Dose-Response Relationship, Drug , Female , Humans , In Situ Hybridization , Middle Aged , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Neuropeptide Y/biosynthesis , Receptors, Neuropeptide Y/genetics , Tumor Cells, Cultured/drug effects
12.
J Med Chem ; 44(13): 2238-46, 2001 Jun 21.
Article in English | MEDLINE | ID: mdl-11405660

ABSTRACT

A family of analogues of des-AA(1,2,5)-[DTrp(8)/D2Nal(8)]-SRIF that contain a 4-(N-isopropyl)-aminomethylphenylalanine (IAmp) at position 9 was identified that has high affinity and selectivity for human somatostatin receptor subtype 1 (sst1). The binding affinities of des-AA(1,2,5)-[DTrp(8),IAmp(9)]-SRIF (c[H-Cys-Lys-Phe-Phe-DTrp-IAmp-Thr-Phe-Thr-Ser-Cys-OH], CH-275) (7), des-AA(1,5)-[Tyr(2),DTrp(8),IAmp(9)]-SRIF (CH-288) (16), des-AA(1,2,5)-[Tyr(7),DTrp(8),IAmp(9)]-SRIF (23), and des-AA(1,2,5)-[DTrp(8),IAmp(9),Tyr(11)]-SRIF (25) are about (1)/(7), (1)/(4), (1)/(125), and (1)/(4) that of SRIF-28 (1) to sst1, respectively, about (1)/(65), (1)/(130), <(1)/(1000), and <(1)/(150) that of 1 to sst3, respectively, and about or less than (1)/(1000) that of 1 to the other three human SRIF receptor subtypes. A substitution of DTrp(8) by D2Nal(8) in 7 to yield des-AA(1,2,5)-[D2Nal(8),IAmp(9)]-SRIF (13) and in 16 to yield des-AA(1,5)-[Tyr(2),D2Nal(8),IAmp(9)]-SRIF (17) was intended to increase chemical stability, selectivity, and affinity and resulted in two analogues that were less potent or equipotent with similar selectivity, respectively. Carbamoylation of the N-terminus as in des-AA(1,2,5)-[DTrp(8),IAmp(9),Tyr(11)]-Cbm-SRIF (27) increased affinity slightly as well as improved selectivity. Monoiodination of 25 to yield 26 and of 27 to yield 28 resulted in an additional 4-fold increase in affinity at sst1. Desamination of the N-terminus of 17 to yield 18, on the other hand, resulted in significant loss of affinity. Attempts at reducing the size of the ring with maintenance of selectivity failed in that des-AA(1,4,5,13)-[Tyr(2),DTrp(8),IAmp(9)]-SRIF (33) and des-AA(1,4,5,6,12,13)-[Tyr(2),DTrp(8),IAmp(9)]-SRIF (34) progressively lost affinity for all receptors. Both des-AA(1,2,5)-[DTrp(8),IAmp(9),Tyr(11)]-Cbm-SRIF (27) and des-AA(1,2,5)-[DCys(3),DTrp(8),IAmp(9),Tyr(11)]-Cbm-SRIF (29) show agonistic activity in a cAMP assay; therefore, the structural basis for the agonist property of this family of analogues is not contingent upon the chirality of the Cys residue at position 3 as shown to be the case in 18-membered ring SRIF octapeptides. None of the high affinity structures described here showed receptor antagonism. We have prepared the radiolabeled des-AA(1,2,5)-[DTrp(8),IAmp(9),(125)ITyr(11)]-SRIF ((125)I-25) and des-AA(1,2,5)-[DTrp(8),IAmp(9), (125)ITyr(11)]-Cbm-SRIF ((125)I-27), used them as in vitro tracers, and found them to be superior to des-AA(1,5)-[(125)ITyr(2),DTrp(8),IAmp(9)]-SRIF ((125)I-16) for the detection of sst1 tumors in receptor autoradiography studies.


