Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Swiss Med Wkly ; 139(15-16): 233-40, 2009 Apr 18.
Article in English | MEDLINE | ID: mdl-19418307

ABSTRACT

Metzincins, such as matrix metalloproteases (MMP), and extracellular matrix (ECM) proteins are differentially regulated in inflammation. We hypothesised that metzincins are also dysregulated in experimental acute cardiac allograft rejection. We investigated the Dark Agouti-to-Lewis (DA-to-Lew) rat model of acute cardiac allograft rejection. Cyclosporine (CsA) (7.5 mg/kg/d) was given from transplantation to sacrifice (day +5). At that time, mRNA levels were analysed by Affymetrix genechip and quantitative reverse transcription polymerase chain reaction (qRTPCR). MMP protein and activities were analysed by immunohistology, fluorometry, zymography and Western blots. In untreated rejected DA allografts, mRNA levels of MMP-2/-7/-9/-/12-/14, a disintegrin and metalloprotease (ADAM)-17, tissue inhibitor of metalloprotease (TIMP)-1/-3 were increased, whereas MMP-11/-16/-24 and TIMP-2/-4 were lowered compared to native DA hearts. With respect to these untreated allografts, CsA lowered mRNA levels of MMP-7, TIMP-1/-3 (TIMP-2/-4 remained relatively low) and ADAM17, but augmented mRNA levels of MMP-11/-16/-23 and of many ECM genes. Immunohistology showed increased staining of MMP-2 in acute rejection (AR). Overall MMP activity was augmented in both transplanted groups, but CsA reduced MMP-9 activity and MMP-14 production. Taken together, MMP and TIMP were upregulated during acute AR. CsA ameliorated histology of rejection but showed potential pro-fibrotic effects. Thus, MMP and TIMP may play a role in acute cardiac allograft rejection, and beneficial modification of the MMP-ECM balance requires interventions beyond CsA.


Subject(s)
Cyclosporine/administration & dosage , Graft Rejection/metabolism , Heart Transplantation/physiology , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Animals , Extracellular Matrix/metabolism , Gene Expression Profiling , Heart Transplantation/immunology , Models, Animal , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Rats, Inbred Strains , Transplantation, Homologous , Up-Regulation/physiology
2.
Eur J Endocrinol ; 153(3): R7-R10, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16131595

ABSTRACT

OBJECTIVE: Adrenocorticotropic hormone (ACTH)-dependent Cushing's syndrome is biochemically characterized by increased plasma concentrations of ACTH inducing hypersecretion of cortisol. Somatostatin is known to inhibit ACTH secretion, and in vitro data have shown the inhibition of ACTH secretion by agonists activating sst2 and sst5 receptors. The present study aimed to determine the inhibitory effect of the multireceptor ligand SOM230, compared with the sst2-preferring agonist octreotide, on corticotropin-releasing hormone (CRH)-stimulated secretion of ACTH and corticosterone in rats. METHODS: Secretion of ACTH and corticosterone was induced by i.v. application of CRH (0.5 microg/kg) in rats pretreated 1 h before by i.v. application of SOM230 (1, 3, or 10 microg/kg), octreotide (10 microg/kg) or NaCl 0.9%. RESULTS: SOM230 (3 and 10 microg/kg) inhibited CRH-induced ACTH release by 45+/-3% and 51+/-2%, respectively, and corticosterone release by 43+/-5% and 27+/-16%, respectively. 10 microg/kg of octreotide tended to be less potent at inhibiting ACTH release (34+/-6% inhibition) and did not alter the secretion of corticosterone. CONCLUSION: SOM230 has a stronger inhibitory effect on ACTH and corticosterone secretion than octreotide in rats. This difference can be explained by its higher affinity to sst1, sst3 and especially sst5 receptors compared with octreotide.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Corticosterone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Somatostatin/analogs & derivatives , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Male , Octreotide/pharmacology , Rats , Rats, Sprague-Dawley , Somatostatin/pharmacology
4.
Eur J Endocrinol ; 146(5): 707-16, 2002 May.
Article in English | MEDLINE | ID: mdl-11980628

