1
|
Tiwari A. The OCEAN Study: Has Octreotide Quenched the Thirst for Angiodysplasia-Related Bleed? Gastroenterology 2024:S0016-5085(24)00489-X. [PMID: 38679395 DOI: 10.1053/j.gastro.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Affiliation(s)
- Avinash Tiwari
- Department of Gastroenterology, Regency Hospital Ltd, Kanpur, Uttar Pradesh, India
| |
Collapse
|
2
|
Milewska-Kranc A, Ćwikła JB, Kolasinska-Ćwikła A. The Role of Receptor-Ligand Interaction in Somatostatin Signaling Pathways: Implications for Neuroendocrine Tumors. Cancers (Basel) 2023; 16:116. [PMID: 38201544 PMCID: PMC10778465 DOI: 10.3390/cancers16010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Neuroendocrine tumors (NETs) arise from neuroendocrine cells and manifest in diverse organs. Key players in their regulation are somatostatin and its receptors (SSTR1-SSTR5). Understanding receptor-ligand interactions and signaling pathways is vital for elucidating their role in tumor development and therapeutic potential. This review highlights SSTR characteristics, localization, and expression in tissues, impacting physiological functions. Mechanisms of somatostatin and synthetic analogue binding to SSTRs, their selectivity, and their affinity were analyzed. Upon activation, somatostatin initiates intricate intracellular signaling, involving cAMP, PLC, and MAP kinases and influencing growth, differentiation, survival, and hormone secretion in NETs. This review explores SSTR expression in different tumor types, examining receptor activation effects on cancer cells. SSTRs' significance as therapeutic targets is discussed. Additionally, somatostatin and analogues' role in hormone secretion regulation, tumor growth, and survival is emphasized, presenting relevant therapeutic examples. In conclusion, this review advances the knowledge of receptor-ligand interactions and signaling pathways in somatostatin receptors, with potential for improved neuroendocrine tumor treatments.
Collapse
Affiliation(s)
| | - Jarosław B. Ćwikła
- School of Medicine, University of Warmia and Mazury, Aleja Warszawska 30, 10-082 Olsztyn, Poland
- Diagnostic Therapeutic Center–Gammed, Lelechowska 5, 02-351 Warsaw, Poland
| | | |
Collapse
|
3
|
Mithanthaya A, Ismail AGM, Muwanwella N, Venugopal K. Blue rubber bleb nevus syndrome in a patient on anticoagulation: a management dilemma. BMJ Case Rep 2023; 16:e256702. [PMID: 37949469 PMCID: PMC10649631 DOI: 10.1136/bcr-2023-256702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023] Open
Abstract
Blue rubber bleb nevus syndrome (BRBNS) is a rare condition characterised by vascular malformations mostly of the skin and gastrointestinal tract and less commonly of the central nervous system, liver, thyroid, spleen and lungs. We report a rare case of BRBNS in a patient on anticoagulation who presented with gastrointestinal bleeding and no cutaneous or other organ involvement. We discuss the difficulty in balancing bleeding and clotting risks in this patient who developed two episodes of venous thromboembolism while off anticoagulation to minimise gastrointestinal bleeding. We also highlight the potential role of somatostatin analogues such as lanreotide in decreasing gastrointestinal bleeding risk in BRBNS, particularly in the setting of anticoagulation. The occurrence of two episodes of venous thromboembolism within a short time frame in this case, in conjunction with known associations between other vascular anomalies and venous thromboembolism, raises the question of whether BRBNS may be associated with a prothrombotic state.
Collapse
Affiliation(s)
- Aditya Mithanthaya
- Gastroenterology, Royal Perth Hospital, Perth, Western Australia, Australia
| | | | | | - Kannan Venugopal
- Gastroenterology, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
4
|
Is There a Place for Somatostatin Analogues for the Systemic Treatment of Hepatocellular Carcinoma in the Immunotherapy Era? LIVERS 2022. [DOI: 10.3390/livers2040024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Patients with advanced hepatocellular carcinoma (HCC) have a very limited survival rate even after the recent inclusion of kinase inhibitors or immune checkpoint inhibitors in the therapeutic armamentarium. A significant problem with the current proposed therapies is the considerable cost of treatment that may be a serious obstacle in low- and middle-income countries. Implementation of somatostatin analogues (SSAs) has the potential to overcome this obstacle, but due to some negative studies their extensive evaluation came to a halt. However, experimental evidence, both in vitro and in vivo, has revealed various mechanisms of the anti-tumor effects of these analogues, including inhibition of cancer cell proliferation and angiogenesis and induction of apoptosis. Favorable indirect effects such as inhibition of liver inflammation and fibrosis and influence on macrophage-mediated innate immunity have also been noted and are presented in this review. Furthermore, the clinical application of SSAs is both presented and compared with clinical trials of kinase and immune checkpoint inhibitors (ICIs). No direct trials have been performed to compare survival in the same cohort of patients, but the cost of treatment with SSAs is a fraction compared to the other modalities and with significantly less serious side effects. As in immunotherapy, patients with viral HCC (excluding alcoholics), as well as Barcelona stage B or C and Child A patients, are the best candidates, since they usually have a survival prospect of at least 6 months, necessary for optimum results. Reasons for treatment failures are also discussed and further research is proposed.
Collapse
|
5
|
Role of Somatostatin Signalling in Neuroendocrine Tumours. Int J Mol Sci 2022; 23:ijms23031447. [PMID: 35163374 PMCID: PMC8836266 DOI: 10.3390/ijms23031447] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Somatostatin (SST) is a small peptide that exerts inhibitory effects on a wide range of neuroendocrine cells. Due to the fact that somatostatin regulates cell growth and hormone secretion, somatostatin receptors (SSTRs) have become valuable targets for the treatment of different types of neuroendocrine tumours (NETs). NETs are a heterogeneous group of tumours that can develop in various parts of the body, including the digestive system, lungs, and pituitary. NETs are usually slow growing, but they are often diagnosed in advanced stages and can display aggressive behaviour. The mortality rate of NETs is not outstandingly increased compared to other malignant tumours, even in the metastatic setting. One of the intrinsic properties of NETs is the expression of SSTRs that serve as drug targets for SST analogues (SSAs), which can delay tumour progression and downregulate hormone overproduction. Additionally, in many NETs, it has been demonstrated that the SSTR expression level provides a prognostic value in predicting a therapeutic response. Furthermore, higher a SSTR expression correlates with a better survival rate in NET patients. In recent studies, other epigenetic regulators affecting SST signalling or SSA–mTOR inhibitor combination therapy in NETs have been considered as novel strategies for tumour control. In conclusion, SST signalling is a relevant regulator of NET functionality. Alongside classical SSA treatment regimens, future advanced therapies and treatment modalities are expected to improve the disease outcomes and overall health of NET patients.
Collapse
|
6
|
Antitumoral and Anti-inflammatory Roles of Somatostatin and Its Analogs in Hepatocellular Carcinoma. Anal Cell Pathol (Amst) 2021; 2021:1840069. [PMID: 34873567 PMCID: PMC8643256 DOI: 10.1155/2021/1840069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and affects about 8% of cirrhotic patients, with a recurrence rate of over 50%. There are numerous therapies available for the treatment of HCC, depending on cancer staging and condition of the patient. The complexity of the treatment is also justified by the unique pathogenesis of HCC that involves intricate processes such as chronic inflammation, fibrosis, and multiple molecular carcinogenesis events. During the last three decades, multiple in vivo and in vitro experiments have used somatostatin and its analogs (SSAs) to reduce the proliferative and metastatic potential of hepatoma cells by inducing their apoptosis and reducing angiogenesis and the inflammatory component of HCC. Most experiments have proven successful, revealing several different pathways and mechanisms corresponding to the aforementioned functions. Moreover, a correlation between specific effects and expression of somatostatin receptors (SSTRs) was observed in the studied cells. Clinical trials have tested either somatostatin or an analog, alone or in combination with other drugs, to explore the potential effects on HCC patients, in various stages of the disease. While the majority of these clinical trials exhibited minor to moderate success, some other studies were inconclusive or even reported negative outcomes. A complete evaluation of the efficacy of somatostatin and SSAs is still the matter of intense debate, and, if deemed useful, these substances may play a beneficial role in the management of HCC patients.
Collapse
|
7
|
Goltstein LCMJ, Grooteman KV, Rocco A, Holleran G, Frago S, Salgueiro PS, Aparicio T, Scaglione G, Chetcuti Zammit S, Prados-Manzano R, Benamouzig R, Nardone G, McNamara D, Benallaoua M, Michopoulos S, Sidhu R, Kievit W, Drenth JPH, van Geenen EJM. Effectiveness and predictors of response to somatostatin analogues in patients with gastrointestinal angiodysplasias: a systematic review and individual patient data meta-analysis. Lancet Gastroenterol Hepatol 2021; 6:922-932. [PMID: 34508668 DOI: 10.1016/s2468-1253(21)00262-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastrointestinal angiodysplasias are vascular malformations that often cause red blood cell transfusion-dependent anaemia. Several studies suggest that somatostatin analogues might decrease rebleeding rates, but the true effect size is unknown. We therefore aimed to investigate the efficacy of somatostatin analogues on red blood cell transfusion requirements of patients with gastrointestinal angiodysplasias and to identify subgroups that might benefit the most from somatostatin analogue therapy. METHODS We did a systematic review and individual patient data meta-analysis. We searched MEDLINE, Embase, and Cochrane on Jan 15, 2016, with an updated search on April 25, 2021. All published randomised controlled trials and cohort studies that reported on somatostatin analogue therapy in patients with gastrointestinal angiodysplasias were eligible for screening. We excluded studies without original patient data, single case reports, small case series (ie, <10 participants), studies in which patients had a specific aetiology of gastrointestinal angiodysplasias, and studies in which somatostatin analogue therapy was initiated simultaneously with other treatment modalities. Authors of eligible studies were invited to share individual patient data. Aggregated data was used if individual patient data were not provided. The primary outcome was the mean reduction in the number of red blood cell transfusions during somatostatin analogue therapy, compared with baseline, expressed as the incidence rate ratio (IRR) and absolute mean decrease. We defined patients as either good responders (≥50% reduction in the number of red blood cell transfusions) or poor responders (<50% reduction). A mixed-effects negative binomial regression was used to account for clustering of patients and skewness in data. This study was registered in the International Prospective Register of Systematic Reviews (PROSPERO), number CRD42020213985. FINDINGS We identified 11 eligible studies (one randomised controlled trial and ten cohort studies) of moderate-to-high quality and obtained individual patient data from the authors of nine (82%) studies. The remaining two (18%) studies provided sufficient information in the published manuscript to extract individual patient data. In total, we analysed data from 212 patients. Somatostatin analogues reduced the number of red blood cell transfusions with an IRR of 0·18 (95% CI 0·14-0·24; p<0·0001) during a median treatment duration of 12 months (IQR 6·0-12·0) and follow-up period of 12 months (12·0-12·0), correlating with a mean absolute decrease in the number of red blood cell transfusions from 12·8 (95% CI 10·4-15·8) during baseline to 2·3 (1·9-2·9) during follow-up-ie, a reduction of 10·5 red blood cell transfusions (p<0·0001). 177 (83%) of 212 patients had a good response to somatostatin analogue therapy (defined as at least a 50% reduction in the number of red blood cell transfusions). Heterogeneity across studies was moderate (I2=53%; p=0·02). Location of gastrointestinal angiodysplasias in the stomach compared with angiodysplasias in the small bowel and colon (IRR interaction 1·92 [95% CI 1·13-3·26]; p=0·02) was associated with worse treatment response. Octreotide was associated with a better treatment response than lanreotide therapy (IRR interaction 2·13 [95% CI 1·12-4·04]; p=0·02). The certainty of evidence was high for the randomised controlled trial and low for the ten cohort studies. Adverse events occurred in 38 (18%) of 212 patients receiving somatostatin analogue therapy, with ten (5%) discontinuing this therapy because of adverse events. The most common adverse events were loose stools (seven [3%] of 212), cholelithiasis (five [2%]), flatulence (four [2%]), and administration site reactions (erythema, five [2%]). INTERPRETATION Somatostatin analogue therapy is safe and effective in most patients with red blood cell transfusion-dependent bleeding due to gastrointestinal angiodysplasias. Somatostatin analogue therapy is more effective in patients with angiodysplasias located in the small bowel and colon, and octreotide therapy seems to be more effective than lanreotide therapy. FUNDING The Netherlands Organisation for Health Research and Development and the Radboud University Medical Center.