Subject(s)
Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , Somatostatin/agonists , Somatostatin/chemical synthesis , Adenylyl Cyclases/metabolism , Animals , Autoradiography , CHO Cells , Cricetinae , Female , Humans , In Situ Hybridization , Leiomyoma/metabolism , Molecular Conformation , Protein Binding , Recombinant Proteins/metabolism , Somatostatin/chemistry , Somatostatin/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured , Uterine Neoplasms/metabolism
13.
Proc Natl Acad Sci U S A ; 97(25): 13973-8, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11095748

ABSTRACT

A family of octapeptide derivatives of somatostatin cyclized via a disulfide bridge (des-AA(1,2,4,5,12,13)[d-2Nal(8)]-somatostatin-14, ODN-8) was identified that has high affinity and selectivity for the human sst(3) somatostatin receptor subtype transfected in CCL39 cells. The binding affinity of carbamoyl-des-AA(1,2,4,5,12, 13)[d-Cys(3),Tyr(7),d-Agl(8)(Me,2-naphthoyl)]-somatostatin-14 (sst(3)-ODN-8) is equal to that of somatostatin-28 for sst(3) and less than one-thousandth that for the other four somatostatin receptor subtypes. Compound sst(3)-ODN-8 potently reverses the somatostatin-28-induced inhibition of forskolin-stimulated cAMP production (pK(B) = 9.07) and reverses the somatostatin-28-induced stimulation of phospholipase C activity (pK(i) = 9.22) in sst(3)-transfected CCL39 cells. [(125)I-Tyr(7)]sst(3)-ODN-8 selectively labels sst(3)-expressing cells with subnanomolar binding affinity (K(D) = 0.27 nM). With the use of this radioligand, sst(3)-expressing human tumors, particularly inactive pituitary adenomas, can be identified with receptor autoradiography; moreover, areas of the human lymphoreticular system express sst(3) binding sites selectively displaced by nanomolar concentrations of sst(3)-ODN-8. Based on the structure-activity relationship of selected analogs substituted at positions 3, 7, and 8, we hypothesize that the basis for sst(3) selectivity, high affinity, and possibly antagonism resides in the ring size of the analog and the unique conformational and structural character of the N-methylated amino-2-naphthoyl side chain of aminoglycine at position 8 and not in the Tyr(7) substitution or in the d-configuration at position 3. The family of labeled and unlabeled sst(3)-ODN-8 analogs represents highly innovative, potent, and specific sst(3)-selective antagonist tools for the study of sst(3)-mediated physiological and pathophysiological conditions that may suggest novel clinical applications.


Subject(s)
Receptors, Somatostatin/antagonists & inhibitors , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Enzyme Activation , Humans , Type C Phospholipases/metabolism
14.
J Clin Endocrinol Metab ; 85(10): 3882-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11061553

ABSTRACT

The distribution of the sst2A receptor was investigated, using immunohistochemistry, with the specific antipeptide antibody R2-88, in a total of 120 tumors of the nervous and the neuroendocrine systems, including small-cell lung carcinomas, medulloblastomas, neuroblastomas, pheochromocytomas, and paragangliomas. The great majority of the tumor samples, frozen or formalin-fixed, showed a positive immunohistochemical staining with R2-88, and an excellent correlation with receptor autoradiography using 125I[Tyr3]-octreotide. Whereas small-cell lung carcinomas and medulloblastomas had a predominantly plasma membrane staining, pheochromocytomas and neuroblastomas had variable ratios of cell surface and intracellular staining. Strikingly, a preferentially cytoplasmic staining was seen in tumors with a high level of somatostatin gene expression, whereas a more plasma membranous staining was seen in tumors lacking somatostatin messenger RNA. Specificity of both the plasma membrane and the cytoplasmic staining pattern was confirmed in immunoblots, which showed the immunoreactive receptor migrating as a characteristic 70-kDa broad band. In both immunohistochemical and immunoblotting experiments, staining was abolished by antibody blockade with 100 nM antigen peptide. These results describe, for the first time, the localization of the sst2A receptor protein in human small-cell lung carcinomas, medulloblastomas, neuroblastomas, and paragangliomas. Moreover, it is the first report investigating possible causes for distinct subcellular localizations of sst2A in human tissues. We show that the subcellular distribution of the receptor may be dependent on the surrounding somatostatin concentration, consistent with both the known effect of somatostatin to cause sst2A receptor internalization and an autocrine regulation of tumors by the peptide they produce. Moreover, our demonstration that the sst2A receptor can be identified in this group of tumors using simple immunohistochemical methods in formalin-fixed, paraffin-embedded material opens numerous diagnostic, therapeutic, and prognostic opportunities.


