51
|
Lacerda S, Campello MP, Marques F, Gano L, Kubíček V, Fousková P, Tóth É, Santos I. A novel tetraazamacrocycle bearing a thiol pendant arm for labeling biomolecules with radiolanthanides. Dalton Trans 2009:4509-18. [DOI: 10.1039/b820375j] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
52
|
Feytens D, De Vlaeminck M, Cescato R, Tourwé D, Reubi JC. Highly Potent 4-Amino-indolo[2,3-c]azepin-3-one-Containing Somatostatin Mimetics with a Range of sst Receptor Selectivities. J Med Chem 2008; 52:95-104. [DOI: 10.1021/jm801205x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Debby Feytens
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Magali De Vlaeminck
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Renzo Cescato
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Dirk Tourwé
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Jean Claude Reubi
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| |
Collapse
|
53
|
Stronge RL, Turner GB, Johnston BT, McCance DR, McGinty A, Patterson CC, Ardill JES. A rapid rise in circulating pancreastatin in response to somatostatin analogue therapy is associated with poor survival in patients with neuroendocrine tumours. Ann Clin Biochem 2008; 45:560-6. [DOI: 10.1258/acb.2008.008033] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aim To assess the value of pancreastatin as a predictive factor for identifying patients with neuroendocrine tumours (NETs) who respond poorly to somatostatin analogues. Methods A retrospective study of patients with NETs. Patient records from the Northern Ireland Neuroendocrine Tumour Register were interrogated. Those who had pancreastatin concentrations measured on two or more occasions, before and during somatostatin analogue therapy (within the set time-limits) were selected. Data relating to diagnosis, surgery, somatostatin analogue therapy and survival outcome were noted. Data were subjected to univariate and multivariate analysis using Cox proportional hazard model. Results Fifty-nine patients with gastroenteropancreatic NETs fulfilled the inclusion criteria. Factors associated with a poor survival outcome on univariate analysis were primary tumour site ( P = 0.006) and rapid rise in pancreastatin during somatostatin analogue treatment ( P < 0.001). In multivariate analysis, highly significant clinical prognostic indicators were: tumour location ( P < 0.001), pre-treatment pancreastatin ( P < 0.001) and pancreastatin change ( P < 0.001). Conclusions This study endorses the finding that pancreastatin is a useful prognostic indicator of neuroendocrine disease. On commencement of treatment, one-third of the subjects showed an immediate negative pancreastatin response to somatostatin analogues, which was associated with poor survival. This is the first study to document such an association. These findings have significant therapeutic consequences. In the presence of a rapidly rising pancreastatin alternative, treatment modalities should be sought.
Collapse
Affiliation(s)
- R L Stronge
- St George's Hospital Medical School, University of London
| | | | - B T Johnston
- Neuroendocrine Tumour Group, Royal Victoria Hospital
| | - D R McCance
- Neuroendocrine Tumour Group, Royal Victoria Hospital
| | | | | | - J E S Ardill
- Neuroendocrine Tumour Group, Royal Victoria Hospital
| |
Collapse
|
54
|
Pyronnet S, Bousquet C, Najib S, Azar R, Laklai H, Susini C. Antitumor effects of somatostatin. Mol Cell Endocrinol 2008; 286:230-7. [PMID: 18359151 DOI: 10.1016/j.mce.2008.02.002] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 01/15/2008] [Accepted: 02/02/2008] [Indexed: 02/07/2023]
Abstract
Since its discovery three decades ago as an inhibitor of GH release from the pituitary gland, somatostatin has attracted much attention because of its functional role in the regulation of a wide variety of physiological functions in the brain, pituitary, pancreas, gastrointestinal tract, adrenals, thyroid, kidney and immune system. In addition to its negative role in the control of endocrine and exocrine secretions, somatostatin and analogs also exert inhibitory effects on the proliferation and survival of normal and tumor cells. Over the past 15 years, studies have begun to reveal some of the molecular mechanisms underlying the antitumor activity of somatostatin. This review covers the present knowledge in the antitumor effect of somatostatin and analogs and discusses the perspectives of novel clinical strategies based on somatostatin receptor sst2 gene transfer therapy.
Collapse
Affiliation(s)
- Stéphane Pyronnet
- INSERM U858, Institut de Médecine Moléculaire de Rangueil, Dpt Cancer/E16, CHU Rangueil, Toulouse Cedex 4, France
| | | | | | | | | | | |
Collapse
|
55
|
|
56
|
Bapst JP, Froidevaux S, Calame M, Tanner H, Eberle AN. Dimeric DOTA-alpha-melanocyte-stimulating hormone analogs: synthesis and in vivo characteristics of radiopeptides with high in vitro activity. J Recept Signal Transduct Res 2008; 27:383-409. [PMID: 18097939 DOI: 10.1080/10799890701723528] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Dimeric analogs of alpha-melanocyte-stimulating hormone (alpha-MSH) labeled with radiometals are potential candidates for diagnosis and therapy of melanoma by receptor-mediated tumor targeting. Both melanotic and amelanotic melanomas (over-)express the melanocortin-1 receptor (MC1-R), the target for alpha-MSH. In the past, dimerized MSH analogs have been shown to display increased receptor affinity compared to monomeric MSH, offering the possibility of improving the ratio between specific uptake of radiolabeled alpha-MSH by melanoma and nonspecific uptake by the kidneys. We have designed three linear dimeric analogs containing a slightly modified MSH hexapeptide core sequence (Nle-Asp-His-d-Phe-Arg-Trp) in parallel or antiparallel orientation, a short spacer, and the DOTA chelator for incorporation of the radiometal. In vitro, all three peptides were more potent ligands of the mouse B16-F1 melanoma cell melanocortin-1 receptor (MC1-R) than DOTA-NAPamide, which served as standard. The binding activity of DOTA-diHexa(NC-NC)-amide was 1.75-fold higher, that of diHexa(NC-NC)-Gly-Lys(DOTA)-amide was 3.37-fold higher, and that of DOTA-diHexa(CN-NC)-amide was 2.34-fold higher. Using human HBL melanoma cells, the binding activity of diHexa(NC-NC)-Gly-Lys(DOTA)-amide was sixfold higher than that of DOTA-NAPamide. Uptake by cultured B16-F1 cells was rapid and almost quantitative. In vivo, however, the data were less promising: tumor-to-kidney ratios 4 hr postinjection were 0.11 for [(111)In]DOTA-diHexa(NC-NC)-amide, 0.26 for diHexa(NC-NC)-Gly-Lys([(111)In]DOTA)-amide, and 0.36 for [(111)In]DOTA-diHexa(CN-NC)-amide, compared to 1.67 for [(111)In]DOTA-NAPamide. It appears that despite the higher affinity to the MC1-R of the peptide dimers and their excellent internalization in vitro, the uptake by melanoma tumors in vivo was lower, possibly because of reduced tissue penetration. More striking, however, was the marked increase of kidney uptake of the dimers, explaining the unfavorable ratios. In conclusion, although radiolabeled alpha-MSH dimer peptides display excellent receptor affinity and internalization, they are no alternative to the monomeric DOTA-NAPamide for in vivo application.
Collapse
Affiliation(s)
- Jean-Philippe Bapst
- Laboratory of Endocrinology, Department of Research, University Hospital and University Children's Hospital, Basel, Switzerland
| | | | | | | | | |
Collapse
|
57
|
Tanaka K, Fukase K. PET (positron emission tomography) imaging of biomolecules using metal-DOTA complexes: a new collaborative challenge by chemists, biologists, and physicians for future diagnostics and exploration of in vivo dynamics. Org Biomol Chem 2008; 6:815-28. [PMID: 18292870 DOI: 10.1039/b718157b] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Recently, PET has been paid a great deal of attention as a non-invasive imaging method. In this review, the recent advances of PET using biomolecules, such as peptides, monoclonal antibodies, proteins, oligonucleotides, and glycoproteins will be described. So far, PET of biomolecules has been mainly used for diagnosis of cancers. The biomolecules have been conjugated with the DOTA ligand, labeled with radiometals as the beta+ emitter, and targeted to specific tumors, where they have enabled visualization of even small metastatic lesions, due to the high sensitivity of the PET scanners. Some of the biomolecules have been used not only for PET diagnosis, but also for radiotherapeutic treatments by simply changing the radiometals to beta(-) emitters. Collaborative work between chemists, biologists, and physicians will be important for the future of biomolecule-based targeting and diagnosis.