Collapse
Affiliation(s)
- Lia C M J Goltstein
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands.
| | - Karina V Grooteman
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alba Rocco
- Department of Clinical Medicine and Surgery, Gastroenterology Unit, University Federico II, Naples, Italy
| | - Grainne Holleran
- Department of Clinical Medicine, Trinity College Dublin, Tallaght Hospital, Dublin, Ireland
| | - Santiago Frago
- Department of Digestive Diseases, Miguel Servet University Hospital, Zaragoza, Spain
| | - Paulo S Salgueiro
- Gastroenterology Department, Centro Hospitalar Universitário do Porto-Hospital de Santo António, Porto, Portugal
| | - Thomas Aparicio
- Department of Gastroenterology, Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France
| | | | | | - Raul Prados-Manzano
- Department of Gastroenterology, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - Robert Benamouzig
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Bobigny, France
| | - Gerardo Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology Unit, University Federico II, Naples, Italy
| | - Deirdre McNamara
- Department of Clinical Medicine, Trinity College Dublin, Tallaght Hospital, Dublin, Ireland
| | - Mourad Benallaoua
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Bobigny, France
| | | | - Reena Sidhu
- Academic Department of Gastroenterology, Royal Hallamshire Hospital, Sheffield, UK
| | - Wietske Kievit
- Radboud Institute for Health Science, Department of Health Evidence, Radboud University Medical Center, Nijmegen, Netherlands
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Erwin J M van Geenen
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
8
|
Wang X, Luo D, Basilion JP. Photodynamic Therapy: Targeting Cancer Biomarkers for the Treatment of Cancers. Cancers (Basel) 2021; 13:cancers13122992. [PMID: 34203805 PMCID: PMC8232794 DOI: 10.3390/cancers13122992] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Photodynamic therapy (PDT) is a minimally invasive treatment option that can kill cancerous cells by subjecting them to light irradiation at a specific wavelength. The main problem related to most photosensitizers is the lack of tumor selectivity, which leads to undesired uptake in normal tissues resulting in side effects. Passive targeting and active targeting are the two strategies to improve uptake in tumor tissues. This review focused on active targeting and summarizes recent active targeting approaches in which highly potent photosensitizers are rendered tumor-specific by means of an appended targeting moiety that interacts with a protein unique to, or at least significantly more abundant on, tumor cell surfaces compared to normal cells. Abstract Photodynamic therapy (PDT) is a well-documented therapy that has emerged as an effective treatment modality of cancers. PDT utilizes harmless light to activate non- or minimally toxic photosensitizers to generate cytotoxic species for malignant cell eradication. Compared with conventional chemotherapy and radiotherapy, PDT is appealing by virtue of the minimal invasiveness, its safety, as well as its selectivity, and the fact that it can induce an immune response. Although local illumination of the cancer lesions renders intrinsic selectivity of PDT, most photosensitizers used in PDT do not display significant tumor tissue selectivity. There is a need for targeted delivery of photosensitizers. The molecular identification of cancer antigens has opened new possibilities for the development of effective targeted therapy for cancer patients. This review provides a brief overview of recent achievements of targeted delivery of photosensitizers to cancer cells by targeting well-established cancer biomarkers. Overall, targeted PDT offers enhanced intracellular accumulation of the photosensitizer, leading to improved PDT efficacy and reduced toxicity to normal tissues.
Collapse
Affiliation(s)
- Xinning Wang
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH 44106, USA
- Correspondence: (X.W.); (J.P.B.); Tel.: +216-844-4848 (X.W.); +216-983-3246 (J.P.B.); Fax: +216-844-4987 (X.W. & J.P.B.)
| | - Dong Luo
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-44, Cleveland, OH 44106, USA;
| | - James P. Basilion
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH 44106, USA
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-44, Cleveland, OH 44106, USA;
- Correspondence: (X.W.); (J.P.B.); Tel.: +216-844-4848 (X.W.); +216-983-3246 (J.P.B.); Fax: +216-844-4987 (X.W. & J.P.B.)
| |
Collapse
|
9
|
Sakthivel P, Kumar R, Arunraj ST, Bhalla AS, Prashanth A, Kumar R, Sharma SC, Thakar A. 68 Ga DOTANOC PET/CT Scan in Primary Juvenile Nasopharyngeal Angiofibroma - A Pilot Study. Laryngoscope 2020; 131:1509-1515. [PMID: 33355921 DOI: 10.1002/lary.29332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/25/2020] [Accepted: 12/03/2020] [Indexed: 11/11/2022]
Abstract
OBJECTIVES/HYPOTHESIS Somatostatin receptors (SSTRs) are highly expressed in neuroendocrine tumors and is exploited for its imaging and treatment. SSTRs expression is also demonstrated in diverse benign and malignant tumor cell types and proliferating peri-tumoral vessels. Similarly, Juvenile Nasopharyngeal Angiofibroma (JNA) expresses different SSTRs and may be utilized for its imaging and treatment using DOTA, 1-Nal3-octreotide (DOTANOC)-PET/CT scan. STUDY DESIGN Prospective cohort. METHODS Nineteen clinico-radiologically diagnosed primary JNA patients underwent a 68 Ga-DOTANOC PET-CT scan. Using a dedicated PET/CT scanner, a low-dose head and neck spot CT scan was performed after 45 to 60 minutes of intravenous injection of 2 to 3 mCi(74-111 MBq) of DOTANOC. The primary objective was to assess the intensity and pattern of DOTANOC uptake in these patients. RESULTS DOTANOC expression was noted in all cases (n = 19) of primary JNA (100%). The mean (SD) DOTANOC SUVmax ratio of tumor and background was 6.9+/-1.4(range, 3.8-9.5). Intra-cranial extension in all 13/19 patients was prominently visualized due to the absence of DOTANOC uptake in the brain. Compared to the background all stages of JNA showed significant DOTANOC uptake (P < .0001). No difference in uptake between advanced-stage tumors and early tumors was noted (P = .47). A statistically non-significant negative trend was noted for decreasing uptake with increasing age (Spearman correlation coefficient, r = -0.19). CONCLUSIONS This first study of 68 Ga-DOTANOC-PET/CT scan in JNA demonstrates consistent and reliable uptake activity in all patients irrespective of age and stage. This opens up possibilities to physiological diagnostic imaging with a promise of greater specificity and sensitivity and may have applications in ambivalent diagnostic situations such as the detection of recurrence. LEVEL OF EVIDENCE 3 Laryngoscope, 131:1509-1515, 2021.
Collapse
Affiliation(s)
- Pirabu Sakthivel
- Department of Otorhinolaryngology & Head and Neck Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Department of Nuclear medicine, All India Institute of Medical Sciences, New Delhi, India
| | | | - Ashu Seith Bhalla
- Department of Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Arun Prashanth
- Department of Nuclear medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Department of Otorhinolaryngology & Head and Neck Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Suresh Chandra Sharma
- Department of Otorhinolaryngology & Head and Neck Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Alok Thakar
- Department of Otorhinolaryngology & Head and Neck Surgery, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
10
|
Danalev D, Borisova D, Yaneva S, Georgieva M, Balacheva A, Dzimbova T, Iliev I, Pajpanova T, Zaharieva Z, Givechev I, Naydenova E. Synthesis, in vitro biological activity, hydrolytic stability and docking of new analogs of BIM-23052 containing halogenated amino acids. Amino Acids 2020; 52:1581-1592. [PMID: 33215308 DOI: 10.1007/s00726-020-02915-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/06/2020] [Indexed: 11/25/2022]
Abstract
One of the potent somatostatin analogs, BIM-23052 (DC-23-99) D-Phe-Phe-Phe-D-Trp-Lys-Thr-Phe-Thr-NH2, has established in vitro growth hormone inhibitory activity in nM concentrations. It is also characterized by high affinity to some somatostatin receptors which are largely distributed in the cell membranes of many tumor cells. Herein, we report the synthesis of a series of analogs of BIM-23052 containing halogenated Phe residues using standard solid-phase peptide method Fmoc/OtBu-strategy. The cytotoxic effects of the compounds were tested in vitro against two human tumor cell lines-breast cancer cell line and hepatocellular cancer cell line, as well as on human non-tumorigenic epithelial cell line. Analogs containing fluoro-phenylalanines are cytotoxic in μM range, as the analog containing Phe (2-F) showed better selectivity against human hepatocellular cancer cell line. The presented study also reveals that accumulation of halogenated Phe residues does not increase the cytotoxicity according to tested cell lines. The calculated selective index reveals different mechanisms of antitumor activity of the parent compound BIM-23052 and target halogenated analogs for examined breast tumor cell lines. All peptides tested have high antitumor activity against the HepG2 cell line (IC50 ≈ 100 μM and SI > 5) compared to breast cells. This is probably due to the high permeability of the cell membrane and the higher metabolic activity of hepatocytes. In silico docking studies confirmed that all obtained analogs bind well with the somatostatin receptors with preference to ssrt3 and ssrt5. All target compounds showed high hydrolytic stability at acid and neutral pH, which mimic physiological condition in stomach and human plasma.