Subject(s)
Nervous System Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Receptors, Somatostatin/metabolism , Subcellular Fractions/metabolism , Autoradiography , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Intracellular Membranes/metabolism , Membranes/metabolism , Octreotide , Somatostatin/biosynthesis
15.
Cancer Res ; 60(11): 3105-12, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850463

ABSTRACT

The evaluation of peptide receptors in man is needed not only to discover the physiological target tissues of a given peptide but also to identify diseases with a sufficient receptor overexpression for diagnostic or therapeutic interventions. Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP) receptors have been evaluated in human tumors and in their tissues of origin using in vitro receptor autoradiography with 125I-VIP or 125I-acetyl-PACAP-27 in tissue sections. The VIP/PACAP receptor subtypes VPAC1, VPAC2, and PAC1 were evaluated in these tissues by determining the rank order of potencies of VIP and PACAP as well as VPAC1- and VPAC2-selective analogues. The VIP/PACAP receptors expressed in the great majority of the most frequently occurring human tumors, including breast (100% receptor incidence), prostate (100%), pancreas (65%), lung (58%), colon (96%), stomach (54%), liver (49%), and urinary bladder (100%) carcinomas as well as lymphomas (58%) and meningiomas (100%), are predominantly of the VPAC1 type. Their cells or tissues of origin, i.e., hepatocytes, breast lobules and ducts, urothelium, prostate glands, pancreatic ducts, lung acini, gastrointestinal mucosa, and lymphocytes, also predominantly express VPAC1. Leiomyomas predominantly express VPAC2 receptors, whereas paragangliomas, pheochromocytomas, and endometrial carcinomas preferentially express PAC1 receptors. Conversely, VPAC2 receptors are found mainly in smooth muscle (i.e., stomach), in vessels, and in stroma (e.g., of the prostate), whereas PAC1 receptors are present in the adrenal medulla and in some uterine glands. Whereas the very wide distribution of VIP/PACAP receptors in the normal human body is indicative of a key role of these peptides in human physiology, the high VIP/PACAP receptor expression in tumors may represent the molecular basis for clinical applications of VIP/PACAP such as in vivo scintigraphy and radiotherapy of tumors as well as VIP/PACAP analogue treatment for tumor growth inhibition.


Subject(s)
Neoplasms/metabolism , Receptors, Pituitary Hormone/biosynthesis , Receptors, Vasoactive Intestinal Peptide/biosynthesis , Adrenal Gland Neoplasms/metabolism , Adrenal Medulla/metabolism , Autoradiography , Breast/metabolism , Breast Neoplasms/metabolism , Dose-Response Relationship, Drug , Female , Gastric Mucosa/metabolism , Humans , Leiomyoma/metabolism , Lymph Nodes/metabolism , Male , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Peptides/metabolism , Pheochromocytoma/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Stomach Neoplasms/metabolism
16.
Eur J Nucl Med ; 27(3): 273-82, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10774879