Collapse
Affiliation(s)
- Katsunori Tanaka
- Department of Chemistry, Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan.
| | | |
Collapse
|
58
|
Torchilin V. Intracellular delivery of protein and peptide therapeutics. DRUG DISCOVERY TODAY. TECHNOLOGIES 2008; 5:e95-e103. [PMID: 24981097 DOI: 10.1016/j.ddtec.2009.01.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Many proteins and peptides are used as highly specific and effective therapeutic agents. Their use is, however, complicated by their instability and side effects. Because many protein and peptide drugs have their therapeutic targets inside cells, there is also an important task to bring these drugs into target cells without subjecting them to the lysosomal degradation. This review describes current approaches to the intracellular delivery of protein and peptide drugs. Various drug delivery systems and methods are considered allowing for safe and effective transport of protein and peptide drugs into the cell cytoplasm.:
Collapse
Affiliation(s)
- Vladimir Torchilin
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
59
|
Petrou C, Magafa V, Nikolopoulou A, Pairas G, Nock B, Maina T, Cordopatis P. Synthesis and sst2 binding profiles of new [Tyr3]octreotate analogs. J Pept Sci 2008; 14:725-30. [DOI: 10.1002/psc.989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
60
|
Acar Ç, Teksöz S, Ünak P, Müftüler FZB, Medine EI. Somatostatin with 99mTc and Biodistribution Studies in Rats. Cancer Biother Radiopharm 2007; 22:748-54. [DOI: 10.1089/cbr.2007.0340] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Çigdem Acar
- Department of Nuclear Applications, Institute of Nuclear Sciences, Ege University, Bornova Izmir, Turkey
| | - Serap Teksöz
- Department of Nuclear Applications, Institute of Nuclear Sciences, Ege University, Bornova Izmir, Turkey
| | - Perihan Ünak
- Department of Nuclear Applications, Institute of Nuclear Sciences, Ege University, Bornova Izmir, Turkey
| | - F. Zümrüt Biber Müftüler
- Department of Nuclear Applications, Institute of Nuclear Sciences, Ege University, Bornova Izmir, Turkey
| | - E. Ilker Medine
- Department of Nuclear Applications, Institute of Nuclear Sciences, Ege University, Bornova Izmir, Turkey
| |
Collapse
|
61
|
Knör S, Modlinger A, Poethko T, Schottelius M, Wester HJ, Kessler H. Synthesis of novel 1,4,7,10-tetraazacyclodecane-1,4,7,10-tetraacetic acid (DOTA) derivatives for chemoselective attachment to unprotected polyfunctionalized compounds. Chemistry 2007; 13:6082-90. [PMID: 17503419 DOI: 10.1002/chem.200700231] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A convenient synthesis of novel bifunctional poly(amino carboxylate) chelating agents allowing chemoselective attachment to highly functionalized biomolecules is described. Based on the well known chelator 1,4,7,10-tetraazacyclodecane-1,4,7,10-tetraacetic acid (DOTA), we synthesized novel bifunctional chelating agents bearing additional functional groups by alkylating 1,4,7,10-tetraazacyclododecane (cyclen) with one equivalent of para-functionalized alkyl 2-bromophenyl-acetate and three equivalents of tert-butyl 2-bromoacetate. The resulting compounds, which contain an additional carbonyl or alkyne functionality, allow site specific labeling of appropriately functionalized unprotected biomolecules in a rapid manner via click reactions. This was demonstrated by the attachment of our new DOTA derivatives to the somatostatin analogue Tyr3-octreotate by chemoselective oxime ligation and CuI-catalyzed azide-alkyne cycloaddition. Initial biodistribution studies in mice with the radiometalated compound demonstrated the applicability of the described DOTA conjugation.
Collapse
Affiliation(s)
- Sebastian Knör
- Department Chemie, Lehrstuhl für Organische Chemie II, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany
| | | | | | | | | | | |
Collapse
|
62
|
Falciani C, Fabbrini M, Pini A, Lozzi L, Lelli B, Pileri S, Brunetti J, Bindi S, Scali S, Bracci L. Synthesis and biological activity of stable branched neurotensin peptides for tumor targeting. Mol Cancer Ther 2007; 6:2441-8. [PMID: 17766836 DOI: 10.1158/1535-7163.mct-07-0164] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Receptors for endogenous regulatory peptides, like the neuropeptide neurotensin, are overexpressed in several human cancers and can be targets for peptide-mediated tumor-selective therapy. Peptides, however, have the main drawback of an extremely short half-life in vivo. We showed that neurotensin and other endogenous peptides, when synthesized as dendrimers, retain biological activity and become resistant to proteolysis. Here, we synthesized the neurotensin functional fragment NT(8-13) in a tetrabranched form linked to different units for tumor therapy or diagnosis. Fluorescent molecules were used to monitor receptor binding and internalization in HT29 human adenocarcinoma cells and receptor binding in HT29 tumor xenografts in nude mice. Linking of chemotherapic molecules like chlorin e6 and methotrexate to dendrimers resulted in a dramatic increase in drug selectivity, uptake of which by target cells became dependent on peptide receptor binding. When nude mice carrying human tumor xenografts were treated with branched NT(8-13)-methotrexate, a 60% reduction in tumor growth was observed with respect to mice treated with the free drug.
Collapse
Affiliation(s)
- Chiara Falciani
- Department of Molecular Biology, University of Siena, Siena, Italy 53100.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Nikiforovich GV, Marshall GR, Achilefu S. Molecular modeling suggests conformational scaffolds specifically targeting five subtypes of somatostatin receptors. Chem Biol Drug Des 2007; 69:163-9. [PMID: 17441902 DOI: 10.1111/j.1747-0285.2007.00493.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Several analogs of somatostatin with conformational constraints in their peptide backbones have been modeled to determine energetically feasible conformations. Comparison of low-energy backbone structures of these peptides suggested unique conformations of the central Phe/Ala(i)-D-Trp(i+1)-Lys(i+2)-Thr(i+3) fragment characteristic for specific interactions of somatostatin with each of the five distinct subtypes of somatostatin receptors (SSTRs). The conformations obtained were in good agreement with experimental data obtained earlier by NMR measurements and/or X-ray crystallography. The results help rationalize experimental observations on the specificity of binding of various somatostatin analogs with different subtypes of the SSTRs. They also serve as templates for the design of conformationally constrained non-peptide scaffolds that effectively and selectively interact with different subtypes of SSTRs. Such scaffolds can be convenient carriers of radiolabels and near-infrared labels in specific agents for imaging tumors expressing different SSTR subtypes.
Collapse
Affiliation(s)
- Gregory V Nikiforovich
- Department of Biochemistry and Molecular Biology, Washington University Medical School, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
64
|
Masyuk TV, Masyuk AI, Torres VE, Harris PC, Larusso NF. Octreotide inhibits hepatic cystogenesis in a rodent model of polycystic liver disease by reducing cholangiocyte adenosine 3',5'-cyclic monophosphate. Gastroenterology 2007; 132:1104-16. [PMID: 17383431 DOI: 10.1053/j.gastro.2006.12.039] [Citation(s) in RCA: 185] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 11/30/2007] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS In polycystic liver diseases (PCLDs), increased cholangiocyte proliferation and fluid secretion are key features and cholangiocyte adenosine 3',5'-cyclic monophosphate (cAMP) is an important regulator of these processes. Thus, we assessed cAMP levels and evaluated octreotide (an analogue of somatostatin known to inhibit cAMP) in hepatic cyst growth using an in vitro model of cystogenesis and an in vivo animal model of autosomal recessive polycystic kidney disease (ARPKD), one of the PCLDs. METHODS Expression of somatostatin receptors (SSTRs) was assessed by reverse-transcription polymerase chain reaction and confocal microscopy in cholangiocytes from normal and polycystic kidney (PCK) rats, the ARPKD model of autosomal recessive polycystic kidney disease. Effects of octreotide on cAMP levels and cyst expansion were studied in vitro using PCK bile ducts grown in 3-dimensional culture. The effects of octreotide on hepatic and renal cystogenesis were investigated in PCK rats in vivo. RESULTS In cholangiocytes and serum of PCK rats, cAMP concentrations were approximately 2 times higher than in normal rats. SSTR subtypes that bind octreotide (ie, SSTR2, SSTR3, and SSTR5) were expressed in both normal and PCK cholangiocytes. In vitro, octreotide inhibited cAMP levels by 35% and reduced cyst growth by 44%. In vivo, octreotide lowered cAMP content in cholangiocytes and serum by 32%-39% and inhibited hepatic disease progression, leading to 22%-60% reductions in liver weight, cyst volume, hepatic fibrosis, and mitotic indices. Similar effects were observed in kidneys of PCK rats. CONCLUSIONS This preclinical study provides a strong rationale for assessing the potential value of octreotide in the treatment of PCLDs.