Collapse
Affiliation(s)
- Dancho Danalev
- Biotechnology Department, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | - Desislava Borisova
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | - Spaska Yaneva
- Department of Fundamental of Chemical Technology, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | - Maya Georgieva
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Anelia Balacheva
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tatyana Dzimbova
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, Sofia, Bulgaria.,South-West University "Neofit Rilski", Blagoevgrad, Bulgaria
| | - Ivan Iliev
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev str., bl. 25, 1113, Sofia, Bulgaria
| | - Tamara Pajpanova
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Zdravka Zaharieva
- Biotechnology Department, University of Chemical Technology and Metallurgy, Sofia, Bulgaria.,Testing Center Global Test Ltd, 31 Krushovski vrah Street, Sofia, Bulgaria
| | - Ivan Givechev
- Biotechnology Department, University of Chemical Technology and Metallurgy, Sofia, Bulgaria.,Testing Center Global Test Ltd, 31 Krushovski vrah Street, Sofia, Bulgaria
| | - Emilia Naydenova
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia, Bulgaria.
| |
Collapse
|
11
|
Suzuki T, Tanaka M, Sasaki M, Ichikawa H, Nishie H, Kataoka H. Vascular Shutdown by Photodynamic Therapy Using Talaporfin Sodium. Cancers (Basel) 2020; 12:cancers12092369. [PMID: 32825648 PMCID: PMC7563359 DOI: 10.3390/cancers12092369] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022] Open
Abstract
Photodynamic therapy (PDT) is an attractive cancer treatment modality. Talaporfin sodium, a second-generation photosensitizer, results in lower systemic toxicity and relatively better selective tumor destruction than first-generation photosensitizers. However, the mechanism through which PDT induces vascular shutdown is unclear. In this study, the in vitro effects of talaporfin sodium-based PDT on human umbilical vein endothelial cells (HUVECs) were determined through cell viability and endothelial tube formation assays, and evaluation of the tubulin and F-actin dynamics and myosin light chain (MLC) phosphorylation. Additionally, the effects on tumor blood flow and tumor vessel destruction were assessed in vivo. In the HUVECs, talaporfin sodium-based PDT induced endothelial tube destruction and microtubule depolymerization, triggering the formation of F-actin stress fibers and a significant increase in MLC phosphorylation. However, pretreatment with the Rho-associated protein kinase (ROCK) inhibitor, Y27632, completely prevented PDT-induced stress fiber formation and MLC phosphorylation. The in vivo analysis and pathological examination revealed that the PDT had significantly decreased the tumor blood flow and the active area of the tumor vessel. We concluded that talaporfin sodium-based PDT induces the shutdown of existing tumor vessels via the RhoA/ROCK pathway by activating the Rho-GTP pathway and decreasing the tumor blood flow.
Collapse
Affiliation(s)
| | - Mamoru Tanaka
- Correspondence: ; Tel.: +81-52-853-8211; Fax: +81-52-852-0952
| | | | | | | | | |
Collapse
|
12
|
Molecular and Cellular Mechanisms Underlying Somatostatin-Based Signaling in Two Model Neural Networks, the Retina and the Hippocampus. Int J Mol Sci 2019; 20:ijms20102506. [PMID: 31117258 PMCID: PMC6566141 DOI: 10.3390/ijms20102506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
Neural inhibition plays a key role in determining the specific computational tasks of different brain circuitries. This functional "braking" activity is provided by inhibitory interneurons that use different neurochemicals for signaling. One of these substances, somatostatin, is found in several neural networks, raising questions about the significance of its widespread occurrence and usage. Here, we address this issue by analyzing the somatostatinergic system in two regions of the central nervous system: the retina and the hippocampus. By comparing the available information on these structures, we identify common motifs in the action of somatostatin that may explain its involvement in such diverse circuitries. The emerging concept is that somatostatin-based signaling, through conserved molecular and cellular mechanisms, allows neural networks to operate correctly.
Collapse
|
13
|
Abstract
Neuroendocrine tumors, including carcinoids, are rare and insidiously growing tumors. Related to their site of origin, tumors can be functional, causing various forms of the carcinoid syndrome, owing to the overproduction of serotonin, histamine, or other bioactive substances. They often invade adjacent structures or metastasize to the liver and elsewhere. Treatment includes multimodal approaches, including cytoreductive surgery, locoregional embolization, cytotoxic therapy, peptide receptor radionuclide therapy, and various targeted therapies with goals of symptom relief and control of tumor growth. This article summarizes current and emerging approaches to management and reviews several promising future therapies.
Collapse
Affiliation(s)
- Paul Benjamin Loughrey
- Department of Ophthalmology, Royal Victoria Hospital, Belfast Trust, Grosvenor Road, Belfast, BT12 6BA, UK; Department of Endocrinology and Diabetes, Royal Victoria Hospital, Belfast Trust, Grosvenor Road, Belfast, BT12 6BA, UK
| | - Dongyun Zhang
- Department of Medicine, David Geffen School of Medicine, University of California, 700 Tiverton Avenue, Los Angeles, CA 90095, USA
| | - Anthony P Heaney
- Department of Medicine, David Geffen School of Medicine, University of California, 700 Tiverton Avenue, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine, University of California, 700 Tiverton Avenue, Los Angeles, CA 90095, USA.
| |
Collapse
|
14
|
Enzler T, Fojo T. Long-acting somatostatin analogues in the treatment of unresectable/metastatic neuroendocrine tumors. Semin Oncol 2017; 44:141-156. [PMID: 28923213 DOI: 10.1053/j.seminoncol.2017.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neuroendocrine tumors (NETs) are a relatively rare and heterogeneous group of neoplasms with an annual incidence of ~35 cases per 100,000 people in the United States. The updated World Health Organization (WHO) classification system of gastroenteropancreatic (GEP)-NETs categorizes these tumors according to site of origin, clinical syndrome, and degree of differentiation. Well-differentiated NETs arising from the gastrointestinal tract or lungs (formerly known as carcinoid tumors) are often indolent and slow-growing. In contrast, poorly differentiated neuroendocrine carcinomas (NECs) are aggressive and have a poor prognosis. Due to their insidious onset, most NETs are diagnosed at an advanced stage and a curative approach is not possible. In these patients, medical therapy is limited to disease control, including relief of symptoms that arise from overproduction of peptide hormones by the tumors. Somatostatin analogues (SSAs) have remained the mainstay of symptoms control. In addition to symptoms control, clinical data also support an anti-proliferative effect of SSAs in patients with well- to moderately differentiated NETs. Long-acting SSAs have greatly facilitated their use. This review will focus on two long-acting SSAs, octreotide LAR and lanreotide, and their use in the clinical setting. Information necessary to assess their relative merits is summarized. We conclude these two therapies are interchangeable making value a very important consideration in their use.
Collapse
Affiliation(s)
- Thomas Enzler
- Department of Medicine, Division of Hematology Oncology, Columbia University, New York, NY
| | - Tito Fojo
- Department of Medicine, Division of Hematology Oncology, Columbia University, New York, NY; James J. Peter VAMC, Bronx, NY.
| |
Collapse
|
15
|
Öberg K, Lamberts SWJ. Somatostatin analogues in acromegaly and gastroenteropancreatic neuroendocrine tumours: past, present and future. Endocr Relat Cancer 2016; 23:R551-R566. [PMID: 27697899 DOI: 10.1530/erc-16-0151] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/03/2016] [Indexed: 11/08/2022]
Abstract
Acromegaly is a hormonal disorder that arises when the pituitary gland secretes excess growth hormone (GH), which in turn stimulates a concomitant increase in serum insulin-like growth factor 1 (IGF-1) levels. Gastroenteropancreatic neuroendocrine tumours (GEP-NET) constitute a heterogeneous group of tumours that can secrete serotonin and a variety of peptide hormones that may cause characteristic symptoms known as carcinoid syndrome or other symptoms and hormonal hypersecretion syndromes depending on the tumour's site of origin. Current medical therapy for the treatment of acromegaly and GEP-NET involves the administration of somatostatin analogues that effectively suppress excess hormone secretion. After its discovery in 1979, octreotide became the first synthetic biologically stable somatostatin analogue with a short-acting formulation of octreotide introduced into clinical practice in the late 1980s. Lanreotide, another somatostatin analogue, became available in the mid-1990s initially as a prolonged-release formulation administered every 10 or 14 days. Long-acting release formulations of both octreotide (Sandostatin LAR and Novartis) and lanreotide (Somatuline Autogel, Ipsen), based on microparticle and nanoparticle drug-delivery technologies, respectively, were later developed, which allowed for once-monthly administration and improved convenience. First-generation somatostatin analogues remain one of the cornerstones of medical therapy in the management of pituitary and GEP-NET hormone hypersecretion, with octreotide having the longest established efficacy and safety profile of the somatostatin analogue class. More recently, pasireotide (Signifor), a next-generation multireceptor-targeted somatostatin analogue, has emerged as an alternative therapeutic option for the treatment of acromegaly. This review summarizes the development and clinical success of somatostatin analogues.