ABSTRACT

OBJECTIVE: The aim of the present study was to identify a small, metabolically stable somatotropin release inhibiting factor (SRIF) analog with a more universal binding profile similar to that of natural somatostatin, resulting in improved pharmacological properties and hence new therapeutic uses. DESIGN: A rational drug design approach was followed by synthesizing alanine-substituted SRIF-14 analogs to determine the importance of single amino acids in SRIF-14 for SRIF receptor subtype binding. The incorporation of structural elements of SRIF-14 in a stable cyclohexapeptide template in the form of modified unnatural amino acids resulted in the identification of the novel cyclohexapeptide SOM230. RESULTS: SOM230 binds with high affinity to SRIF receptor subtypes sst1, sst2, sst3 and sst5 and displays a 30- to 40-fold higher affinity for sst1 and sst5 than Sandostatin (octreotide; SMS 201-995) or Somatuline (BIM 23014). In vitro, SOM230 effectively inhibited the growth hormone releasing hormone (GHRH)-induced growth hormone (GH) release in primary cultures of rat pituitary cells with an IC(50) of 0.4+/-0.1 nmol/l (n=5). In vivo, SOM230 also potently suppressed GH secretion in rats. The ED(50) values determined at 1 h and 6 h post injection of SOM230 indicated its very long duration of action in vivo. This property was also reflected in pharmacokinetic studies comparing plasma levels of SMS 201-995 and SOM230 after subcutaneous application. Whereas SMS 201-995 had a terminal elimination half life of 2 h, this was markedly prolonged in SOM230-treated animals (t(1/2)=23 h). Furthermore, in rats SOM230 demonstrated a much higher efficacy in lowering plasma insulin-like growth factor-I (IGF-I) levels compared with SMS 201-995. The infusion of 10 microg/kg/h of SOM230 using subcutaneously implanted minipumps decreased plasma IGF-I levels far more effectively than SMS 201-995. After 126 days of continuous infusion of SOM230 plasma IGF-I levels were decreased by 75% of placebo-treated control animals. For comparison SMS 201-995, when used under the same experimental conditions, resulted in only a 28% reduction of plasma IGF-I levels, indicating a much higher efficacy for SOM230 in this animal model. It is important to note that the inhibitory effect of SOM230 was relatively selective for GH and IGF-I in that insulin and glucagon secretion was inhibited only at higher doses of SOM230. This lack of potent inhibition of insulin and glucagon release was also reflected in the lack of effect on plasma glucose levels. Even after high dose treatment over 126 days no obvious adverse side effects were noticed, including changes in plasma glucose levels. CONCLUSION: We have identified a novel short synthetic SRIF peptidomimetic, which exhibits high affinity binding to four of the five human SRIF receptor subtypes and has potent, long lasting inhibitory effects on GH and IGF-I release. Therefore SOM230 is a promising development candidate for effective GH and IGF-I inhibition and is currently under evaluation in phase 1 clinical trials.


Subject(s)
Human Growth Hormone/antagonists & inhibitors , Insulin-Like Growth Factor I/antagonists & inhibitors , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Somatostatin/pharmacology , Animals , Binding, Competitive , Dose-Response Relationship, Drug , Glucagon/antagonists & inhibitors , Humans , Insulin Antagonists/pharmacology , Male , Protein Isoforms/metabolism , Rats , Rats, Inbred Lew , Somatostatin/analogs & derivatives , Somatostatin/chemistry , Somatostatin/pharmacokinetics
5.
J Med Chem ; 44(12): 1986-92, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11384243

ABSTRACT

The prevention of xenograft rejection is substantially dependent on inhibiting antibodies (Ab) produced by B-cells independently of T-cell signals (TI-1). Due to their ubiquitous biochemical mechanisms of action, the immunosuppressants currently employed not only fail to discriminate between B- and T-cells but also have a narrow therapeutic window and, thus, their prolonged use in complex immunosuppressive regimens is problematic. By capitalizing on the target enzyme-bound (DHODH) structure 1b of one of these compounds, leflunomide, and modulating part of its multiple mechanisms of action to gain selectivity, the quinoline-8-carboxamide 3 was designed as a potentially weak enzyme inhibitor but effective immunosuppressant. Compound 3 fulfilled the mechanistic criteria set and had 10-fold B-cell over T-cell selectivity. Its pyridyl analogue 4 was found to be a highly potent and selective B-cell immunosuppressant with a 75-fold selectivity for B- over T-cells (as judged by the MLR data) and no general cytotoxicity at concentrations up to 160-fold higher than those required to inhibit B-cells. In the mouse, 4 effectively blocked TI-1 Ab production and suppressed Ab-mediated xenograft rejection in a xenotransplantation model under a once-daily dosing regimen, with efficacy down to 0.3 mg/kg/day po. These are the first data demonstrating the feasibility of the development of drugs specific for impeding Ab production.