Collapse
Affiliation(s)
- Tatyana V Masyuk
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
65
|
Kvols LK, Woltering EA. Role of somatostatin analogs in the clinical management of non-neuroendocrine solid tumors. Anticancer Drugs 2007; 17:601-8. [PMID: 16917205 DOI: 10.1097/01.cad.0000210335.95828.ed] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The somatostatin analogs octreotide, lanreotide and RC-160 (vapreotide) are known to have direct and indirect antitumor effects. Direct effects include the arrest of tumor growth and stimulation of apoptosis, resulting in tumor shrinkage. Indirect antiproliferative effects may occur through antiangiogenesis, immunomodulatory effects and the suppression of tumor-stimulating growth factors. With a safety profile of somatostatin analogs established over 20 years of clinical use in the treatment of neuroendocrine tumors, somatostatin analogs are attractive therapeutic options for patients with non-neuroendocrine tumors. In early clinical trials of somatostatin analogs, however, some cancer patients responded well, while others showed a lack of benefit. This variability in clinical response may reflect the selective binding affinities of octreotide, lanreotide and RC-160, which bind with high affinity to just two of the five different somatostatin receptor subtypes. Treatment response may therefore depend on the specific receptor subtype(s) present in the tumor, the relative proportion of receptor(s) expressed on the tumor cell surface and the absolute quantity of each receptor subtype. Greater understanding of the role of somatostatin receptors, their binding affinities and modes of action has led to increased research into the use of somatostatin analogs, particularly octreotide, in cancer treatment as monotherapies, in combination with hormonal treatments and cytotoxic therapies, and in both adjuvant and neoadjuvant settings. A review of the literature suggests that the antitumor potential of somatostatin analogs should be investigated further and additional studies might determine how these analogs can best be used to improve the treatment of patients with non-neuroendocrine tumors.
Collapse
Affiliation(s)
- Larry K Kvols
- Inter Disciplinary Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, USA.
| | | |
Collapse
|
66
|
Lu Y, Yang J, Sega E. Issues related to targeted delivery of proteins and peptides. AAPS JOURNAL 2006; 8:E466-78. [PMID: 17025264 PMCID: PMC2761053 DOI: 10.1208/aapsj080355] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
While modern genomic and proteomic technology enables rapid screening of novel proteins and peptides as potential drug candidates, design of delivery systems for these biologics remains challenging especially to achieve site-specific pharmacological actions. This article discusses the issues associated with targeted delivery of protein and peptide drugs at physiochemical, physiological, and intracellular levels with a special focus on cancer therapy.
Collapse
Affiliation(s)
- Yingjuan Lu
- Endocyte, Inc, 3000 Kent Avenue, Suite A1-100, West Lafayette, IN 47906, USA.
| | | | | |
Collapse
|
67
|
Abstract
BACKGROUND There is a need for novel antitumor agents that demonstrate efficacy in currently refractory tumors without adding to the toxicity of therapy. The somatostatin analogs, which have demonstrated antineoplastic activities in experimental tumor models, and good tolerability and safety profiles are attractive candidates. MATERIALS AND METHODS Data from preclinical studies provide evidence for direct and indirect mechanisms by which somatostatin analogs exert antitumor effects. RESULTS Direct antitumor activities, mediated through somatostatin receptors (sst(1)-sst(5)) expressed in tumor cells, include blockade of autocrine/paracrine growth-promoting hormone and growth factor production, inhibition of growth factor-mediated mitogenic signals and induction of apoptosis. Indirect antitumor effects include inhibition of growth-promoting hormone and growth factor secretion, and antiangiogenic actions. Many human tumors express more than one somatostatin receptor subtype, with sst(2) being predominant. Somatostatin analogs such as octreotide and lanreotide, which present a high affinity for sst(2), are in current clinical use to alleviate symptoms in patients with endocrine tumors, and radiolabeled somatostatin analogs have been developed for diagnosis and radiotherapy. CONCLUSIONS While the rationale exists for the use of somatostatin analogs as antitumor agents, studies are ongoing to identify analogs with activity across the range of receptor subtypes to maximize the potential of such treatment.
Collapse
Affiliation(s)
- C Susini
- INSERM U151, Institut Louis Bugnard, Toulouse, France.
| | | |
Collapse
|
68
|
Singh R, Pantarotto D, Lacerda L, Pastorin G, Klumpp C, Prato M, Bianco A, Kostarelos K. Tissue biodistribution and blood clearance rates of intravenously administered carbon nanotube radiotracers. Proc Natl Acad Sci U S A 2006; 103:3357-62. [PMID: 16492781 PMCID: PMC1413890 DOI: 10.1073/pnas.0509009103] [Citation(s) in RCA: 623] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Carbon nanotubes (CNT) are intensively being developed for biomedical applications including drug and gene delivery. Although all possible clinical applications will require compatibility of CNT with the biological milieu, their in vivo capabilities and limitations have not yet been explored. In this work, water-soluble, single-walled CNT (SWNT) have been functionalized with the chelating molecule diethylentriaminepentaacetic (DTPA) and labeled with indium ((111)In) for imaging purposes. Intravenous (i.v.) administration of these functionalized SWNT (f-SWNT) followed by radioactivity tracing using gamma scintigraphy indicated that f-SWNT are not retained in any of the reticuloendothelial system organs (liver or spleen) and are rapidly cleared from systemic blood circulation through the renal excretion route. The observed rapid blood clearance and half-life (3 h) of f-SWNT has major implications for all potential clinical uses of CNT. Moreover, urine excretion studies using both f-SWNT and functionalized multiwalled CNT followed by electron microscopy analysis of urine samples revealed that both types of nanotubes were excreted as intact nanotubes. This work describes the pharmacokinetic parameters of i.v. administered functionalized CNT relevant for various therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Ravi Singh
- *Centre for Drug Delivery Research, School of Pharmacy, University of London, London WC1N 1AX, United Kingdom
| | - Davide Pantarotto
- *Centre for Drug Delivery Research, School of Pharmacy, University of London, London WC1N 1AX, United Kingdom
- Institut de Biologie Moléculaire et Cellulaire, Unité Propre de Recherche 9021, Centre National de la Recherche Scientifique, Immunologie et Chimie Thérapeutiques, 67084 Strasbourg, France; and
- Dipartimento di Scienze Farmaceutiche, Università di Trieste, 34127 Trieste, Italy
| | - Lara Lacerda
- *Centre for Drug Delivery Research, School of Pharmacy, University of London, London WC1N 1AX, United Kingdom
| | - Giorgia Pastorin
- Institut de Biologie Moléculaire et Cellulaire, Unité Propre de Recherche 9021, Centre National de la Recherche Scientifique, Immunologie et Chimie Thérapeutiques, 67084 Strasbourg, France; and
| | - Cédric Klumpp
- Institut de Biologie Moléculaire et Cellulaire, Unité Propre de Recherche 9021, Centre National de la Recherche Scientifique, Immunologie et Chimie Thérapeutiques, 67084 Strasbourg, France; and
- Dipartimento di Scienze Farmaceutiche, Università di Trieste, 34127 Trieste, Italy
| | - Maurizio Prato
- Dipartimento di Scienze Farmaceutiche, Università di Trieste, 34127 Trieste, Italy
| | - Alberto Bianco
- Institut de Biologie Moléculaire et Cellulaire, Unité Propre de Recherche 9021, Centre National de la Recherche Scientifique, Immunologie et Chimie Thérapeutiques, 67084 Strasbourg, France; and
| | - Kostas Kostarelos
- *Centre for Drug Delivery Research, School of Pharmacy, University of London, London WC1N 1AX, United Kingdom
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
69
|
Yang SK, Parkington HC, Blake AD, Keating DJ, Chen C. Somatostatin increases voltage-gated K+ currents in GH3 cells through activation of multiple somatostatin receptors. Endocrinology 2005; 146:4975-84. [PMID: 16081634 DOI: 10.1210/en.2005-0696] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The secretion of GH by somatotropes is inhibited by somatostatin (SRIF) through five specific membrane receptors (SSTRs). SRIF increases both transient outward (IA) and delayed rectifying (IK) K+ currents. We aim to clarify the subtype(s) of SSTRs involved in K+ current enhancement in GH3 somatotrope cells using specific SSTR subtype agonists. Expression of all five SSTRs was confirmed in GH3 cells by RT-PCR. Nystatin-perforated patch clamp was used to record voltage-gated K+ currents. We first established the presence of IA and IK type K+ currents in GH3 cells using different holding potentials (-40 or -70 mV) and specific blockers (4-aminopirimidine and tetraethylammonium chloride). SRIF (200 nM) increased the amplitude of both IA and IK in a fully reversible manner. Various concentrations of each specific SRTR agonist were tested on K+ currents to find the maximal effective concentration. Activation of SSTR2 and SSTR4 by their respective agonists, L-779,976 and L-803,087 (10 nM), increased K+ current amplitude without preference to IA or IK, and abolished any further increase by SRIF. Activation of SSTR1 and SSTR5 by their respective agonists, L-797,591 or L-817,818 (10 nM), increased K+ current amplitude, but SRIF evoked a further increase. The SSTR3 agonist L-797,778 (10 nM) did not affect the K+ currents or the response to SRIF. These results indicate that SSTR1, -2, -4, and -5 may all be involved in the enhancement of K+ currents by SRIF but that only the activation of SSTR2 or -4 results in the full activation of K+ current caused by SRIF.