Collapse
|
16
|
Lequoy M, Desbois-Mouthon C, Wendum D, Gupta V, Blachon JL, Scatton O, Dumont S, Bonnemaire M, Schmidlin F, Rosmorduc O, Fartoux L. Somatostatin receptors in resected hepatocellular carcinoma: status and correlation with markers of poor prognosis. Histopathology 2016; 70:492-498. [DOI: 10.1111/his.13034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Marie Lequoy
- Department of Hepatology; AP-HP; Saint-Antoine Hospital; Paris France
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
| | | | - Dominique Wendum
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Pathology; AP-HP; Saint-Antoine Hospital; Paris France
| | - Vandana Gupta
- Oncology and Biomarkers; Ipsen Biosci. Inc.; Cambridge MA USA
| | | | - Olivier Scatton
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Hepatobiliary Surgery; AP-HP; Pitié Salpêtrière Hospital; Paris France
| | - Sylvie Dumont
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Pathology; AP-HP; Saint-Antoine Hospital; Paris France
| | | | | | - Olivier Rosmorduc
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Hepatology; AP-HP; Pitié Salpêtrière Hospital; Paris France
| | - Laetitia Fartoux
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Hepatology; AP-HP; Pitié Salpêtrière Hospital; Paris France
| |
Collapse
|
17
|
Synthetic Peptide Analogs of Somatostatin: Trends in the Synthesis of and Prospects in the Search for New Anticancer Drugs. Pharm Chem J 2015. [DOI: 10.1007/s11094-015-1284-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
SAKAI N, YAMASHITA S, TAKEHARA Y, SAKAHARA H, BABA S, OKI Y, TAKAHASHI G, KOIZUMI S, SAMESHIMA T, NAMBA H. Evaluation of the Antiangiogenic Effects of Octreotide on Growth Hormone-producing Pituitary Adenoma using Arterial Spin-labeling Perfusion Imaging. Magn Reson Med Sci 2015; 14:73-6. [DOI: 10.2463/mrms.2014-0026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Naoto SAKAI
- Department of Neurosurgery, Hamamatsu University School of Medicine
| | - Shuhei YAMASHITA
- Department of Radiology, Hamamatsu University School of Medicine
| | | | - Harumi SAKAHARA
- Department of Radiology, Hamamatsu University School of Medicine
| | - Satoshi BABA
- Department of Diagnostic Pathology, Hamamatsu University School of Medicine
| | - Yutaka OKI
- Second Department of Internal Medicine, Division of Endocrinology and Metabolism, Hamamatsu University School of Medicine
| | - Goro TAKAHASHI
- Department of Otorhinolaryngology, Division of Head and Neck Surgery, Hamamatsu University School of Medicine
| | | | | | - Hiroki NAMBA
- Department of Neurosurgery, Hamamatsu University School of Medicine
| |
Collapse
|
19
|
Kaščáková S, Hofland LJ, De Bruijn HS, Ye Y, Achilefu S, van der Wansem K, van der Ploeg-van den Heuvel A, van Koetsveld PM, Brugts MP, van der Lelij AJ, Sterenborg HJCM, ten Hagen TLM, Robinson DJ, van Hagen MP. Somatostatin analogues for receptor targeted photodynamic therapy. PLoS One 2014; 9:e104448. [PMID: 25111655 PMCID: PMC4128677 DOI: 10.1371/journal.pone.0104448] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/08/2014] [Indexed: 12/15/2022] Open
Abstract
Photodynamic therapy (PDT) is an established treatment modality, used mainly for anticancer therapy that relies on the interaction of photosensitizer, light and oxygen. For the treatment of pathologies in certain anatomical sites, improved targeting of the photosensitizer is necessary to prevent damage to healthy tissue. We report on a novel dual approach of targeted PDT (vascular and cellular targeting) utilizing the expression of neuropeptide somatostatin receptor (sst2) on tumor and neovascular-endothelial cells. We synthesized two conjugates containing the somatostatin analogue [Tyr3]-octreotate and Chlorin e6 (Ce6): Ce6-K3-[Tyr3]-octreotate (1) and Ce6-[Tyr3]-octreotate-K3-[Tyr3]-octreotate (2). Investigation of the uptake and photodynamic activity of conjugates in-vitro in human erythroleukemic K562 cells showed that conjugation of [Tyr3]-octreotate with Ce6 in conjugate 1 enhances uptake (by a factor 2) in cells over-expressing sst2 compared to wild-type cells. Co-treatment with excess free Octreotide abrogated the phototoxicity of conjugate 1 indicative of a specific sst2-mediated effect. In contrast conjugate 2 showed no receptor-mediated effect due to its high hydrophobicity. When compared with un-conjugated Ce6, the PDT activity of conjugate 1 was lower. However, it showed higher photostability which may compensate for its lower phototoxicity. Intra-vital fluorescence pharmacokinetic studies of conjugate 1 in rat skin-fold observation chambers transplanted with sst2+ AR42J acinar pancreas tumors showed significantly different uptake profiles compared to free Ce6. Co-treatment with free Octreotide significantly reduced conjugate uptake in tumor tissue (by a factor 4) as well as in the chamber neo-vasculature. These results show that conjugate 1 might have potential as an in-vivo sst2 targeting photosensitizer conjugate.
Collapse
Affiliation(s)
- Slávka Kaščáková
- Center for Optical Diagnostics and Therapy, Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Leo J. Hofland
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Henriette S. De Bruijn
- Department of Otolaryngology and Head & Neck Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Yunpeng Ye
- Department of Radiology, School of Medicine, Washington University, St. Louis, Missouri, United States of America
| | - Samuel Achilefu
- Department of Radiology, School of Medicine, Washington University, St. Louis, Missouri, United States of America
| | | | | | | | - Michael P. Brugts
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Henricus J. C. M. Sterenborg
- Center for Optical Diagnostics and Therapy, Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Timo L. M. ten Hagen
- Department of Surgical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dominic J. Robinson
- Department of Otolaryngology and Head & Neck Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Martin P. van Hagen
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
20
|
Vilar L, Fleseriu M, Naves LA, Albuquerque JL, Gadelha PS, dos Santos Faria M, Nascimento GC, Montenegro RM, Montenegro RM. Can we predict long-term remission after somatostatin analog withdrawal in patients with acromegaly? Results from a multicenter prospective trial. Endocrine 2014; 46:577-84. [PMID: 24272601 DOI: 10.1007/s12020-013-0094-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 10/17/2013] [Indexed: 10/26/2022]
Abstract
Somatostatin analogs (SSAs) represent the mainstay of therapy in acromegaly. One of the potential disadvantages is the expected need to maintain therapy indefinitely in previously non-irradiated patients. The aim of this multicenter prospective open trial was to evaluate the likelihood of successful discontinuation of SSA therapy in well-controlled acromegalic patients who fulfilled very strict criteria: two or more years of treatment with the long-acting SSA octreotide LAR (OCT-LAR), a stable dose and injections interval every 4 weeks or longer for the previous year, GH levels <2.5 ng/ml and normal IGF-1 levels for age, a tumor remnant <10 mm, no history of radiotherapy, and no use of cabergoline or pegvisomant over the previous 6 months. Disease recurrence was defined as an increase of IGF-1 to levels above 1.2-fold the upper limit of normal (ULN). Out of 220 patients, 20 patients (12 women and 8 men; mean age, 48.1 ± 10.3 years; age range, 27-64) treated for 2.74 ± 0.64 years (range, 2.0-4.4) were included in this prospective study and OCT-LAR therapy was stopped. Four patients (20 %) remained without clinical and biochemical/neuroradiological evidence of disease recurrence after 12-18 months of follow-up. Sixteen patients (80 %) relapsed biochemically within 9 months after drug withdrawal and restarted OCT-LAR at the same previous dose. Compared to recurring subjects, non-recurring patients had significantly lower mean IGF-1 (× ULN) levels but there were some overlapping values in both groups. No other characteristic could be identified as a predictor of successful OCT-LAR discontinuation. Our findings demonstrated that OCT-LAR withdrawal, though rare, is possible in well-selected acromegalic patients treated for at least 2 years and considered optimally controlled in hormonal and neuroradiological terms.
Collapse
Affiliation(s)
- Lucio Vilar
- Division of Endocrinology, Hospital das Clínicas, Federal University of Pernambuco, Rua Clovis Silveira Barros, 84/1202, Boa Vista, Recife, CEP 50050-270, Brazil,
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abdel-Rahman O, Lamarca A, Valle JW, Hubner RA. Somatostatin receptor expression in hepatocellular carcinoma: prognostic and therapeutic considerations. Endocr Relat Cancer 2014; 21:R485-93. [PMID: 25336571 DOI: 10.1530/erc-14-0389] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sorafenib is the only systemic therapy to demonstrate a significant survival benefit over supportive care in robust randomised controlled trials for advanced hepatocellular carcinoma (HCC). In the context of an intense search for prognostic and predictive factors for response and efficacy of different systemic therapies (including sorafenib), a number of molecular targets have been identified, paving new avenues for potential therapeutic opportunities. Such molecular targets include somatostatin receptor (SSTR)-related alterations. In this review, we provide an overview of the various considerations relating to SSTRs as potentially novel prognostic and predictive biomarkers for HCC with special emphasis on the therapeutic potential of somatostatin analogues in HCC management.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology DepartmentFaculty of Medicine, Ain Shams University, Cairo, EgyptDepartment of Medical OncologyThe Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UKUniversity of ManchesterManchester Academic Health Sciences Centre (MAHSC), Manchester, UK
| | - Angela Lamarca
- Clinical Oncology DepartmentFaculty of Medicine, Ain Shams University, Cairo, EgyptDepartment of Medical OncologyThe Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UKUniversity of ManchesterManchester Academic Health Sciences Centre (MAHSC), Manchester, UK
| | - Juan W Valle
- Clinical Oncology DepartmentFaculty of Medicine, Ain Shams University, Cairo, EgyptDepartment of Medical OncologyThe Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UKUniversity of ManchesterManchester Academic Health Sciences Centre (MAHSC), Manchester, UK Clinical Oncology DepartmentFaculty of Medicine, Ain Shams University, Cairo, EgyptDepartment of Medical OncologyThe Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UKUniversity of ManchesterManchester Academic Health Sciences Centre (MAHSC), Manchester, UK
| | - Richard A Hubner
- Clinical Oncology DepartmentFaculty of Medicine, Ain Shams University, Cairo, EgyptDepartment of Medical OncologyThe Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UKUniversity of ManchesterManchester Academic Health Sciences Centre (MAHSC), Manchester, UK
| |
Collapse
|
22
|
Chéret J, Lebonvallet N, Carré JL, Misery L, Le Gall-Ianotto C. Role of neuropeptides, neurotrophins, and neurohormones in skin wound healing. Wound Repair Regen 2013; 21:772-88. [PMID: 24134750 DOI: 10.1111/wrr.12101] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 07/01/2013] [Indexed: 12/01/2022]
Abstract
Due to the close interactions between the skin and peripheral nervous system, there is increasing evidence that the cutaneous innervation is an important modulator of the normal wound healing process. The communication between sensory neurons and skin cells involves a variety of molecules (neuropeptides, neurohormones, and neurotrophins) and their specific receptors expressed by both neuronal and nonneuronal skin cells. It is well established that neurotransmitters and nerve growth factors released in skin have immunoregulatory roles and can exert mitogenic actions; they could also influence the functions of the different skin cell types during the wound healing process.