ABSTRACT

In vivo somatostatin receptor scintigraphy using Octreoscan is a valuable method for the visualisation of human endocrine tumours and their metastases. Recently, several new, alternative somatostatin radioligands have been synthesised for diagnostic and radiotherapeutic use in vivo. Since human tumours are known to express various somatostatin receptor subtypes, it is mandatory to assess the receptor subtype affinity profile of such somatostatin radiotracers. Using cell lines transfected with somatostatin receptor subtypes sst1, sst2, sst3, sst4 and sst5, we have evaluated the in vitro binding characteristics of labelled (indium, yttrium, gallium) and unlabelled DOTA-[Tyr3]-octreotide, DOTA-octreotide, DOTA-lanreotide, DOTA-vapreotide, DTPA-[Tyr3]-octreotate and DOTA-[Tyr3]-octreotate. Small structural modifications, chelator substitution or metal replacement were shown to considerably affect the binding affinity. A marked improvement of sst2 affinity was found for Ga-DOTA-[Tyr3]-octreotide (IC50 2.5 nM) compared with the Y-labelled compound and Octreoscan. An excellent binding affinity for sst2 in the same range was also found for In-DTPA-[Tyr3]-octreotate (IC50 1.3 nM) and for Y-DOTA-[Tyr3]-octreotate (IC50 1.6 nM). Remarkably, Ga-DOTA-[Tyr3]-octreotate bound at sst2 with a considerably higher affinity (IC50 0.2 nM). An up to 30-fold improvement in sst3 affinity was observed for unlabelled or Y-labelled DOTA-octreotide compared with their Tyr3-containing analogue, suggesting that replacement of Tyr3 by Phe is crucial for high sst3 affinity. Substitution in the octreotide molecule of the DTPA by DOTA improved the sst3 binding affinity 14-fold. Whereas Y-DOTA-lanreotide had only low affinity for sst3 and sst4, it had the highest affinity for sst5 among the tested compounds (IC50 16 nM). Increased binding affinity for sst3 and sst5 was observed for DOTA-[Tyr3]-octreotide, DOTA-lanreotide and DOTA-vapreotide when they were labelled with yttrium. These marked changes in subtype affinity profiles are due not only to the different chemical structures but also to the different charges and hydrophilicity of these compounds. Interestingly, even the coordination geometry of the radiometal complex remote from the pharmacophoric amino acids has a significant influence on affinity profiles as shown with Y-DOTA versus Ga-DOTA in either [Tyr3]-octreotide or [Tyr3]-octreotate. Such changes in sst affinity profiles must be identified in newly designed radiotracers used for somatostatin receptor scintigraphy in order to correctly interpret in vivo scintigraphic data. These observations may represent basic principles relevant to the development of other peptide radioligands.


Subject(s)
Radiopharmaceuticals , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Animals , Autoradiography , Binding, Competitive , Humans , In Situ Hybridization , Octreotide/chemistry , Peptides, Cyclic/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use , Rats , Receptors, Somatostatin/classification , Somatostatin/chemistry , Somatostatin/therapeutic use
17.
Gut ; 45(5): 766-74, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10517918

ABSTRACT

BACKGROUND: Overexpression of regulatory peptide receptors in selected human tumours is of diagnostic and therapeutic relevance. AIMS: To evaluate the expression of somatostatin, vasoactive intestinal peptide (VIP), substance P, cholecystokinin (CCK) A and B, and neurotensin receptors in hepatocellular carcinoma (HCC). METHODS: In vitro receptor autoradiography for the various peptide receptors using selective iodinated radioligands on tissue sections in 59 cases of HCC. RESULTS: 41% of HCC expressed somatostatin receptors; 47% expressed VIP receptors. VIP receptors were always identified in non-neoplastic liver tissue. Substance P receptors were only identified in 5% of HCC but in the majority of their peritumorous and intratumorous vessels. CCK-A and -B and neurotensin receptors were not detected in HCC. The somatostatin receptors showed high affinity for somatostatin and octreotide. The VIP receptors had high affinity for VIP, pituitary adenylate cyclase activating peptide (PACAP) 27, and a VIP1 selective analogue, suggesting the presence of VIP1/PACAP II type receptors. PACAP I receptors were identified in two cases. Substance P receptors were all of the NK1 subtype. The density of somatostatin receptors in HCC was low compared with the density found in liver metastases of neuroendocrine tumours. The VIP receptor density was always lower in HCC than in adjacent liver tissue. CONCLUSIONS: Somatostatin, VIP, and substance P may have a receptor mediated role in HCC. Substance P receptors may be involved in regulation of tumour associated blood flow; somatostatin receptors and VIP receptors may mediate tumour growth. Diagnostic and therapeutic evaluation of somatostatin and VIP analogues may be of interest in receptor positive HCC.