Collapse
Affiliation(s)
- Seung-Kwon Yang
- Prince Henry's Institute of Medical Research, Monash University, Melbourne, Australia
| | | | | | | | | |
Collapse
|
70
|
Abstract
There has been an exponential growth in the development of radiolabeled peptides for diagnostic and therapeutic applications in the last decade. The automated means of synthesizing these compounds in large quantities and the simplified methods of purifying, characterizing, and optimizing them have kindled attention to peptides as carrier molecules. These new techniques have accelerated the commercial development of radiolabelled peptides, which has provided additional radiopharmaceuticals for the nuclear medicine community. Peptides have many key properties including fast clearance, rapid tissue penetration, and low antigenicity, and can be produced easily and inexpensively. However, there may be problems with in vivo catabolism, unwanted physiologic effects, and chelate attachment. Radiolabeled peptides have made their greatest impact in the management of relatively rare neuroendocrine malignancies. Indeed, Indium-111 ((111)In)-pentetreotide ((111)In-DTPA-octreotide, Octreoscan), which binds to somatostatin receptors (SSTRs), has become the diagnostic 'gold standard' in these diseases. However, (111)In-pentetreotide has been less successful in the diagnosis of other more prevalent diseases in which SSTRs are upregulated. Technetium-99m (99mTc)-depreotide (NeoTect), a 99mTc-labeled SSTR-analog, could have wider impact since it has high sensitivity and specificity for lung cancer lesion detection. However, this impact may be minimized by the increased availability of positron emission tomography imaging with Fluorine-18 (18F)-flourodeoxyglucose, which has similar sensitivity and specificity for lesion identification in this disease, and is currently more widely used. The receptors for bombesin, alpha-melanocyte-stimulating hormone, neurotensin, and the integrin alpha(v)beta3, are under active investigation as targets for radiolabelled peptides, but are still in the pre-clinical stage. Compounds directed at the cholecystokinin-B/gastrin receptor have shown promising results in clinical trials in humans. Radiolabelled peptide therapy is usually indicated for patients with widespread disease that is not amenable to focused radiation therapy or is refractory to chemotherapy. Phase I/II studies using various radiolabelled peptides (including (111)In-pentetreotide, Yttrium-90 [90Y]-DOTA-Phe1-Tyr3-octreotide, 90Y-DOTA-lanreotide, and Lutetium-177 [177Lu]-DOTA-octreotate) for the treatment of patients with neuroendocrine malignancy are in progress. Over 400 patients have been treated, and the response rate has ranged from 60% to 75%, although few patients have had a complete response. Patients have been given individual doses ranging from 2 to 11 GBq with a slow infusion every 4-8 weeks (up to 12 times). The kidney is the dose-limiting organ and most patients experience a transient decline in blood cell counts. A concomitant infusion of an amino acid mixture can reduce kidney toxicity and increase the effective tumor dose. Other peptides currently under investigation, some of which have shown promising results, include Rhenium-188 (188Re)-P2045 and 90Y-alpha(v)beta3 antagonist.
Collapse
Affiliation(s)
- Ronald E Weiner
- Department of Diagnostic Imaging and Therapeutics, University of Connecticut Health Center, Farmington, Connecticut 06030-2804, USA.
| | | |
Collapse
|
71
|
Rivera JA, Alturaihi H, Kumar U. Differential regulation of somatostatin receptors 1 and 2 mRNA and protein expression by tamoxifen and estradiol in breast cancer cells. J Carcinog 2005; 4:10. [PMID: 16018813 PMCID: PMC1184089 DOI: 10.1186/1477-3163-4-10] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2004] [Accepted: 07/14/2005] [Indexed: 11/10/2022] Open
Abstract
Somatostatin (SST) inhibition of hormone hypersecretion from tumors is mediated by somatostatin receptors (SSTRs). SSTRs also play an important role in controlling tumor growth through specific antiproliferative actions. These receptors are well expressed in numerous normal and tumor tissues and are susceptible to regulation by a variety of factors. Estradiol, a potent trophic and mitogenic hormone in its target tissues, is known to modulate the expression of SST and its receptors. Accordingly, in the present study, we determined the effects of tamoxifen, a selective estrogen receptor (ER) modulator (SERM), and estradiol on SSTR1 and SSTR2 expression at the mRNA and protein levels in ER-positive and -negative breast cancer cells. We found that SSTR1 was upregulated by tamoxifen in a dose-dependent manner but no effect was seen with estradiol. In contrast, SSTR2 was upregulated by both tamoxifen and estradiol. Combined treatment caused suppression of SSTR1 below control levels but had no significant effect on SSTR2. Treatment with SSTR1-specific agonist was significantly more effective in suppressing cell proliferation of cells pre-treated with tamoxifen. Taking these data into consideration, we suggest that tamoxifen and estradiol exert variable effects on SSTR1 and SSTR2 mRNA and protein expression and distributional pattern of the receptors. These changes are cell subtype-specific and affect the ability of SSTR agonists to inhibit cell proliferation.
Collapse
Affiliation(s)
- Juan A Rivera
- Fraser Laboratories For Diabetes Research, Department of Medicine, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, H3A 1A1, Canada
| | - Haydar Alturaihi
- Fraser Laboratories For Diabetes Research, Department of Medicine, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, H3A 1A1, Canada
| | - Ujendra Kumar
- Fraser Laboratories For Diabetes Research, Department of Medicine, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, H3A 1A1, Canada
| |
Collapse
|
72
|
Li Y, Si JM, Zhang J, Du J, Wang F, Jia B. Somatostatin receptor subtype 2-mediated scintigraphy and localization using 99mTc-HYNIC-Tyr 3-octreotide in human hepatocellular carcinoma-bearing nude mice. World J Gastroenterol 2005; 11:3953-7. [PMID: 15991301 PMCID: PMC4504904 DOI: 10.3748/wjg.v11.i25.3953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the uptake of 99mTc-HYNIC-Tyr3-octreotide (99mTc-HYNIC-TOC) in human hepatocellular carcinoma (HCC), which can provide the localizable diagnosis in hepatic carcinoma.
METHODS: The expression of somatostatin receptor 2 (SSTR2) messenger RNA (mRNA) in human HCC cell line HepG2 was examined by reverse transcriptase-polymerase chain reaction (RT-PCR). Uptake of 99mTc-HYNIC-TOC was evaluated in the human HCC implanted into BALB/c nude mice. ANMIS2000 nuclear medicine analysis system was used to calculate the ratio of 99mTc uptake between tumor tissue and vital organs.
RESULTS: We demonstrated the expression of SSTR2 mRNA in human HCC cell line HepG2 by RT-PCR. The size of the RT-PCR products was 364 bp detected by sequence analysis of the human SSTR2 mRNA. Scintigraphy proved that 99mTc-HYNIC-TOC was uptaken in the tumor tissue, liver and kidney of the tumor-bearing mice.
CONCLUSION: Based on expression of the SSTR2 mRNA in human HCC, 99mTc-HYNIC-TOC can markedly bind with and be uptaken by human HCC tissues as compared with normal liver tissue. The significant retention of radionuclide in kidney and bladder is probably related to non-specific peptide uptake in the tubulus cells of kidney and possibly due to excretion by kidney. Our results show that localizable diagnosis and targeting radiotherapy with radionuclide-labeled somatostatin analog for HCC are of great value to be further studied.