Collapse
Affiliation(s)
- Jérémy Chéret
- Laboratory of Neurosciences of Brest (EA4685), University of Western Brittany, Brest, France
| | | | | | | | | |
Collapse
|
23
|
Veenstra MJ, de Herder WW, Feelders RA, Hofland LJ. Targeting the somatostatin receptor in pituitary and neuroendocrine tumors. Expert Opin Ther Targets 2013; 17:1329-43. [DOI: 10.1517/14728222.2013.830711] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
24
|
Starkey JR, Pascucci EM, Drobizhev MA, Elliott A, Rebane AK. Vascular targeting to the SST2 receptor improves the therapeutic response to near-IR two-photon activated PDT for deep-tissue cancer treatment. Biochim Biophys Acta Gen Subj 2013; 1830:4594-603. [PMID: 23747302 DOI: 10.1016/j.bbagen.2013.05.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 05/11/2013] [Accepted: 05/29/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Broader clinical acceptance of photodynamic therapy is currently hindered by (a) poor depth efficacy, and (b) predisposition towards establishment of an angiogenic environment during the treatment. Improved depth efficacy is being sought by exploiting the NIR tissue transparency window and by photo-activation using two-photon absorption (2PA). Here, we use two-photon activation of PDT sensitizers, untargeted and targeted to SST2 receptors or EGF receptors, to achieve deep tissue treatment. METHODS Human tumor lines, positive or negative for SST2r expression were used, as well as murine 3LL cells and bovine aortic endothelial cells. Expression of SST2 receptors on cancer cells and tumor vasculature was evaluated in vitro and frozen xenograft sections. PDT effects on tumor blood flow were followed using in vivo scanning after intravenous injection of FITC conjugated dextran 150K. Dependence of the PDT efficacy on the laser pulse duration was evaluated. Effectiveness of targeting to vascular SST2 receptors was compared to that of EGF receptors, or no targeting. RESULTS Tumor vasculature stained for SST2 receptors even in tumors from SST2 receptor negative cell lines, and SST2r targeted PDT led to tumor vascular shutdown. Stretching the pulse from ~120fs to ~3ps led to loss of the PDT efficacy especially at greater depth. PDT targeted to SST2 receptors was much more effective than untargeted PDT or PDT targeted to EGF receptors. GENERAL SIGNIFICANCE The use of octreotate to target SST2 receptors expressed on tumor vessels is an excellent approach to PDT with few recurrences and some long term cures.
Collapse
Affiliation(s)
- Jean R Starkey
- Montana State University, Department of Microbiology, Bozeman, MT 59717, USA.
| | | | | | | | | |
Collapse
|
25
|
Keskin O, Yalcin S. A review of the use of somatostatin analogs in oncology. Onco Targets Ther 2013; 6:471-83. [PMID: 23667314 PMCID: PMC3650572 DOI: 10.2147/ott.s39987] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Indexed: 12/12/2022] Open
Abstract
Somatostatin is a neuropeptide produced by paracrine cells that are located throughout the gastrointestinal tract, lung, and pancreas, and is also found in various locations of the nervous system. It exerts neural control over many physiological functions including inhibition of gastrointestinal endocrine secretion through its receptors. Potent and biologically stable analogs of somatostatin have been developed. These somatostatin analogs show different efficacy on different receptors, and receptors are varyingly concentrated in specific tissues. Antitumor and antisecretory effects of somatostatin analogs in cancer have been shown in several in vivo and in vitro studies. However, these activities have not always yielded into clinically relevant patient outcome benefit. Somatostatin analogs are of clinical benefit in treating symptoms of ectopic hormone secretion (adrenocorticotropic hormone, growth hormone-releasing hormone) in lung cancer, without inducing a significant tumor response. They have also been shown to induce a statistically significant decrease in bone pain and increase in Karnofsky performance status in patients with metastatic prostate cancer. Somatostatin analogs alone or in combination with other agents have only limited antitumoral effect in breast cancer. In gastrointestinal cancers, studies have not shown an objective tumor response to somatostatin analogs except in endocrine tumors of the liver with symptomatic and biochemical improvement. In neuroendocrine tumors of the gastrointestinal system and pancreas, very high symptomatic and biochemical response rates have been achieved with somatostatin analogs. Antiproliferative activity has been clearly shown in metastatic midgut neuroendocrine tumors.
Collapse
Affiliation(s)
- Ozge Keskin
- Department of Medical Oncology, Hacettepe University Institute of Cancer, Ankara, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University Institute of Cancer, Ankara, Turkey
| |
Collapse
|
26
|
Ramírez C, Vargas G, González B, Grossman A, Rábago J, Sosa E, Espinosa-de-Los-Monteros AL, Mercado M. Discontinuation of octreotide LAR after long term, successful treatment of patients with acromegaly: is it worth trying? Eur J Endocrinol 2012; 166:21-6. [PMID: 21993154 DOI: 10.1530/eje-11-0738] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Somatostatin analogs (SA) have been used for over 25 years in the treatment of acromegaly. A major disadvantage is the need to continue therapy indefinitely. OBJECTIVE To evaluate the feasibility of discontinuing therapy in well-controlled patients with acromegaly treated chronically with SA. DESIGN AND METHODS Of the 205 subjects on octreotide LAR, we selected those who met the following criteria: two or more years of treatment, a stable dose and injection interval of 20 mg every 8 weeks or longer for the previous year, no history of radiation, no cabergoline for the previous 6 months, a GH <1.5 ng/ml, and an IGF1 <1.2×upper limit of normal (ULN). Octreotide LAR was stopped and both GH and IGF1 were measured monthly for 4 months; a glucose-suppressed GH value and magnetic resonance imaging were obtained at the 4th month, thereafter, basal GH and IGF1 were measured q. 3 months, for 12-18 months. Patients were removed from the study if GH or IGF1 rose to 1.5 ng/ml or 1.2×ULN respectively. RESULTS Twelve patients (ten women, mean age 48±13 years) were studied. Seven patients (58.3%) relapsed biochemically within 1 year of having stopped the SA; two patients relapsed by GH and IGF1 criteria, the remaining five patients kept GH levels within target. Five patients (41.7%) remain in remission after 12 months of follow-up. Non-recurring patients were on longer injection intervals but no other characteristic was associated with a successful withdrawal. CONCLUSION Withdrawal of SA is possible in a small but distinct subset of patients, particularly in those who are very well controlled on relatively low doses administered at long intervals.
Collapse
Affiliation(s)
- Claudia Ramírez
- Endocrinology Service and Experimental Endocrinology Unit, Hospital de Especialidades, CMN S.XXI, IMSS, Aristoteles 68, Polanco 11560, Mexico City, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Lee JS, Park YS, Kwon JT, Nam TK, Lee TJ, Kim JK. Radiological apoplexy and its correlation with acute clinical presentation, angiogenesis and tumor microvascular density in pituitary adenomas. J Korean Neurosurg Soc 2011; 50:281-7. [PMID: 22200007 DOI: 10.3340/jkns.2011.50.4.281] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 07/26/2011] [Accepted: 10/17/2011] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE Pituitary apoplexy is life-threatening clinical syndrome caused by the rapid enlargement of a pituitary tumor due to hemorrhage and/or infarction. The pathogenesis of pituitary apoplexy is not completely understood. We analyzed the magnetic resonance imaging (MRI) of pituitary tumors and subsequently correlated the radiological findings with the clinical presentation. Additionally, immunohistochemistry was also performed to determine whether certain biomarkers are related to radiological apoplexy. METHODS Thirty-four cases of pituitary adenoma were enrolled for retrospective analysis. In this study, the radiological apoplexy was defined as cases where hemorrhage, infarction or cysts were identified on MRI. Acute clinical presentation was defined as the presence of any of the following symptoms: severe sudden onset headache, decreased visual acuity and/or visual field deficit, and acute mental status changes. Angiogenesis was quantified by immunohistochemical expression of fetal liver kinase 1 (Flk-1), neuropilin (NRP) and vascular endothelial growth factor (VEGF) expression, while microvascular density (MVD) was assessed using Endoglin and CD31. RESULTS Clinically, fourteen patients presented with acute symptoms and 20 for mild or none clinical symptoms. Radiologically, fifteen patients met the criteria for radiological apoplexy. Of the fifteen patients with radiologic apoplexy, 9 patients presented acute symptoms whereas of the 19 patient without radiologic apoplexy, 5 patients presented acute symptoms. Of the five biomarkers tracked, only VEGF was found to be positively correlated with both radiological and nonradiological apoplexy. CONCLUSION While pituitary apoplexy is currently defined in cases where clinical symptoms can be histologically confirmed, we contend that cases of radiologically identified pituitary hemorrhages that present with mild or no symptoms should be designated subacute or subclinical apoplexy. VEGF is believed to have a positive correlation with pituitary hemorrhage. Considering the high rate of symptomatic or asymptomatic pituitary tumor hemorrhage, additional studies are needed to detect predictors of the pituitary hemorrhage.
Collapse
Affiliation(s)
- Jung-Sup Lee
- Department of Neurosurgery, College of Medicine, Chung-Ang University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
28
|
Kukwa W, Andrysiak R, Kukwa A, Hubalewska-Dydejczyk A, Gronkiewicz Z, Wojtowicz P, Krolicki L, Wierzchowski W, Grochowski T, Czarnecka AM. 99mTC-octreotide scintigraphy and somatostatin receptor subtype expression in juvenile nasopharyngeal angiofibromas. Head Neck 2011; 33:1739-46. [DOI: 10.1002/hed.21668] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Revised: 09/02/2010] [Accepted: 10/04/2010] [Indexed: 11/08/2022] Open
|
29
|
Walter T, Hommell-Fontaine J, Gouysse G, Pourreyron C, Nejjari M, Villaume K, Causeret S, Hervieu V, Poncet G, Roche C, Scoazec JY. Effects of somatostatin and octreotide on the interactions between neoplastic gastroenteropancreatic endocrine cells and endothelial cells: a comparison between in vitro and in vivo properties. Neuroendocrinology 2011; 94:200-8. [PMID: 21677423 DOI: 10.1159/000328134] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 04/02/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Experimental studies in vitro suggest that somatostatin and some of its analogues used in clinical practice, such as octreotide, may have potent antiangiogenic properties. However, the clinical transposition of these data is difficult. METHODS To address this issue, we designed a comparative study of the effects of somatostatin and octreotide on the interactions between neoplastic endocrine cells and endothelial cells in several in vitro and in vivo experimental models, including primary cultures of human umbilical vein endothelial cells (HUVEC), indirect cocultures between HUVEC and the somatostatin-producing endocrine cell line STC-1, and an animal model of intrahepatic dissemination of STC-1 cells. RESULTS 10(-8)M octreotide markedly inhibited both basal and VEGF-stimulated HUVEC proliferation, had no effect on endothelial cell migration, but inhibited endothelial tubule formation. HUVEC cocultured with the somatostatin- and VEGF-producing STC-1 cells presented a markedly decreased proliferation, a slightly increased motility and an increased capacity of tubule formation; in this system, the inhibition of endothelial cell proliferation was abolished by neutralizing anti-somatostatin but was restored in the presence of anti-VEGF antibodies. This suggests that somatostatin is able to antagonize the effects of VEGF on endothelial cell proliferation but not on endothelial cell sprouting. Finally, no significant effect of octreotide on tumor growth and intratumoral microvascular density was detected in an experimental model of intrahepatic dissemination of STC-1 cells. CONCLUSION The in vitro antiangiogenic effects of somatostatin and its analogues are likely to be efficiently counterbalanced in the tumor microenvironment by the concomitant release of proangiogenic factors like VEGF.