Subject(s)
Carcinoma, Hepatocellular/chemistry , Liver Neoplasms/chemistry , Neoplasm Proteins/analysis , Receptors, Peptide/analysis , Adult , Aged , Autoradiography , Carcinoma, Hepatocellular/diagnosis , Female , Humans , Liver Neoplasms/diagnosis , Male , Middle Aged , Protein Binding , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/analysis , Receptors, Neurokinin-1/analysis , Receptors, Neurotensin/analysis , Receptors, Somatostatin/analysis , Receptors, Vasoactive Intestinal Peptide/analysis
18.
J Clin Endocrinol Metab ; 84(8): 2942-50, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10443702

ABSTRACT

The cellular distribution of the somatostatin sst2A receptor protein was investigated in the lymphatic, smooth muscular, and nervous components of the human gastrointestinal tract using subtype-specific antibody R2-88 for immunohistochemical staining of cryostat and formalin-fixed, paraffin-embedded tissue sections. Germinal centers of intestinal lymphatic follicles were immunostained, exhibiting a predominantly plasma membrane localization of the receptor. Similarly, nerve fibers and cells in the submucosal and myenteric plexus were stained for sst2A. Antibody preabsorption with 100 nmol/L antigen peptide abolished staining in all of these tissues, and immunohistochemical staining correlated with the labeling observed after receptor autoradiography using the sst2-preferring radioligand 125I-[Tyr3]octreotide. Cytoplasmic immunostaining was detected in gastrointestinal smooth muscle cells and was inhibited by antibody pre-absorption with antigen peptide. However, 125I-[Tyr3]octreotide autoradiography was negative, and Western blots showed no band at the usual 70-90 kDa location for sst2A. Instead, a band was observed at 205 kDa. This band comigrated with the rabbit myosin standard, which was also stained with R2-88, although antibody sensitivity for myosin was less than 0.002% of that for the sst2A receptor. Rigorous computer-based sequence analysis demonstrated the peptide sequence chosen for antibody production was unique. Moreover, standard sequence alignment protocols were unable to identify the sequences in myosin responsible for the observed reactivity with the R2-88 antiserum. The observed cross-reactivity emphasizes the need for extensive controls to prove the specificity of immunostaining for such low abundance proteins as receptors even when the peptide sequence chosen for antibody production is unique. This study demonstrates for the first time the presence of specific sst2A receptor protein by immunohistochemistry in the human gastrointestinal lymphatic and nervous components, but not in gastrointestinal circular and longitudinal smooth muscle.


Subject(s)
Digestive System/chemistry , Lymphatic System/chemistry , Muscle, Smooth/chemistry , Peripheral Nervous System/chemistry , Receptors, Somatostatin/analysis , Animals , Artifacts , Cross Reactions , Humans , Immunohistochemistry , Myosins/chemistry , Myosins/immunology , Rabbits , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/immunology , Sequence Alignment
19.
Int J Cancer ; 82(2): 213-8, 1999 Jul 19.
Article in English | MEDLINE | ID: mdl-10389755