Collapse
Affiliation(s)
- Yong Li
- Gastrointestinal Laboratory of Clinical Medical Institute of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | | | | | | | | | | |
Collapse
|
73
|
Fridkin G, Bonasera TA, Litman P, Gilon C. Backbone metal-cyclization: a novel approach for simultaneous peptide cyclization and radiolabeling. Application to the combinatorial synthesis of rhenium-cyclic somatostatin analogs. Nucl Med Biol 2005; 32:39-50. [PMID: 15691660 DOI: 10.1016/j.nucmedbio.2004.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2004] [Revised: 08/08/2004] [Accepted: 08/14/2004] [Indexed: 11/25/2022]
Abstract
A novel approach for the combinatorial synthesis of backbone-derived metal-cyclic peptide libraries is presented. In this approach the metalo-cyclic peptides are prepared from their linear precursors through complexation of a metal atom via two hemi-chelating arms located on the peptide backbone. Thus, cyclization and metal labeling of the peptides are achieved simultaneously. A library, composed of 48 rhenium-cyclic somatostatin analogs, was prepared. All rhenium somatostatin complexes exhibited high to moderate in vitro binding affinities toward cloned human somatostatin receptor subtype 2 (hsstr2). Five rhenium-cyclic peptides were found to be most potent with IC50 values between 1 and 3 nM making them promising leads for further development of tumor diagnostic and therapeutic radiolabeled agents. A 99mTc somatostatin cyclic analog was successfully prepared by the same method.
Collapse
Affiliation(s)
- Gil Fridkin
- Department of Organic Chemistry, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | | | | | |
Collapse
|
74
|
Slijkhuis WA, Stadheim L, Hassoun ZM, Nzeako UC, Kremers WK, Talwalkar JA, Gores GJ. Octreotide therapy for advanced hepatocellular carcinoma. J Clin Gastroenterol 2005; 39:333-8. [PMID: 15758629 DOI: 10.1097/01.mcg.0000155136.35315.de] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Treatment options for advanced hepatocellular carcinoma (HCC) remain limited. Recently, octreotide has been proposed for therapy, although its efficacy remains controversial. Thus, the aim of this open-label pilot study was to evaluate the response of HCC to long-acting octreotide (Sandostatin LAR). Thirty patients were enrolled for this prospective 2-year trial. Initially, patients were given short acting octreotide to ensure drug tolerability. Thereafter, patients received long-acting octreotide 30 mg IM every 4 to 6 weeks. Measurable disease was assessed at 3-month intervals. Five of 30 patients were unable to tolerate the test dose, and 1 patient was reevaluated and underwent hepatic resection. The remaining 24 patients, who received long-acting octreotide, all had advanced stage of disease with multifocal-massive morphology (67%), vascular thrombosis (63%), or extrahepatic spread (17%), but well compensated liver disease. The treatment was well tolerated, except for diarrhea. Median time to tumor progression was 3.6 months, and median survival was 5.1 months. Seven patients (29%) had stable disease (median duration of 8.0 months) with 2 patients demonstrating disease stability for 24 months. In conclusion, although occasional patients appear to have stable disease on long-acting octreotide therapy, overall the beneficial response in terms of time to tumor progression and survival is limited.
Collapse
Affiliation(s)
- Wilco A Slijkhuis
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
75
|
Sukopp M, Schwab R, Marinelli L, Biron E, Heller M, Várkondi E, Pap A, Novellino E, Kéri G, Kessler H. Structure−Activity Relationship Studies Optimizing the Antiproliferative Activity of Novel Cyclic Somatostatin Analogues Containing a Restrained Cyclic β-Amino Acid. J Med Chem 2005; 48:2916-26. [PMID: 15828830 DOI: 10.1021/jm049500j] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The cyclic somatostatin analogue cyclo[Pro(1)-Phe(2)-D-Trp(3)-Lys(4)-Thr(5)-Phe(6)] (L-363,301) displays high biological activity in inhibiting the release of growth hormone, insulin, and glucagon. According to the sequence of L-363,301, we synthesized a number of cyclic hexa- and pentapeptides containing nonnatural alpha- and beta-amino acids. The N- fluorenylmethoxycarbonyl protected cyclic beta-amino acid [1S, 2S, 5R]-2-amino-3,5-dimethyl-2-cyclohex-3-enecarboxylic acid (cbetaAA), for the replacement of the Phe(6)-Pro(1) moiety of L-363,301, was synthesized in two steps by an enantioselective multicomponent reaction using (-)-8-phenylmenthol as a chiral auxiliary. The resulting peptide cyclo[cbetaAA(1)-Tyr(2)-D-Trp(3)-Nle(4)-Thr(Trt)(5)] (Trt = triphenylmethyl) shows high antiproliferative effects in an in vitro assay with A431 cancer cells. The same peptide without the Trt group does not reveal any biological activity, whereas L-363,301 and closely related hexapeptides show only minor activity. By comparison of the solution structure of cyclo[cbetaAA(1)-Tyr(2)-D-Trp(3)-Nle(4)-Thr(Trt)(5)] with the structure of l-363,301, a nearly perfect match of the betaII'-turn region with d-Trp in the i + 1 position was observed. The cyclic beta-amino acid cbetaAA is likely needed for the bioactive conformation of the peptide.
Collapse
Affiliation(s)
- Martin Sukopp
- Lehrstuhl für Organische Chemie und Biochemie II, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Rivier JE, Kirby DA, Erchegyi J, Waser B, Eltschinger V, Cescato R, Reubi JC. Somatostatin receptor 1 selective analogues: 3. Dicyclic peptides. J Med Chem 2005; 48:515-22. [PMID: 15658865 DOI: 10.1021/jm049519m] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The binding affinity of short chain somatostatin (SRIF) analogues at the five human SRIF receptors (sst) was determined to identify sterically constrained somatostatin receptor subtype 1 (sst(1)) selective scaffolds. Des-AA(1,2,4,13)-[d-Trp(8)]SRIF (2) retained high binding affinity at all receptors but sst(1), Des-AA(1,2,4,5)-[d-Trp(8)]SRIF (3) at sst(4) and sst(5), and Des-AA(1,2,4,5,13)-[d-Trp(8)]SRIF (4) at sst(2) and sst(4) (AA = amino acid). Des-AA(1,2,4,12,13)-[d-Trp(8)]SRIF (6) was potent and sst(4)-selective (>25-fold); Des-AA(1,2,5,12,13)-[d-Trp(8)]SRIF (7) and Des-AA(1,2,4,5,12,13)-[d-Trp(8)]-SRIF (9, ODT-8) were most potent at sst(4) and moderately potent at all other receptors. Dicyclic SRIF agonists of the sst(1)-selective Des-AA(1,5)-[Tyr(2),d-Trp(8),IAmp(9)]SRIF, (14, sst(1) IC(50) = 14 nM) were prepared in which a lactam bridge introduced additional conformational constraint (IAmp = 4-(N-isopropyl)-aminomethylphenylalanine). Cyclo(7-12)Des-AA(1,5)-[Tyr(2),Glu(7),d-Trp(8),IAmp(9),hhLys(12)]SRIF (31) (sst(1) IC(50) = 16 nM) and cyclo(7-12) Des-AA(1,2,5)-[Glu(7),d-Trp(8),IAmp(9),m-I-Tyr(11),hhLys(12)]SRIF (45) (sst(1) IC(50) = 6.1 nM) had equal or improved affinities over that of the parent 14. Binding affinity was decreased in all other cases with alternate bridging constraints such as cyclo (6-11), cyclo (6-12), and cyclo (7-11). Compound 45 is an agonist (EC(50) = 8.8 nM) in the adenylate cyclase assay.