Collapse
Affiliation(s)
- Thomas Walter
- Lyon Cancer Research Center, Team 4, INSERM UMR1052/CNRS UMR5286, Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhao Y, Liu M, Chagnault V, Wang J, Zhang X, Murphy PV. Biological study of a somatostatin mimetic based on the 1-deoxynojrimycin scaffold. Bioorg Med Chem Lett 2011; 21:824-8. [DOI: 10.1016/j.bmcl.2010.11.088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 11/17/2010] [Accepted: 11/18/2010] [Indexed: 12/01/2022]
|
31
|
GPCR somatostatin receptor extracellular loop 2 is a key ectodomain for making subtype-selective antibodies with agonist-like activities in the pancreatic neuroendocrine tumor BON cell line. Pancreas 2010; 39:1155-66. [PMID: 20531241 DOI: 10.1097/mpa.0b013e3181de8c05] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The extracellular loop 2 (ECL2) ectodomain of the G protein-coupled receptor class A is thought to function like an inactivation "lid." We created polyclonal somatostatin receptor ECL2 (anti-SSTR ECL2) antibodies to target this lid and to examine if these antibodies can selectively activate the SSTR. METHODS Western blots and live-cell immunofluorescence microscopy determined anti-SSTR ECL2 antibody receptor binding selectivity. 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay (MTS assay) and cell cycle assay (fluorescence-activated cell sorting) checked for antibody effect on antiproliferation. Nexin assay examined the antibody's ability to induce apoptosis. LANCE cAMP kit (Perkin Elmer) detected antibody-dependent cAMP decrease. Enzyme-linked immunosorbent assay measured antibody effect on suppressing serotonin secretion. Ligand-receptor binding interference assay with the fluorescent somatostatin (FAM-SST) was used to examine antibody interference to SST-SSTR binding. RESULTS Anti-SSTR ECL2 antibodies are SSTR subtype selective and agonist-like, and they suppress cell proliferation via cell cycle arrest and apoptosis. In addition, these antibodies decrease cAMP production and inhibit serotonin secretion. Interestingly, these antibodies do not interfere with SST-SSTR binding. CONCLUSIONS The ECL2 is an important ectodomain for G protein-coupled receptor activation and required for ligand binding selectivity. The anti-SSTR2, anti-SSTR3, and anti-SSTR5 ECL2 antibodies independently inhibited BON proliferation and decreased hormone secretion. Unlike octreotide, our antibodies do not interfere with SST-SSTR binding.
Collapse
|
32
|
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | | |
Collapse
|
33
|
Bornschein J, Drozdov I, Malfertheiner P. Octreotide LAR: safety and tolerability issues. Expert Opin Drug Saf 2010; 8:755-68. [PMID: 19998528 DOI: 10.1517/14740330903379525] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Somatostatin analogues are the cornerstone in therapy of acromegaly and functioning neuroendocrine tumors. Long-acting retard formulations have improved patient survival and contributed considerably to quality of life. The first such compound was octreotide LAR ('long-acting release'), characterized by high affinity to somatostatin receptor subtypes 2 and 5, which has to be injected intramuscularly every 4 weeks. OBJECTIVE The aim was to screen all octreotide LAR-related literature and assess the compound's profile for safety and tolerability. METHODS An extensive literature search has been performed using the MEDLINE database to retrieve data from clinical studies evaluating the efficacy and tolerability of octreotide LAR. RESULTS/CONCLUSION Octreotide LAR is well tolerated; however, diarrhea and gallstone formation were identified as the main adverse events. Impairment of glucose homeostasis was a regular phenomenon, but its occurrence was unpredictable. General side effects such as headache, abdominal discomfort or fatigue were also reported. According to incidental case reports, administration during pregnancy appears to be safe for both mother and child; however, definitive evidence is missing. In addition, octreotide LAR has been evaluated for further indications including treatment of solid tumor entities, due to its antiproliferative effect. Currently, several compounds (lanreotide, SOM230) with a broader receptor spectrum are under evaluation and may improve treatment efficacy and lower incidence of side effects.
Collapse
Affiliation(s)
- Jan Bornschein
- Otto-von-Guericke University of Magdeburg, Department of Gastroenterology, Hepatology and Infectious Diseases, Germany
| | | | | |
Collapse
|
34
|
Feelders RA, Hofland LJ, van Aken MO, Neggers SJ, Lamberts SWJ, de Herder WW, van der Lely AJ. Medical therapy of acromegaly: efficacy and safety of somatostatin analogues. Drugs 2009; 69:2207-26. [PMID: 19852525 DOI: 10.2165/11318510-000000000-00000] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Acromegaly is a chronic disease with signs and symptoms due to growth hormone (GH) excess. The most frequent cause of acromegaly is a GH-producing pituitary adenoma. Chronic GH excess is accompanied by long-term complications of the locomotor (arthrosis) and cardiovascular (atherosclerosis, cardiomyopathy) systems and is, when untreated, associated with an increased mortality. The aim of treatment of acromegaly is to improve symptoms, to achieve local tumour mass control, and to decrease morbidity and mortality. Treatment options include surgery, medical therapy and radiotherapy. Transsphenoidal surgery is the first choice of treatment when a definitive cure can be achieved, particularly in the case of microadenomas and when decompression of surrounding structures (optic chiasm, ophthalmic motor nerves) is indicated. Primary medical therapy has been increasingly applied in recent years, especially when a priori chances of surgical cure are low (because of adenoma size and localization) and in patients with advanced age and/or serious co-morbidity. In addition, preoperative primary medical therapy may result in tumour shrinkage, facilitating tumour resection, and may reduce perioperative complications due to GH excess. Within the spectrum of medical therapy, long-acting somatostatin analogues (somatostatins) are considered as first-line treatment. Treatment with somatostatin analogues results in GH control in approximately 60% of patients. In addition, somatostatin analogues induce tumour shrinkage in 30-50% of patients, particularly when applied as primary therapy. Prolonged treatment with somatostatin analogues appears to be safe and is usually well tolerated. The currently available somatostatin analogues, octreotide and lanreotide, seem to be equally effective; however, this should still be evaluated in prospective, randomized trials evaluating efficacy with respect to GH control and tumour shrinkage. In patients with an insufficient clinical and biochemical response to somatostatin analogues, combination therapy with dopamine receptor agonists or the GH receptor antagonist pegvisomant usually leads to disease control. New developments in the medical therapy of acromegaly include the universal somatostatin receptor agonist pasireotide, which has a broader affinity for all somatostatin receptor (sst) subtypes compared with the currently available somatostatin analogues with preferential affinity for the sst2 receptor, and chimeric compounds that interact with both somatostatin and dopamine receptors with synergizing effects on GH secretion.
Collapse
Affiliation(s)
- Richard A Feelders
- Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
35
|
Colao A, Pivonello R, Di Somma C, Savastano S, Grasso LFS, Lombardi G. Medical therapy of pituitary adenomas: effects on tumor shrinkage. Rev Endocr Metab Disord 2009; 10:111-23. [PMID: 18791829 DOI: 10.1007/s11154-008-9107-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The efficacy of dopamine-agonists (DA) in patients with prolactinomas and that of somatostatin analogues (SSA) in those with GH- and TSH-secreting adenomas is well established. More recently, data are accumulating suggesting a potential therapeutic role of DA also in patients with ACTH-secreting and clinically non-functioning (NFA) pituitary adenomas. This review aims at summarizing published results of DA and SSA on tumor shrinkage in patients with different histotypes of pituitary adenomas. Results of tumor shrinkage are of clinical relevance as tumor size is the one of the most important determinant of surgical outcome. While reduction of tumor size more than 50% of baseline size in macroprolactinomas treated with DA is a frequent finding in patients with GH-secreting adenomas treated with SSA tumor shrinkage only recently is becoming frequent thanks to the availability of depot formulations. Data on tumor shrinkage in patients with TSH-secreting adenomas treated with SSA are limited because of the rarity of these tumors. Very recently, DA have been reported of some efficacy also in patients with ACTH-secreting adenomas but data are still very limited. NFA respond very scantly to both DA and SSA even if receptors targeting these drugs are present. Whether this is due to limited receptor number or alterations of post-receptor pathway is still unknown.
Collapse
Affiliation(s)
- Annamaria Colao
- Department of Molecular & Clinical Endocrinology and Oncology, Federico II University of Naples, Naples, Italy.
| | | | | | | | | | | |
Collapse
|
36
|
Somatostatin analogues as therapeutics in retinal disease. Pharmacol Ther 2009; 122:324-33. [DOI: 10.1016/j.pharmthera.2009.03.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 03/24/2009] [Indexed: 12/30/2022]
|
37
|
Hoffman WH, Jain A, Chen H, Fedarko NS. Matrix extracellular phosphoglycoprotein (MEPE) correlates with serum phosphorus prior to and during octreotide treatment and following excisional surgery in hypophosphatemic linear sebaceous nevus syndrome. Am J Med Genet A 2008; 146A:2164-8. [PMID: 18627046 DOI: 10.1002/ajmg.a.32395] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- William H Hoffman
- Department of Pediatrics, Pediatric Endocrinology, Medical College of Georgia, Augusta, Georgia
| | | | | | | |
Collapse
|
38
|
Impact of different anticancer regimens on biomarkers of angiogenesis in patients with advanced hepatocellular cancer. J Cancer Res Clin Oncol 2008; 135:271-81. [PMID: 18642029 DOI: 10.1007/s00432-008-0443-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 06/18/2008] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Advanced hepatocellular cancer (HCC) is a highly vascularised tumor with limited treatment options. We wanted to evaluate the impact of different treatments on systemic biomarkers linked to angiogenesis. METHODS Two subsequent prospective, randomised, phase-I/II trials in patients with advanced HCC were performed. A total of 38 patients was randomised to a total of 4 regimens consisting of 3 cycles of 4 weeks each: Trial 1 included group 1 receiving octreotide 30 mg im on day 1, and group 2 octreotide 30 mg on day 1 plus Imatinib 400 mg po daily; Trial 2 included group 3 with oxaliplatin on day 1 (60 mg-90 mg/m(2)), and group 4 with oxaliplatin on day 1, 8, 15 (20 mg-30 mg/m(2)) in combination with octreotide 30 mg on day 1 plus imatinib 400 mg po daily. Primary outcome measure was the relative changes in plasma biomarkers over time. RESULTS Time-to-progression and overall survival was not different between the the two study trials. Within group 1-4, the mean relative increase from baseline to week 12 of treatment was 17, 18, 37, and 2% for s-E-selectin; -1, 90, 10, and -9% for VEGF-A; 18, 84, 141, and 74% for PDGF-BB, and 111, 142, 30, and 7% for serum AFP, respectively. CONCLUSIONS The increase of plasma levels for s-E-selectin and PDGF-BB seen in patients receiving chemotherapy alone may reflect activation of angiogenesis. In contrast, low-dose metronomic chemotherapy in combination with anti-angiogenic drugs seems to correlate with the least increase in biomarkers. Imatinib-octreotide temporarily leads to a decrease in PDGF-BB, whereas octreotide alone had no effect on PDGF-BB plasma levels.