ABSTRACT

Receptors for regulatory peptides, such as somatostatin or vasoactive intestinal peptide (VIP), expressed at high density by neoplastic cells, can be instrumental for tumor diagnosis and therapy. Little is known about the expression of neurotensin receptors in human tumors. In the present study, 464 human neoplasms of various types were investigated for their neurotensin receptor content by in vitro receptor autoradiography on tissue sections using 125I-[Tyr3]-neurotensin as radioligand. Neurotensin receptors were identified and localized in tumor cells of 11/17 Ewing's sarcomas, 21/40 meningiomas, 10/23 astrocytomas, 5/13 medulloblastomas, 7/24 medullary thyroid cancers and 2/8 small cell lung cancers. They were rarely found in non-small cell lung cancers and breast carcinomas; they were absent in prostate, ovarian, renal cell and hepatocellular carcinomas, neuroendocrine gut tumors, pituitary adenomas, schwannomas, neuroblastomas and lymphomas. When present, the receptors bound with nanomolar affinity neurotensin and acetyl-neurotensin-(8-13), with lower affinity neuromedin N, diethylenetriamine penta-acetic acidneurotensin-(8-13) and SR 48692, but not neurotensin-(1-11). They were all of the NT1 type, without high affinity for levocabastine. Further, in 2 receptor-positive Ewing's sarcomas, neurotensin mRNA was detected by in situ hybridization techniques. Since neurotensin is known to stimulate cell proliferation, the presence of neurotensin receptors in human neoplasia may be of biological relevance, possibly as an integrative part of an autocrine feedback mechanism of tumor growth stimulation.


Subject(s)
Bone Neoplasms/chemistry , Neoplasm Proteins/analysis , Receptors, Neurotensin/analysis , Sarcoma, Ewing/chemistry , Animals , Humans , Neoplasm Proteins/metabolism , Neoplasms/chemistry , Neurotensin/analogs & derivatives , Neurotensin/metabolism , Organ Specificity , Peptide Fragments/metabolism , Piperidines/metabolism , Pyrazoles/metabolism , Quinolines/metabolism , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Rats , Receptors, Neurotensin/metabolism
20.
Int J Cancer ; 81(3): 376-86, 1999 May 05.
Article in English | MEDLINE | ID: mdl-10209952

ABSTRACT

Receptors for cholecystokinin (CCK), gastrin, neurotensin, somatostatin and vasoactive intestinal peptide (VIP) are over-expressed in several human tumors, where they have diagnostic and therapeutic implications. Since reports on the expression of these peptide receptors in primary gastric and colonic adenocarcinomas are either non-existent or conflicting, a detailed evaluation with particular emphasis on the tissue localization was undertaken. CCK-A, CCK-B, neurotensin, somatostatin and VIP receptors were localized by in vitro receptor autoradiography with iodinated radioligands on histological sections of surgical samples of 27 gastric and 25 colonic adenocarcinomas. CCK-A, CCK-B and neurotensin-1 receptors were found in a minority of both tumor types. Somatostatin receptors were found in 18/27 gastric and 2/25 colonic cancers. VIP receptors were found in 14/26 gastric and 23/25 colonic cancers; subtype characterization suggests VIP1 receptors. In addition, resected tumor samples contained non-malignant tissues (mucosa, smooth muscle, nerves or vessels) with high amounts of the various peptide receptors. Therefore, regulatory peptide receptors are expressed differentially in gastric and colonic cancers but also very frequently in "contaminating" non-malignant tissues. Since results using morphological techniques are superior to those using homogenates, we recommend that localization of these receptors to the tissues should always be attempted, to minimize receptor over-estimation in tumors and to prevent spurious results.


Subject(s)
Adenocarcinoma/chemistry , Gastrointestinal Neoplasms/chemistry , Receptors, Cholecystokinin/analysis , Receptors, Neurotensin/analysis , Receptors, Somatostatin/analysis , Receptors, Vasoactive Intestinal Peptide/analysis , Autoradiography , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...