Collapse
Affiliation(s)
- Jean E Rivier
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, California 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
77
|
Marques F, Paulo A, Campello MP, Lacerda S, Vitor RF, Gano L, Delgado R, Santos I. Radiopharmaceuticals for targeted radiotherapy. RADIATION PROTECTION DOSIMETRY 2005; 116:601-4. [PMID: 16604708 DOI: 10.1093/rpd/nci251] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
This work intends to find specific radiopharmaceuticals for cancer therapy based on beta (153Sm and 166Ho) or Auger (99Tc(m)) emitter radionuclides, using cyclic and acyclic polyamines as bifunctional chelators. These chelators are designed to allow the binding of a tumour seeking biomolecule and/or a DNA intercalator. The cyclic amines, such as 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid, 1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid and 1,4,7,10-tetraazacyclotridecane-1,4,7,10-tetraacetic acid, were radiolabelled with 153Sm and 166Ho. The radiochemical and biological behaviour of the resulting complexes were evaluated in order to assess their potential as building blocks for the attachment of selected biomolecules, with the aim of further applying them for the development of specific therapeutic radiopharmaceuticals. Novel pyrazolyldiamines, bearing a DNA intercalating anthracenyl fragment, were also explored to synthesize radioactive complexes with the fac-[99Tc(m)(CO)]3]+ moiety. The identity of these 99Tc(m) tricarbonyl complexes was confirmed by high-performance liquid chromatography comparison with rhenium congeners fully characterized. By including a DNA intercalator into the chelator framework, we expect to induce more efficient and selective damage to the DNA of cancer cells by the action of the short-range Auger electrons emitted by 99Tc(m).
Collapse
Affiliation(s)
- Fernanda Marques
- Departamento de Química, Instituto Tecnológico e Nuclear, EN 10, Apartado 21, 2686-953 Sacavém, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Eberle AN, Mild G, Froidevaux S. Receptor-Mediated Tumor Targeting with Radiopeptides. Part 1. General Concepts and Methods: Applications to Somatostatin Receptor-Expressing Tumors. J Recept Signal Transduct Res 2004; 24:319-455. [PMID: 15648449 DOI: 10.1081/rrs-200040939] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Radiolabeled peptides have become important tools in nuclear oncology, both as diagnostics and more recently also as therapeutics. They represent a distinct sector of the molecular targeting approach, which in many areas of therapy will implement the old "magic bullet" concept by specifically directing the therapeutic agent to the site of action. In this three-part review, we present a comprehensive overview of the literature on receptor-mediated tumor targeting with the different radiopeptides currently studied. Part I summarizes the general concepts and methods of targeting, the selection of radioisotopes, chelators, and the criteria of peptide ligand development. Then, the >400 studies on the application to somatostatin/somatostatin-release inhibiting factor receptor-mediated tumor localization and treatment will be reviewed, demonstrating that peptide radiopharmaceuticals have gained an important position in clinical medicine.
Collapse
Affiliation(s)
- Alex N Eberle
- Laboratory of Endocrinology, Department of Research, University Hospital and University Children's Hospital, Basel, Switzerland.
| | | | | |
Collapse
|
79
|
Abstract
Accurate and rapid detection of tumors is of great importance for interrogating the molecular basis of cancer pathogenesis, preventing the onset of complications, and implementing a tailored therapeutic regimen. In this era of molecular medicine, molecular probes that respond to, or target molecular processes are indispensable. Although numerous imaging modalities have been developed for visualizing pathologic conditions, the high sensitivity and relatively innocuous low energy radiation of optical imaging method makes it attractive for molecular imaging. While many human diseases have been studied successfully by using intrinsic optical properties of normal and pathologic tissues, molecular imaging of the expression of aberrant genes, proteins, and other pathophysiologic processes would be enhanced by the use of highly specific exogenous molecular beacons. This review focuses on the development of receptor-specific molecular probes for optical imaging of tumors. Particularly, bioconjugates of probes that absorb and fluoresce in the near infrared wavelengths between 750 and 900 nm will be reviewed.
Collapse
Affiliation(s)
- Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, 4525 Scot Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
80
|
Dasgupta P. Somatostatin analogues: multiple roles in cellular proliferation, neoplasia, and angiogenesis. Pharmacol Ther 2004; 102:61-85. [PMID: 15056499 DOI: 10.1016/j.pharmthera.2004.02.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Angiogenesis, the development of new blood vessels is a crucial process both for tumor growth and metastatic dissemination. Additionally, dysregulation in angiogenesis has been implicated in the pathogenesis of cardiovascular disease, proliferative retinopathy, diabetic nephropathy, and rheumatoid arthritis (RA). The neuropeptide somatostatin has been shown to be a powerful inhibitor of neovascularization in several experimental models. Furthermore, somatostatin receptors (sst) are expressed on endothelial cells; particularly, sst2 has been found to be uniquely up-regulated during the angiogenic switch, from quiescent to proliferative endothelium. The present manuscript reviews the anti-angiogenic activity of somatostatin and its analogues in neoplastic and nonneoplastic disease. The role of sst subtypes particularly sst2 in mediating its angioinhibitory activity is described. Somatostatin agonists may also exert their anti-angiogenic activity indirectly by inhibition of growth factors like vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and the growth hormone (GH)/insulin-like growth factor-I (IGF-I) axis or through its immunomodulatory effects. However, the therapeutic utility of somatostatin agonists as anti-angiogenic drugs in these diseases remains confusing because of conflicting results from different studies. More basic research, as well as patient-oriented studies, is required to firmly establish the clinical potential of somatostatin agonists in therapeutic angiogenesis. The currently available somatostatin agonists have high affinity of sst2 with lower affinities for sst3 and sst5. The emergence of novel somatostatin agonists especially bispecific analogues (agonists targeting multiple cellular receptors) and conjugates (synthesized by chemically linking somatostatin analogues with other antineoplastic agents) with improved receptor specificity signify a new generation of anti-angiogenics, which may represent novel strategies in the treatment of neovascularization-related diseases.
Collapse
Affiliation(s)
- Piyali Dasgupta
- Department of Interdisciplinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Room 2068A, MRC-2 East, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| |
Collapse
|
81
|
Marques F, Guerra KP, Gano L, Costa J, Campello MP, Lima LMP, Delgado R, Santos I. 153Sm and 166Ho complexes with tetraaza macrocycles containing pyridine and methylcarboxylate or methylphosphonate pendant arms. J Biol Inorg Chem 2004; 9:859-72. [PMID: 15338314 DOI: 10.1007/s00775-004-0587-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Accepted: 07/27/2004] [Indexed: 11/26/2022]
Abstract
A set of tetraaza macrocycles containing pyridine and methylcarboxylate (ac(3)py14) or methylphosphonate (MeP(2)py14 and P(3)py14) pendant arms were prepared and their stability constants with La(3+), Sm(3+), Gd(3+) and Ho(3+) determined by potentiometry at 25 degrees C and 0.10 M ionic strength in NMe(4)NO(3). The metal:ligand ratio for (153)Sm and (166)Ho and for ac(3)py14, MeP(2)py14 and P(3)py14, as well as the pH of the reaction mixtures, were optimized to achieve a chelation efficiency higher than 98%. These radiocomplexes are hydrophilic and have a significant plasmatic protein binding. In vitro stability was studied in physiological solutions and in human serum. All complexes are stable in saline and PBS, but 20% of radiochemical impurities were detected after 24 h of incubation in serum. Biodistribution studies in mice indicated a slow rate of clearance from blood and muscle, a high and rapid liver uptake and a very slow rate of total radioactivity excretion. Some bone uptake was observed for complexes with MeP(2)py14 and P(3)py14, which was enhanced with time and the number of methylphosphonate groups. This biological profile supports the in vitro instability found in serum and is consistent with the thermodynamic stability constants found for these complexes.
Collapse
Affiliation(s)
- Fernanda Marques
- Instituto Tecnológico e Nuclear, Estrada Nacional 10, 2686-953 Sacavém, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Affiliation(s)
- Michael Doubrovin
- Cotzias Neuro-Oncology Lab, Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | | | | |
Collapse
|
83
|
Lu B, Xu Y, Ding ZS. Effect of octreotide on growth and apoptosis of human gastric cancer cell line SGC-7901. Shijie Huaren Xiaohua Zazhi 2004; 12:527-529. [DOI: 10.11569/wcjd.v12.i3.527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the effects of somatostatin analog (octreotide) on the growth and apoptosis of gastric cancer cell line.
METHODS: The proliferation of gastric cancer cell line SGC-7901 was determined in octreotide groups and control group by MTT. The cell morphology was observed under HE staining and electron microscopy. The cell cycle and apoptosis ratio were measured using flow cytometry analysis.
RESULTS: The growth of gastric cancer cell line SGC-7901 was significantly inhibited by octreotide with 160-480 mg/L (vs controls, P < 0.001), and the maximal inhibitory rate was 25.4%. Morphological variations of apoptosis were observed at 48 hours treated with octreotide by HE staining and apoptosis body was observed under electron microscope. Apoptotic rate was 9.42%±5.29% (P < 0.05).