Collapse
|
39
|
Lania A, Mantovani G, Spada A. Genetic abnormalities of somatostatin receptors in pituitary tumors. Mol Cell Endocrinol 2008; 286:180-6. [PMID: 17913341 DOI: 10.1016/j.mce.2007.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 08/01/2007] [Accepted: 08/24/2007] [Indexed: 10/22/2022]
Abstract
Somatostatin exerts antisecretive and antiproliferative effects on different endocrine cells by acting through a family of G protein-coupled receptors that includes five subtypes (SST1-5). Normal human pituitary and pituitary adenomas have been shown to express almost all SST subtypes, with the exception of SST4. Consistent with the observation that octreotide and other somatostatin analogs bind to SST2 and SST5 with high affinity, these genes have been screened for quantitative/qualitative abnormalities in tumors removed from patients with poor responsiveness to somatostatin analogs treatment. Data obtained in GH-secreting adenomas suggested that resistance to octreotide was frequently associated with low expression of SST2 mRNA, although other authors failed to confirm this finding. To date, the only mutational change involving SST2 and SST5 is the Arg to Trp substitution in codon 240 of the SST5 gene that was found in one acromegalic patient resistant to octreotide. Similarly, loss of heterozygosis at SST5 gene locus in pituitary adenomas has been described in individual tumors. In recent years, molecular studies investigated the possible association of gene polymorphisms and susceptibility to diseases and/or resistance to drugs. As far as polymorphic variants of SST genes are concerned, a possible role of SST5 C1004T and T-461C alleles in influencing GH and IGF-I levels in patients with acromegaly has been proposed. Nevertheless, polymorphic variants in SST2 and SST5 genes seem to have a minor, if any, role in determining the different responsiveness to somatostatin analogs in patients with acromegaly.
Collapse
Affiliation(s)
- A Lania
- Department of Medical Sciences, University of Milan, Fondazione Ospedale Maggiore IRCCS, Milan, Italy
| | | | | |
Collapse
|
40
|
Cervia D, Casini G, Bagnoli P. Physiology and pathology of somatostatin in the mammalian retina: a current view. Mol Cell Endocrinol 2008; 286:112-22. [PMID: 18242820 DOI: 10.1016/j.mce.2007.12.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Revised: 10/11/2007] [Accepted: 12/12/2007] [Indexed: 12/30/2022]
Abstract
In the retina, peptidergic signalling participates in multiple circuits of visual information processing. The neuropeptide somatostatin (SRIF) is localised to amacrine cells and, in some instances, in a subset of ganglion cells. The variegated expression patterns of SRIF receptors (sst(1)-sst(5)) and the variety of signalling mechanisms activated by retinal SRIF suggest that this peptide may exert multiple actions on retinal neurons and on retinal physiology, although our current understanding reflects a rather complicated picture. SRIF, mostly through sst(2), may act as a positive factor in the retina by regulating retinal homeostasis and protecting neurons against damage. In this respect, SRIF analogues seem to constitute a promising therapeutic arsenal to cure different retinal diseases, as for instance, ischemic and diabetic retinopathies. However, further investigations are needed not only to fully understand the functional role of the SRIF system in the retina but also to exploit new chemical space for drug-like molecules.
Collapse
Affiliation(s)
- Davide Cervia
- Department of Environmental Sciences, University of Tuscia, Viterbo, Italy
| | | | | |
Collapse
|
41
|
Grozinsky-Glasberg S, Grossman AB, Korbonits M. The role of somatostatin analogues in the treatment of neuroendocrine tumours. Mol Cell Endocrinol 2008; 286:238-50. [PMID: 18037561 DOI: 10.1016/j.mce.2007.10.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2007] [Revised: 09/30/2007] [Accepted: 10/10/2007] [Indexed: 02/08/2023]
Abstract
Neuroendocrine tumours belong to a heterogeneous family of neoplasms, originating in endocrine glands (such as the pituitary, parathyroid or the neuroendocrine adrenal glands), in endocrine islets (within the thyroid or pancreas) as well as in endocrine cells dispersed between exocrine cells throughout the digestive or respiratory tracts. The clinical behaviour of neuroendocrine tumours is variable; they may be functioning or not functioning, ranging from well-differentiated slow growing neuroendocrine tumours to poorly differentiated neuroendocrine tumours, which are highly aggressive malignant tumours. The development of somatostatin analogues as important diagnostic and treatment tools have revolutionised the clinical management of patients with neuroendocrine tumours. However, although symptomatic relief and stabilisation of tumour growth for various periods of time are observed in many patients treated with somatostatin analogues, tumour regression is rare. Development of new somatostatin analogues and new drug combination therapies should further improve the clinical management of these patients.
Collapse
Affiliation(s)
- Simona Grozinsky-Glasberg
- Department of Endocrinology, William Harvey Research Institute, Barts and the London, Queen Mary School of Medicine, University of London, London, UK
| | | | | |
Collapse
|
42
|
Adenylyl cyclase/cAMP system involvement in the antiangiogenic effect of somatostatin in the retina. Results from transgenic mice. Neurochem Res 2008; 33:1247-55. [PMID: 18270825 DOI: 10.1007/s11064-007-9576-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Accepted: 12/20/2007] [Indexed: 10/22/2022]
Abstract
Neoangiogenesis is a response to retinal hypoxia that is inhibited by somatostatin (SRIF) through its subtype 2 receptor (sst2). Using a mouse model of hypoxia-induced retinopathy, we investigated whether inhibition of adenylyl cyclase (AC) is involved in SRIF anti-angiogenic actions. Hypoxia increased AC responsiveness in wild type (WT) retinas and in retinas lacking sst2, but not in sst2-overexpressing retinas. Hypoxia also altered AC isoform expression with different patterns depending on sst2 expression level. The AC VII isoform mRNA and protein resulted the most affected. Indeed, in hypoxia AC VII expression was enhanced in WT retinas and it was further increased in sst2-lacking retinas, whereas in sst2 overexpressing retinas the increase of AC VII was lower than in WT retinas. These data suggest an involvement of AC/cAMP in mediating both hypoxia-evoked retinal neoangiogenesis and SRIF protective actions. The AC VII isoform is a candidate to a main role in these mechanisms.
Collapse
|
43
|
Bocci G, Culler MD, Fioravanti A, Orlandi P, Fasciani A, Colucci R, Taylor JE, Sadat D, Danesi R, Del Tacca M. In vitro antiangiogenic activity of selective somatostatin subtype-1 receptor agonists. Eur J Clin Invest 2007; 37:700-8. [PMID: 17696959 DOI: 10.1111/j.1365-2362.2007.01848.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Endothelial cells of human blood vessels (arteries and veins) show high levels of somatostatin subtype-1 receptor (sst(1)). The aim of the present study is to investigate the inhibitory effects of novel somatostatin analogs, highly selective for human sst(1), on in vitro angiogenesis and their modulation of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor-2 (VEGFR-2) expression. MATERIALS AND METHODS Somatostatin analogs BIM-23745 and BIM-23926 were tested for their ability to prevent proliferation and migration of human endothelial HMEC-1 cells, to modulate VEGF and VEGFR-2 expression and to inhibit sprouting of microvessels from cultured human placental vessel explants in fibrin matrix for 28 days. RESULTS The somatostatin sst(1 )receptor-selective agonists, BIM-23745 and BIM-23926 showed a suppression of endothelial proliferation (e.g. 10(-6) M BIM-23475, 40.0 +/- 2.1% vs. 100% of controls; 10(-7) M BIM-23926, 55.3 +/- 3.3% vs. 100% of controls), migration (e.g. 10(-7) M BIM-23475, 35.0 +/- 1.56% vs. 100% of controls; 10(-7) M BIM-23926, 53.7 +/- 1.77% vs. 100% of controls) and microvessel sprouting (e.g. 10(-8) M BIM-23475, 42.8 +/- 5.6% vs. 100% of controls; 10(-7) M BIM-23926, 17.2 +/- 11.8% vs. 100% of controls). A small but significant percentage of cells exposed to BIM-23745 and BIM-23926 for 24 h and for 72 h presented typical apoptotic morphology. Moreover, both the analogs significantly inhibit VEGF and VEGFR-2 gene expression in endothelial cells grown for 144 h in a fibrin matrix and the VEGF secretion in conditioned media. CONCLUSIONS The inhibition of endothelial activities suggests potential therapeutic utility for administration of somatostatin sst(1 )receptor-selective agonists in the proliferative diseases involving angiogenesis.