Subject(s)
B-Lymphocytes/drug effects , Graft Rejection/prevention & control , Immunosuppressive Agents/chemical synthesis , Quinolines/chemical synthesis , Transplantation, Heterologous/immunology , Animals , Antibody Formation/drug effects , B-Lymphocytes/immunology , Cricetinae , Drug Design , Graft Survival/drug effects , Heart Transplantation/immunology , Immunosuppressive Agents/blood , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Lipopolysaccharides/pharmacology , Mesocricetus , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Quinolines/chemistry , Quinolines/pharmacokinetics , Quinolines/pharmacology , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
6.
J Immunol Methods ; 254(1-2): 1-12, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11406149

ABSTRACT

The aim of this study was to develop a new quantitative method for measuring in vitro the effects of T-cell immunosuppressive drugs by flow cytometry. Rat whole blood samples were stimulated with the T-cell mitogen succinylated concanavalin A in the presence or absence of different drugs. After 3 days, the expression of CD25 and CD8alpha in mitogen-stimulated CD4(+) cells increased 10- to 20-fold as measured by flow cytometry. Drug efficacy and potency was calculated based on dose-response curves of the drug-mediated decrease in CD4(+)/CD8alpha(+)/CD25(+) cells. The expression of CD8alpha in mitogen-stimulated CD4(+) cells was blocked completely by calcineurin inhibitors (cyclosporine A and FK-506), and partially by rapamycin and SDZ-RAD. The IC(50) (50% inhibitory concentration) values obtained were (mean+/-S.E.): 99.5+/-16.6 nM for cyclosporine A, 10.4+/-1.3 nM for FK-506, 1.8+/-0.7 nM for rapamycin, and 6.4+/-1.1 nM for SDZ-RAD. Our results show, for the first time, that CD8alpha, used as an activation antigen, is a sensitive marker for monitoring T-cell immunosuppression.


Subject(s)
CD4 Antigens/biosynthesis , CD4-Positive T-Lymphocytes/drug effects , CD8 Antigens/biosynthesis , Immunosuppressive Agents/pharmacology , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , Concanavalin A/pharmacology , Cyclosporine/pharmacology , Everolimus , Fingolimod Hydrochloride , Flow Cytometry/methods , Guanidines/pharmacology , Mitogens/pharmacology , Propylene Glycols/pharmacology , Rats , Rats, Inbred Lew , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sphingosine/analogs & derivatives , Tacrolimus/pharmacology
7.
Eur J Endocrinol ; 143 Suppl 1: S3-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11068933

ABSTRACT

Graft vessel disease (GVD) is a major cause of graft loss after the first year following transplantation. GVD is a complex, multifunctional process that involves immunological as well as non-immunological events such as ischaemia/reperfusion injury. An important target cell to interfere with the development of GVD is the smooth muscle cell (SMC). Somatostatin (SRIF) analogues have been shown previously to inhibit the proliferation of SMC in vitro and in vivo. We provide evidence that Sandostatin, an octapeptide SRIF analogue that is known to have anti-proliferative properties on SMC proliferation, inhibits vascular remodelling in a rat angioplasty model. Furthermore, in two allotransplantation models, Sandostatin effectively interferes with the development of signs of chronic rejection/GVD. The role of the different SRIF receptor subtypes in chronic graft rejection is currently under investigation.