CONCLUSION: Octreotide can suppress the growth of gastric cancer cell line SGC-7901 and induce apoptosis of the cells.
Collapse
|
84
|
Weckbecker G, Lewis I, Albert R, Schmid HA, Hoyer D, Bruns C. Opportunities in somatostatin research: biological, chemical and therapeutic aspects. Nat Rev Drug Discov 2004; 2:999-1017. [PMID: 14654798 DOI: 10.1038/nrd1255] [Citation(s) in RCA: 408] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Gisbert Weckbecker
- Transplantation and Immunology, Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland.
| | | | | | | | | | | |
Collapse
|
85
|
Gibril F, Jensen RT. Diagnostic uses of radiolabelled somatostatin receptor analogues in gastroenteropancreatic endocrine tumours. Dig Liver Dis 2004; 36 Suppl 1:S106-20. [PMID: 15077919 DOI: 10.1016/j.dld.2003.11.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Numerous studies have established that gastroenteropancreatic endocrine tumours (carcinoids and pancreatic endocrine tumours) resemble a number of other tumours in overexpressing somatostatin receptors that can bind octreotide or lanreotide with high affinity (i.e. possess sst2/sst5 receptors). Recent studies report that radiolabelled somatostatin analogues can be used to image these tumours (somatostatin receptor scintigraphy) and may be useful for peptide-directed radionuclide therapy. In this paper the evidence is reviewed that has led to establishing somatostatin receptor scintigraphy as the initial imaging modality of choice in patients with gastroenteropancreatic tumours. This conclusion is based on an understanding of the results with conventional imaging modalities (ultrasound, computed tomographic scan, magnetic resonance imaging, angiography) available prior to somatostatin receptor scintigraphy and the results of studies demonstrating the sensitivity and specificity of somatostatin receptor scintigraphy. Most important in this regard are the results of studies that have assessed the use of somatostatin receptor scintigraphy on clinical management. Each of these areas is reviewed in this paper.
Collapse
Affiliation(s)
- F Gibril
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Building 10, Room 9C-103, 10 Center Drive MSC-1804, Bethesda, MD 20892-1804, USA
| | | |
Collapse
|
86
|
Schwaiger M, Weber W. Molecular imaging: dream or reality? ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2004:1-18. [PMID: 15248514 DOI: 10.1007/978-3-662-07310-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
|
87
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2003; 11:1755-1759. [DOI: 10.11569/wcjd.v11.i11.1755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
88
|
Abstract
PURPOSE OF REVIEW Gastric acid facilitates the digestion of protein and the absorption of iron, calcium, and vitamin B12. It also protects against bacterial overgrowth and enteric infection, including prion disease. When homeostatic mechanisms malfunction, the volume and concentration of acid may overwhelm mucosal defense mechanisms, leading to duodenal ulcer, gastric ulcer, and gastroesophageal reflux disease. This article reviews recent knowledge contributing to understanding of the regulation of gastric acid secretion at the central, peripheral, and intracellular levels. RECENT FINDINGS The vagus nerve contains afferent fibers that transmit sensory information from the stomach to the nucleus of the solitary tract. Input from the nucleus of the solitary tract is relayed to vagal efferent neurons that originate from two brain stem nuclei: the nucleus ambiguus and the dorsal motor nucleus of the vagus. The latter is also influenced by thyrotropin-releasing hormone neurons that act centrally to stimulate acid secretion. The main peripheral stimulants of acid secretion are the hormone gastrin and the paracrine amine histamine. Gastrin stimulates acid secretion directly and, more importantly, indirectly by releasing histamine from fundic enterochromaffin-like cells. Gastrin also exerts trophic effects on various tissues, including the gastric and intestinal mucosa. The main inhibitor of acid secretion is somatostatin. Somatostatin, acting via ssTR2 receptors, exerts a tonic paracrine inhibitory influence on the secretion of gastrin, histamine, and acid secretion. Calcitonin gene-related peptide, adrenomedullin, amylin, atrial natriuretic peptide, and pituitary adenylate cyclase-activating polypeptide all stimulate somatostatin secretion and thus inhibit acid secretion. HK-ATPase, the proton pump of the parietal cell, is stored within cytoplasmic tubulovesicles during the resting state, but during stimulation, it is shuttled to the canalicular membrane by a poorly understood mechanism that probably involves soluble N-ethylmaleimide-sensitive factor attachment protein receptor proteins. The proton pump inhibitor, pantoprazole, is unique in that it binds cysteine 822, located deep within the membrane domain of the alpha-subunit. The difficulty that reducing agents, such as glutathione, have in reaching cysteine 822 may be responsible for the longer half-time for acid recovery observed with pantoprazole. Hypergastrinemia, induced by proton pump inhibitors, enhances expression of cyclooxygenase-2 and hence prostaglandins within parietal cells, a feedback pathway that may protect the stomach against acid-induced damage. SUMMARY In the past year, significant advances have been made in understanding of the regulation of gastric acid secretion. Ultimately, these advances should lead to improved therapies to prevent and treat acid-related disorders. Gastric acid secretion must be precisely controlled at a variety of levels to prevent disease caused by hyperchlorhydria and hypochlorhydria. The mechanisms include neural (central and peripheral), hormonal, paracrine, and intracellular pathways that operate in concert to switch acid secretion on during ingestion of a meal and off during the interdigestive period. A better understanding of the physiology of acid secretion in health and disease should eventually lead to improved therapies to prevent and treat acid-related disorders.
Collapse
Affiliation(s)
- Mitchell L Schubert
- Department of Medicine, Division of Gastroenterology, Medical College of Virginia and McGuire VAMC, Richmond, Virginia, USA.
| |
Collapse
|
89
|
Szepeshazi K, Schally AV, Nagy A, Wagner BW, Bajo AM, Halmos G. Preclinical evaluation of therapeutic effects of targeted cytotoxic analogs of somatostatin and bombesin on human gastric carcinomas. Cancer 2003; 98:1401-10. [PMID: 14508826 DOI: 10.1002/cncr.11649] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND New modalities are necessary for the treatment of patients with unresectable gastric carcinoma. The authors investigated whether receptors for somatostatin and bombesin were present in human gastric carcinoma lines and tested the antitumor effects of cytotoxic somatostatin analog AN-238 and cytotoxic bombesin conjugate AN-215. METHODS Nude mice bearing AGS, Hs 746T, and NCI-N87 human gastric carcinomas were treated with AN-238, AN-215, or their cytotoxic moiety 2-pyrrolinodoxorubicin (AN-201). Tumor growth reduction and tumor doubling times were calculated, and histologic characteristics of cell proliferation and apoptosis were examined. The expression of mRNA for somatostatin and bombesin receptors in tumors was investigated by reverse transcriptase-polymerase chain reaction. Subtypes 2 and 5 of somatostatin receptor proteins (sst2 and sst5, respectively) were analyzed using immunohistochemistry and immunoblotting. Binding assays were performed with radiolabeled somatostatin and bombesin analogs. RESULTS Cytotoxic bombesin analog AN-215 powerfully inhibited the growth of AGS carcinomas that expressed high-affinity subtype 1 bombesin receptors. All three carcinomas expressed high-affinity sst2 and sst5 receptors. Cytotoxic somatostatin analog AN-238 exerted a strong inhibitory effect on NCI-N87 and Hs 746T carcinomas, which exhibited high concentrations of somatostatin receptors, but had a weaker effect on AGS tumors, which expressed the lowest receptor levels. AN-201 had only nonsignificant effects. CONCLUSIONS Experimental human gastric carcinomas that expressed high-affinity subtype 1 bombesin receptors were inhibited by cytotoxic bombesin analog AN-215, and tumors with high concentrations of sst2 or sst5 somatostatin receptors were suppressed by cytotoxic somatostatin analog AN-238. These findings suggest that this class of targeted compounds should be considered for the therapy of patients with advanced gastric carcinoma.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Blotting, Western
- Bombesin/analogs & derivatives
- Bombesin/pharmacology
- Carcinoma/drug therapy
- Carcinoma/pathology
- Disease Models, Animal
- Humans
- Immunohistochemistry
- Injections, Intralesional
- Male
- Mice
- Mice, Nude
- Neoplasm Transplantation
- Probability
- RNA, Neoplasm/analysis
- Receptors, Bombesin/analysis
- Receptors, Bombesin/drug effects
- Receptors, LHRH/metabolism
- Receptors, Somatostatin/analysis
- Receptors, Somatostatin/drug effects
- Reference Values
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Sensitivity and Specificity
- Somatostatin/analogs & derivatives
- Somatostatin/pharmacology
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/pathology
Collapse
Affiliation(s)
- Karoly Szepeshazi
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | |
Collapse
|
90
|
Gao S, Yu BP, Li Y, Dong WG, Luo HS. Antiproliferative effect of octreotide on gastric cancer cells mediated by inhibition of Akt/PKB and telomerase. World J Gastroenterol 2003; 9:2362-5. [PMID: 14562414 PMCID: PMC4656499 DOI: 10.3748/wjg.v9.i10.2362] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the antiproliferative effect of octreotide, a long-acting analogue of somatostatin, on gastric cancer cell line SGC7901 and its possible molecular mechanisms.