Subject(s)
Muscle Development , Muscle, Smooth, Vascular/growth & development , Octreotide/therapeutic use , Receptors, Somatostatin/physiology , Animals , Carotid Arteries/drug effects , Carotid Arteries/transplantation , Cell Division , Disease Models, Animal , Graft Rejection/prevention & control , Kidney/blood supply , Kidney/drug effects , Kidney Transplantation , Mice , Muscle, Smooth, Vascular/drug effects , Octreotide/metabolism , Rats , Receptors, Somatostatin/metabolism
8.
Eur J Pharmacol ; 391(1-2): 31-8, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10720632

ABSTRACT

Increased levels of nitric oxide (NO) are found in rejecting renal allografts. Inducible NO synthase (iNOS) in infiltrating monocytes/macrophages could lead to NO bursts. NO may modulate the inflammatory response of early rejection due to its high reactivity with superoxide to yield peroxynitrite. To define the role of iNOS in acute renal allograft, rejection effects of the specific iNOS blockers iminoethyl-lysine and 7-butylhexahydro-1H-azepin-2-imine, monohydrochloride on renal function and morphology were investigated in renal allografts. Lewis rats received Brown Norway grafts with one kidney left in situ. All recipients were treated with low dose cyclosporine-A (2.5 mg/kg BW/day s.c.) to allow moderate rejection. In addition, one group received iminoethyl-lysine (10 mg/kg BW/day gavage) and one group received butylhexahydro-azepin-imine (3.4 mg/kg BW/day i.p.). Sham operated Brown Norway donor rats served as baseline controls. Compared to controls, low dose cyclosporine-A decreased glomerular filtration rate (P<0.05) and numerically increased renal vascular resistance. Adding iminoethyl-lysine to cyclosporine-A improved renal hemodynamics. Adding butylhexahydro-azepin-imine to cyclosporine-A practically restored glomerular filtration rate and renal vascular resistance (P<0.05) to control levels. Grafts treated with cyclosporine-A alone showed vascular, glomerular and tubulointerstitial lesions. Adding iminoethyl-lysine or butylhexahydro-azepin-imine to cyclosporine-A did not significantly reduce vascular and glomerular injury, but diminished tubulointerstitial injury as well as nitrotyrosine staining in tubular epithelium (P<0.05). Thus, adding the iNOS blockers iminoethyl-lysine or butylhexahydro-azepin-imine to cyclosporine-A improved graft function and reduced tubulointerstitial lesions.


Subject(s)
Azepines/pharmacology , Enzyme Inhibitors/pharmacology , Graft Rejection/prevention & control , Graft Survival/drug effects , Imines/pharmacology , Kidney Transplantation/physiology , Kidney Tubules/pathology , Kidney/drug effects , Lysine/analogs & derivatives , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Cyclosporine/pharmacology , Graft Rejection/pathology , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Kidney/pathology , Kidney Function Tests , Lysine/pharmacology , Male , Nitric Oxide Synthase Type II , Rats , Rats, Inbred BN , Rats, Inbred Lew
9.
Ital J Gastroenterol Hepatol ; 31 Suppl 2: S224-6, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10604136

ABSTRACT

Among various newly synthesized chelator-linked octreotide analogues 90Y-[DOTA-DPhe1, Thyr3]-octreotide (90Y-SMT 487) was finally selected for clinical development. In vitro, SMT 487 binds selectively with nanomolar affinity to the somatostatin receptor subtype 2 (IC30 = 0.39 nM +/- 0.02). In vivo, 90Y-[DOTA-DPhe1, Thyr3]-octreotide shows a rapid blood clearance (T1/2 alpha < 5 min) and high accumulation in somatostatin subtype 2 receptor expressing tumours. The in vivo administration of 90Y-[DOTA-DPhe1, Thyr3]-octreotide induces a rapid tumour shrinkage in three different somatostatin receptor positive tumour models: CA20948 rat pancreatic tumours grown in normal rats, AR42J rat pancreatic tumours and NCI-H69 human small cell lung cancer both grown in nude mice. The radiotherapeutic efficacy of 90Y-SMT 487 was enhanced in combination with standard anticancer drugs, such as mitomycin C, which resulted in a tumour decrease of 70% of the initial volume. In the CA 20948 syngeneic rat tumour model, a single treatment with 10 microCi/kg 90Y-SMT 487 resulted in the disappearance of 5 out of 7 tumours. Thus the new radiotherapeutic agent showed its curative potential for the selective treatment of SRIF receptor-expression tumours. Clinical Phase I studies with 90Y-SMT 487 were started in September 1997.


Subject(s)
Neuroendocrine Tumors/radiotherapy , Octreotide/therapeutic use , Receptors, Somatostatin/analysis , Animals , Disease Models, Animal , Humans , Mice , Octreotide/analogs & derivatives , Octreotide/pharmacokinetics , Rats , Receptors, Somatostatin/metabolism , Sensitivity and Specificity , Tissue Distribution
12.
Eur J Nucl Med ; 25(7): 668-74, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9662587

ABSTRACT

Somatostatin receptor-expressing tumours are potential targets for therapy with radiolabelled somatostatin analogues. We have synthesized a number of such analogues in the past and identified [DOTA-dPhe1, Tyr3]octreotide (SMT 487) as the most promising candidate molecule because of its advantageous properties in cellular and in vivo tumour models. In the current paper we describe the radiotherapeutic effect of yttrium-90 labelled SMT 487 in Lewis rats bearing the somatostatin receptor-positive rat pancreatic tumour CA 20948. SMT 487 binds with nanomolar affinity to both the human and the rat somatostatin receptor subtype 2 (sst2) (human sst2 IC50=0.9 nM, rat sst2 IC50=0.5 nM). In vivo, 90Y-SMT 487 distributed rapidly to the sst2 expressing CA 20948 rat pancreatic tumour, with a tumour-to-blood ratio of 49.15 at 24 h post injection. A single intravenous administration of 10 mCi/kg 90Y-SMT 487 resulted in a complete remission of the tumours in five out of seven CA 20948 tumour-bearing Lewis rats. No regrowth of the tumours occurred 8 months post injection. Control animals that were treated with 30 microg/kg of unlabelled SMT 487 had to be sacrificed 10 days post injection due to excessive growth or necrotic areas on the tumour surface. Upon re-inoculation of tumour cells into those rats that had shown complete remission, the tumours disappeared after 3-4 weeks of moderate growth without any further treatment. The present study shows for the first time the curative potential of 90Y-SMT 487-based radiotherapy for somatostatin receptor-expressing tumours. Clinical phase I studies with yttrium-labelled SMT 487 have started in September 1997.


Subject(s)
Octreotide/analogs & derivatives , Pancreatic Neoplasms/radiotherapy , Radiopharmaceuticals/therapeutic use , Receptors, Somatostatin/metabolism , Yttrium Radioisotopes/therapeutic use , Animals , Humans , Octreotide/therapeutic use , Pancreatic Neoplasms/metabolism , Polymerase Chain Reaction , Rats , Rats, Inbred Lew , Tissue Distribution
15.
Cancer Res ; 57(5): 956-62, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9041201

ABSTRACT

Among the five cloned somatostatin receptor subtypes (sst1 to sst5), sst2 mediates the antiproliferative effect of somatostatin analogues in vitro. Somatostatin analogues have been shown to inhibit cell growth in vitro and in vivo in pancreatic cancer models that expressed sst2. We recently demonstrated the loss of sst2 gene expression in human pancreatic adenocarcinomas and most of the derived pancreatic cancer cell lines. In the present study, we corrected the sst2 defect in human pancreatic cancer BxPC-3 and Capan-1 cells by stable transfection with human sst2 cDNA. In the absence of exogenous ligand, both BxPC-3 and Capan-1 cells expressing sst2 showed a significant reduction in cell growth. This inhibitory effect was blocked by treatment with antiserum to somatostatin. sst2-expressing cells produced somatostatin-like immunoreactivity that mainly corresponded to somatostatin 14, indicating the induction of a negative autocrine loop. In other respects, sst2 expression in Capan-1 cells induced a significant reduction of clonogenicity in soft agar. Moreover, a significantly reduced (Capan-1 cells) or suppressed (BxPC-3 cells) tumor growth in athymic nude mice was observed. The reversal of tumorigenicity induced by the restoration of sst2 expression suggests that the loss of sst2 contributes to the malignancy of human pancreatic cancers.