METHODS: Gastric cancer cell line SGC7901 employed in the study was treated with 0.008, 0.04, 0.2, 1, 5 and 25 μg·mL-1 of octreotide respectively for 24 h to evaluate the antiproliferative effect of somatostatin analog on the tumor cells by MTT assay method. To elucidate the underlying mechanism, the cells were exposed to 1 μg·mL-1 of octreotide for 0, 12, 24 and 48 h, when their Akt/PKB and telomerase activities were respectively determined using PCR-ELSIA and nonradioactive protein kinase assay protocols. The same experimental procedures were also performed in the control cells that were treated with corresponding vehicles instead of somatostatin analog.
RESULTS: After exposed to octreotide for 24 h at the concentrations of more than 1 μg·mL-1, SGC7901 cells exhibited a dose-dependent inhibition of growth with the inhibiting rate to be as high as 34.66% when 25 μg·mL-1 of octreotide was applied. The Akt/PKB and telomerase activity of SGC7901 cells was significantly inhibited when the cells were exposed to 1 μg·mL-1 of octreotide for 12, 24 and 48 h compared with that of their control counterparts (P < 0.01), both of which exhibited in a time-dependent manner.
CONCLUSION: The antiproliferative effect of octreotide on SGC7901 cells might be mediated by the inhibition of Akt/PKB and telomerase.
Collapse
Affiliation(s)
- Shan Gao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | | | | | | | | |
Collapse
|
91
|
Chini B, Chinol M, Cassoni P, Papi S, Reversi A, Areces L, Marrocco T, Paganelli G, Manning M, Bussolati G. Improved radiotracing of oxytocin receptor-expressing tumours using the new [111In]-DOTA-Lys8-deamino-vasotocin analogue. Br J Cancer 2003; 89:930-6. [PMID: 12942128 PMCID: PMC2394487 DOI: 10.1038/sj.bjc.6601189] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Oxytocin receptors (OTR) have been described in a number of tumours of different origin, and represent a new target for specific radiolabelled oxytocin (OT) analogues in cancer diagnosis and therapy. By linking the DOTA chelating agent to position 8 of the deamino derivative of Lys(8)-vasotocin (dLVT), we obtained a new compound (DOTA-dLVT) with the following characteristics: (1) it forms a monomeric and stable compound that binds to OTR with an affinity comparable to that of the endogenous OT ligand; (2) it is characterised by a very good selectivity profile for the human OTR, with a low affinity binding to the closely related V1a, V1b and V2 vasopressin receptor subtypes; (3) it induces rapid and persistent receptor internalisation and (4) when radiolabelled, [(111)In]-DOTA-dLVT is efficiently and selectively taken up by OTR-positive tumours grown in mice. These features makes radiolabelled DOTA-dLVT a very good candidate for the radiotargeting of OTR-expressing tumours.
Collapse
Affiliation(s)
- B Chini
- CNR Institute of Neuroscience, Cellular and Molecular Pharmacology Section, Via Vanvitelli 32, 20129 Milano, Italy
| | - M Chinol
- European Institute of Oncology, Division of Nuclear Medicine, Via Ripamonti 435, 20141 Milano, Italy
| | - P Cassoni
- Department of Biomedical Sciences and Human Oncology, University of Turin, Via Santena 7, 10126 Torino, Italy
| | - S Papi
- European Institute of Oncology, Division of Nuclear Medicine, Via Ripamonti 435, 20141 Milano, Italy
| | - A Reversi
- CNR Institute of Neuroscience, Cellular and Molecular Pharmacology Section, Via Vanvitelli 32, 20129 Milano, Italy
| | - L Areces
- European Institute of Oncology, Division of Nuclear Medicine, Via Ripamonti 435, 20141 Milano, Italy
| | - T Marrocco
- Department of Biomedical Sciences and Human Oncology, University of Turin, Via Santena 7, 10126 Torino, Italy
| | - G Paganelli
- European Institute of Oncology, Division of Nuclear Medicine, Via Ripamonti 435, 20141 Milano, Italy
| | - M Manning
- Department of Biochemistry and Molecular Biology, Medical College of Ohio, Toledo, PO Box 10008, OH, USA
| | - G Bussolati
- Department of Biomedical Sciences and Human Oncology, University of Turin, Via Santena 7, 10126 Torino, Italy
- Department of Biomedical Sciences and Human Oncology, University of Turin, Via Santena 7, 10126 Torino, Italy. E-mail:
| |
Collapse
|
92
|
Lewis I, Bauer W, Albert R, Chandramouli N, Pless J, Weckbecker G, Bruns C. A novel somatostatin mimic with broad somatotropin release inhibitory factor receptor binding and superior therapeutic potential. J Med Chem 2003; 46:2334-44. [PMID: 12773038 DOI: 10.1021/jm021093t] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A rational drug design approach, capitalizing on structure-activity relationships and involving transposition of functional groups from somatotropin release inhibitory factor (SRIF) into a reduced size cyclohexapeptide template, has led to the discovery of SOM230 (25), a novel, stable cyclohexapeptide somatostatin mimic that exhibits unique high-affinity binding to human somatostatin receptors (subtypes sst1-sst5). SOM230 has potent, long-lasting inhibitory effects on growth hormone and insulin-like growth factor-1 release and is a promising development candidate currently under evaluation in phase I clinical trials.
Collapse
Affiliation(s)
- Ian Lewis
- Transplantation Research Department, Novartis Pharma, CH-4002 Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
93
|
Torchilin VP, Lukyanov AN. Peptide and protein drug delivery to and into tumors: challenges and solutions. Drug Discov Today 2003; 8:259-66. [PMID: 12623240 DOI: 10.1016/s1359-6446(03)02623-0] [Citation(s) in RCA: 257] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The potential of peptide and protein anticancer agents has yet to be realized owing to the many unresolved problems concerning their delivery to the site of a tumor and into tumor cells. However, our understanding of the mechanisms underlying the biological fate and biodistribution of protein and peptide drugs has advanced to the stage where methods that use or influence these mechanisms are now available. There are different approaches that can improve the stability, longevity and targeting of peptides and proteins in the body, such as their modification with various soluble polymers, incorporation into microparticular drug carriers, enhanced permeability and retention effect-based tumor targeting and the use of targeting moieties. Furthermore, new approaches to intracellular drug delivery, including the use of transduction proteins and peptides, are being developed. These advances promise the delivery of a new generation of anticancer drugs.
Collapse
Affiliation(s)
- Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Bouve College of Health Sciences, 312 Mugar Bldg. Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA.
| | | |
Collapse
|
94
|
Abstract
Antitumor monoclonal antibodies have shown clinical promise as cancer cell surface targeting agents. More tumor targeting antibodies are likely to be approved by the FDA in the next few years. However, there are two major limitations in antibody-targeted therapy: large size and nonspecific uptake of the antibody molecules by the liver and the reticuloendothelial system. These result in poor tumor penetration of antibody pharmaceuticals and dose-limiting toxicity to the liver and bone marrow. Peptides are excellent alternative targeting agents for human cancers, and they may alleviate some of the problems with antibody targeting. In the last decade, several investigators have successfully used combinatorial library methods to discover cell surface binding peptides that may be useful for cancer targeting. The phage-display library technique and the "one-bead one-compound" combinatorial library method are the two approaches that have been used. Cancer cell surface receptors or endothelial cell surface receptors of the neovasculature are the two popular therapeutic targets for cancer. Results from preclinical studies with some peptides are encouraging in their targeting potential.
Collapse
Affiliation(s)
- Olulanu H Aina
- Univeristy of California Davis Cancer Center, Division of Hematology/Oncology, and Department of Internal Medicine, University of California Davis, 4501 X Street, Sacramento 95817, USA
| | | | | | | |
Collapse
|