Subject(s)
Receptors, Somatostatin/physiology , Animals , Cell Adhesion , Cell Division , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Somatostatin/physiology , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
16.
Yale J Biol Med ; 70(5-6): 549-54, 1997.
Article in English | MEDLINE | ID: mdl-9825482

ABSTRACT

Analogs of somatostatin (SRIF) such as octreotide exert antiproliferative effects that are mediated directly by tumoral SRIF receptors or indirectly by down-modulation of factors that stimulate tumor growth. Direct and indirect antiproliferative effects have been demonstrated in certain SRIF receptor-positive and -negative human breast cancer models in nude mice, respectively. These antiproliferative mechanisms are also being explored in other cancer types including pancreatic cancer. While clinical pilot studies have indicated that a fraction of pancreatic adenocarcinomas respond to high-dose octreotide treatment, it is known from receptor autoradiographic and scintigraphic studies that human pancreatic carcinomas fail to express SRIF receptors, in contrast to rat pancreatic carcinomas or human endocrine pancreatic cancer. Studies on the potential anticancer effect of octreotide on the growth of experimental human pancreatic cancer and its SRIF receptor status have been controversial. Therefore, we investigated in vivo the effects of octreotide on the growth of MIA PaCa-2 human pancreatic carcinomas raised from cultured cells with a low passage number after receipt from the American Type Culture Collection. Nude mice bearing MIA PaCa-2 tumors were treated with a single injection of the recently developed octreotide long-acting release formulation, "SMS pa LAR." This treatment was well tolerated and resulted in a highly significant inhibition of tumor growth during weeks three and eight after administration. MIA PaCa-2 tumors were removed after eight weeks and processed for RT-PCR analysis using probes specific for each of the five somatostatin receptor subtypes sst1-sst5. This analysis revealed that MIA PaCa-2 tumors, like human pancreatic adenocarcinomas, do not express any of the five SRIF receptor subtypes, suggesting an indirect mode of tumor growth inhibition. In summary, the depot formulation SMS pa LAR exerted long-lasting antiproliferative effects in SRIF receptor-negative human pancreatic carcinomas in nude mice.


Subject(s)
Carcinoma/drug therapy , Neoplasms, Experimental/drug therapy , Octreotide/pharmacology , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Hormonal/pharmacology , Carcinoma/metabolism , Cell Division/drug effects , Humans , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Metabolism ; 45(8 Suppl 1): 17-20, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8769372

ABSTRACT

In the past few years, five different somatostatin (SRIF) receptor subtypes (sst1.5) have been identified, which form a distinct group in the superfamily of G-protein-coupled receptors. The naturally occurring somatostatins SRIF-28, SRIF-25, and SRIF-14 all reveal high-affinity binding for sst1.5. In contrast, short synthetic analogs that are in clinical use, such as SMS 201-995, RC-160, or BIM 23014, primarily interact with the sst2 subtype. Some SRIF analogs were previously reported to be selective for one SRIF receptor subtype, eg, the sst2 (MK 678), the sst3 (BIM 23056), or the sst5 (BIM 23052, L362-855) subtype. However, when we studied the binding affinities of these SRIF analogs for human (h) sst1.5 expressed in either CHO or COS-1 cells, we were unable to confirm these previously reported selectivities. The absence of sst antagonists is a major drawback for investigating the functional role of each sst subtype. We used site-directed mutagenesis to identify amino acids that determine ligand specificity for sst2. A single Ser305 to Phe mutation in TM VII increased the affinity of hsst1, for SMS 201-995 nearly 100-fold, and when Gln291 was also exchanged to Asn in TM VII of hsst1, almost full sst2-like binding of SMS 201-995 was obtained. These data may aid in the design and synthesis of new selective type sst ligands. We have identified the expression of sst subtypes in nonclassical SRIF target tissue such as the lung. The pKi values for SRIF and various SRIF analogs in rat lung tissue preparations were in close correlation with those obtained for CHO cells expressing the sst4 subtype. Furthermore, reverse transcriptase polymerase chain reaction (RT-PCR) experiments revealed the predominant expression of mRNA specific for sst4 in mouse, rat, and human lung tissue, confirmed by autoradiographies of rat lung. No specific binding for [125I]Tyr3-SMS 201-995 was detected, since SMS 201-995 has low affinity for sst4. In contrast, specific binding of [125I]SRIF-28 to rat lung sections was demonstrated, which could be displaced by unlabelled SRIF-14 and SRIF-28, indicating specific, high affinity binding of this radioligand to sst4 receptors.


Subject(s)
Receptors, Somatostatin/metabolism , Amino Acid Sequence , Animals , Humans , Ligands , Lung/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , RNA, Messenger/metabolism , Rats , Receptors, Somatostatin/genetics
19.
Digestion ; 57 Suppl 1: 17-21, 1996.
Article in English | MEDLINE | ID: mdl-8813461

ABSTRACT

The aim of the present study was to selectively target a beta-emitter-labelled octreotide analogue to somatostatin (SRIF)-receptor-expressing tumours and to evaluate the feasibility of SRIF-receptor-mediated radiotherapy by delivering a lethal dose of radiation to the tumour. The most promising compound in a series of DTPA-coupled octreotide analogues was DTPA-benzyl-acetamido-D-Phe1, Tyr3-octreotide (SDZ413). In vitro, SDZ413 binds with nanomolar affinity to SRIF-receptors (IC50 = 4.0 nM) and inhibits growth hormone release from primary cultures of rat pituitary cells with an IC50 of 7.2 nM. Biodistribution studies with [90Y]SDZ413 demonstrated a fast and significant SRIF-receptor-specific accumulation of the labelled conjugate (tumour/muscle ratio after 24 h: 52/1). [90Y]SDZ413 was effective in the radiotherapy of SRIF-receptor-positive tumours in a nude mouse model. A single treatment with [90Y]SDZ413 led to a significant decrease (25%) of tumour mass. This effect was mediated by the intact radioligand, since treatment with [90Y]SDZ978, a derivative of SDZ413 which does not bind with high affinity to SRIF-receptors or with the unlabelled SDZ413 alone, failed to affect tumour growth. These results suggest that receptor-targeted radiotherapy with a 90Y-labelled octreotide analogue represents a new strategy for the treatment of SRIF-receptor-positive tumours that have been previously diagnosed with OctreoScan111 (pentetreotide).


Subject(s)
Hormone Antagonists/therapeutic use , Neoplasms, Experimental/radiotherapy , Receptors, Somatostatin/physiology , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Animals , Chelating Agents/pharmacology , Hormone Antagonists/metabolism , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Octreotide/analogs & derivatives , Octreotide/chemical synthesis , Octreotide/pharmacology , Pentetic Acid/analogs & derivatives , Pentetic Acid/chemical synthesis , Pentetic Acid/pharmacology , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Rats , Somatostatin/metabolism , Tissue Distribution , Yttrium Radioisotopes
20.
Digestion ; 57 Suppl 1: 22-8, 1996.
Article in English | MEDLINE | ID: mdl-8813462

ABSTRACT

The somatostatin analogue octreotide (SMS 201-995) exerts potent anti-proliferative effects in a number of experimental cancer models. Here we report on the inhibitory effect of octreotide in combination with the chemotherapeutic agents mitomycin C, doxorubicin, 5-fluorouracil, or taxol on the growth of AR42J pancreatic cancer cells in vitro. The dose-dependent anti-proliferative effects of mitomycin C, doxorubicin and taxol were synergistically enhanced by octreotide. Combinations of octreotide and 5-fluorouracil resulted either in additive or, at high concentrations of the chemotherapeutic agent, in synergistic interactions. Combined treatment with doxorubicin and octreotide was also studied for time dependency and potential efficacy in tumour-bearing animals. Pretreatment (24 h) with doxorubicin resulted in clear synergy. However, pretreatment with octreotide 24 h prior to addition of doxorubicin resulted only in an additive interaction. It was shown in AR42J-tumour-bearing nude mice that the combination of doxorubicin and octreotide was well tolerated. Tumour growth was inhibited to 9% of controls, compared with 44% in the doxorubicin alone arm (day 14 of treatment). Our in vitro and in vivo interaction studies suggest that octreotide potentiates the effect of various chemotherapeutic agents in a synergistic or additive manner.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents/pharmacology , Octreotide/pharmacology , Pancreatic Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Autoradiography , Cell Division/drug effects , Doxorubicin/pharmacology , Drug Synergism , Fluorouracil/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mitomycin/pharmacology , Neoplasm Transplantation , Paclitaxel/pharmacology , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...