Collapse
Affiliation(s)
- G Bocci
- University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ribatti D, Conconi MT, Nussdorfer GG. Nonclassic Endogenous Novel Regulators of Angiogenesis. Pharmacol Rev 2007; 59:185-205. [PMID: 17540906 DOI: 10.1124/pr.59.2.3] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Angiogenesis, the process through which new blood vessels arise from preexisting ones, is regulated by several "classic" factors, among which the most studied are vascular endothelial growth factor (VEGF) and fibroblast growth factor-2 (FGF-2). In recent years, investigations showed that, in addition to the classic factors, numerous endogenous peptides play a relevant regulatory role in angiogenesis. Such regulatory peptides, each of which exerts well-known specific biological activities, are present, along with their receptors, in the blood vessels and may take part in the control of the "angiogenic switch." An in vivo and in vitro proangiogenic effect has been demonstrated for erythropoietin, angiotensin II (ANG-II), endothelins (ETs), adrenomedullin (AM), proadrenomedullin N-terminal 20 peptide (PAMP), urotensin-II, leptin, adiponectin, resistin, neuropeptide-Y, vasoactive intestinal peptide (VIP), pituitary adenylate cyclase-activating polypeptide (PACAP), and substance P. There is evidence that the angiogenic action of some of these peptides is at least partly mediated by their stimulating effect on VEGF (ANG-II, ETs, PAMP, resistin, VIP and PACAP) and/or FGF-2 systems (PAMP and leptin). AM raises the expression of VEGF in endothelial cells, but VEGF blockade does not affect the proangiogenic action of AM. Other endogenous peptides have been reported to exert an in vivo and in vitro antiangiogenic action. These include somatostatin and natriuretic peptides, which suppress the VEGF system, and ghrelin, that antagonizes FGF-2 effects. Investigations on "nonclassic" regulators of angiogenesis could open new perspectives in the therapy of diseases coupled to dysregulation of angiogenesis.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Human Anatomy and Histology, School of Medicine, University of Bari, Bari, Italy.
| | | | | |
Collapse
|
45
|
Abstract
Octreotide has dramatically changed the results of medical treatment of acromegaly. It is the reference drug for the pharmacological treatment of acromegaly, owing to its impressive efficacy in suppressing growth hormome secretion, and excellent compliance. Safe growth hormone and normal insulin-like growth factor I values are reached in 50-60% of unselected patients. Octreotide arrests the growth of the tumor and shrinks tumor in over half of all patients (namely, up to 88% of naive patients and to complete disappearance in anecdotic cases). The safety profile of octreotide is excellent, but in some patients, glucose metabolism worsens and cholelythiasis occurs. This review will address the primary treatment and the relative roles of pharmacological and surgical treatment, as well as the predictivity of octreotide results.
Collapse
Affiliation(s)
- Renato Cozzi
- a Ospedale Niguarda Milano, Division of Endocrinology, via Canonica 81, 20154 Milano, Italy.
| | | |
Collapse
|
46
|
Sathyapalan T, Lowry M, Turnbull LW, Rowland-Hill C, Atkin SL. Mechanism of action of octreotide in acromegalic tumours in vivo using dynamic contrast-enhanced magnetic resonance imaging. Pituitary 2007; 10:233-6. [PMID: 17541750 DOI: 10.1007/s11102-007-0044-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
CONTEXT Octreotide causes significant tumour shrinkage in patients with acromegaly but the exact mechanism of action is unclear in vivo. OBJECTIVE To determine the mechanism of action of octreotide in vivo using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). DESIGN Five patients with acromegaly were treated with octreotide as primary medical therapy. DCE-MRI was done at baseline and 24 weeks. Local ethical committee approval was granted. SETTING Study was done in a tertiary care centre. PATIENTS Five patients with newly diagnosed acromegaly were recruited. INTERVENTION Patients were started on subcutaneous octreotide and DCE-MRI was done on 0 and 24 weeks. MAIN OUTCOME MEASURES Amplitude of contrast intake, exchange rate and maximum enhancement index of tumour tissue was compared before and after treatment. RESULTS Amplitude of contrast intake (9.87 +/- 3.52 vs. 4.97 +/- 1.96 P < or = 0.05) and exchange rate (6.27 +/- 1.57 vs. 1.63 +/- 0.76 P value < or = 0.01) were significantly higher at baseline in adenoma compared to normal pituitary tissue but was comparable to normal pituitary tissue after treatment. There was a significant decrease in amplitude of contrast intake and exchange rate which relates to functional vascularity of adenoma at 24 weeks compared to baseline (P-values 0.026 and 0.002 respectively) but there were no significant changes in the normal pituitary tissue. CONCLUSION DCE-MRI in acromegalic tumours treated with octreotide showed a significant reduction in functional vascularity after octreotide therapy compared to baseline in pituitary adenomas. This supports the antiangiogenic action of somatostatin analogue therapy in vitro, but it remains unclear if this mechanism is important clinically in analogue pre-treatment reducing the effect of radiotherapy on these pituitary tumours.
Collapse
|
47
|
Treiber G, Wex T, Röcken C, Fostitsch P, Malfertheiner P. Impact of biomarkers on disease survival and progression in patients treated with octreotide for advanced hepatocellular carcinoma. J Cancer Res Clin Oncol 2006; 132:699-708. [PMID: 16835748 DOI: 10.1007/s00432-006-0118-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Accepted: 05/04/2006] [Indexed: 01/05/2023]
Abstract
BACKGROUND Current determination of prognosis for advanced hepatocellular carcinoma (HCC) is mainly based on clinical assessment. We aimed to determine the impact of biomarkers as predictive factors for HCC progression and survival during octreotide-based treatments. PATIENTS AND METHODS We included patients who had been prospectively randomised to receive either octreotide (30 mg) alone monthly (n = 39) or in combination with rofecoxib (up to 50 mg bid daily, n = 32) for a minimum of 6 months, or until death occurred. RESULTS Overall median survival (154 days) and median time to progression (94 days) were not different for both treatments and the biomarkers investigated (VEGF-A, IGF-1, PGE-2, ET-A) were similarly distributed amongst treatment groups. Combined univariate group analysis revealed that survival was decreased for an uptake ratio of > 2 on initial octreoscan (P = 0.05); baseline serum VEGF-A and IGF-1 were further significantly associated with survival. On multivariate analysis, uncorrected serum VEGF-A appeared to be the most significant predictor for tumor progression and survival. CONCLUSIONS Biomarkers, in addition to established tumor markers, are independent predictors of tumor progression and survival in patients with advanced HCC treated with octreotide. Furthermore, the involvement of VEGF-A implies the inhibition of angiogenesis as a potential mechanism of action for this drug.
Collapse
Affiliation(s)
- G Treiber
- Department of Gastroenterology and Hepatology, University Hospital of Magdeburg, Magdeburg, Germany.
| | | | | | | | | |
Collapse
|
48
|
Niveiro M, Aranda FI, Peiró G, Alenda C, Picó A. Immunohistochemical analysis of tumor angiogenic factors in human pituitary adenomas. Hum Pathol 2005; 36:1090-5. [PMID: 16226108 DOI: 10.1016/j.humpath.2005.07.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Accepted: 07/29/2005] [Indexed: 11/15/2022]
Abstract
Microvessel density (MVD) has been studied in a number of neoplasias, and apparently, there is a relationship between angiogenesis and tumor progression, response to treatment, and outcome. In pituitary adenoma, the association between MVD and vascular endothelial growth factor (VEGF) with tumor behavior has been described, but correlation with other angiogenic factors such as fetal liver kinase 1 (Flk-1) or proliferative markers is unknown. We investigated MVD, VEGF, and its receptor Flk-1 expression in 60 human pituitary adenomas: 13 growth hormone cell adenomas, 7 prolactin cell adenomas, 5 corticotroph cell adenomas, 2 thyrotroph cell adenomas, and 33 nonfunctioning adenomas (30 gonadotroph cell adenomas and 3 null cell adenomas). We performed immunohistochemistry for CD34, Ki-67, VEGF, and Flk-1. To evaluate MVD, we used 2 methods: the number of vessels per square millimeter and the Chalkley method. Immunohistochemistry results were correlated, as well as with clinicopathologic factors. Adenomas with higher MVD were thyrotroph cell adenomas (299.9 +/- 87.5), and those with lower MVD were prolactin cell adenomas (168.6 +/- 63.3; P = .45, analysis of variance). We found a trend toward higher MVD in the adenomas of older patients (P = .142), but no difference was found regarding sex, extrasellar extension, or Ki-67 (P > .05). However, extrasellar extension was nearly significant when the Chalkley method score was high (P = .056). Low expression of VEGF was seen predominantly in prolactin cell adenomas, and high in nonfunctioning adenomas, or in cases of older patients (P < or = .032). Flk-1 score correlated with VEGF (P = .006). High expression was observed in nonfunctioning adenomas, cases presenting at older ages, and with extrasellar extension (P < or = .022). Our study shows that VEGF and Flk-1 are widely expressed in pituitary adenomas, predominantly in nonfunctioning adenomas and those presenting at older ages. Moreover, Flk-1 is associated with a more aggressive phenotype, and it may have potential therapeutic interest.
Collapse
Affiliation(s)
- María Niveiro
- Department of Pathology, General Universitary Hospital of Alicante, Pintor Baeza s/n, 03010-Alicante, Spain
| | | | | | | | | |
Collapse
|
49
|
Hernández C, Carrasco E, Casamitjana R, Deulofeu R, García-Arumí J, Simó R. Somatostatin molecular variants in the vitreous fluid: a comparative study between diabetic patients with proliferative diabetic retinopathy and nondiabetic control subjects. Diabetes Care 2005; 28:1941-7. [PMID: 16043736 DOI: 10.2337/diacare.28.8.1941] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE There is growing evidence to indicate that somatostatin could be added to the list of natural antiangiogenic factors that exist in the vitreous fluid. In addition, a deficit of intravitreous somatostatin-like immunoreactivity (SLI) has been found in diabetic patients with proliferative diabetic retinopathy (PDR). In the present study, we have determined the main molecular variants of somatostatin (somatostatin-14 and somatostatin-28) in the vitreous fluid and plasma of nondiabetic control subjects and diabetic patients with PDR. In addition, the contribution of cortistatin, a neuropeptide with strong structural similarities to somatostatin, to SLI and its levels in vitreous and plasma in both nondiabetic and diabetic patients has also been measured. RESERCH DESIGN AND METHODS: Plasma and vitreous fluid from 22 diabetic patients with PDR and 22 nondiabetic control subjects were analyzed. Somatostatin-14, somatostatin-28 and cortistatin were measured by radioimmunoassay but separation by high-performance liquid chromatography was required to measure somatostatin-14. RESULTS The predominant molecular form of somatostatin within the vitreous fluid was somatostatin-28 (fivefold higher than somatostatin-14 in control subjects and threefold higher in patients with PDR). Cortistatin significantly contributed to SLI and its intravitreous levels were higher than those detected in plasma (nondiabetic control subjects: 147 [102-837] vs. 78 [24-32] pg/ml; patients with PDR: 187 [87-998] vs. 62 [24-472] pg/ml; P = 0.01 for both). Intravitreous somatostatin-14 was similar in both subjects with PDR and the control group (P = 0.87). By contrast, somatostatin-28 concentration was lower in patients with PDR than in nondiabetic control subjects (350 +/- 32 vs. 595 +/- 66 pg/ml; P = 0.004). CONCLUSIONS Somatostatin-28 is the main molecular variant in the vitreous fluid. The intravitreous SLI deficit detected in patients with PDR is mainly due to somatostatin-28. Cortistatin is abundant in the vitreous fluid and significantly contributes to SLI. These findings could open up new strategies for PDR treatment.
Collapse
Affiliation(s)
- Cristina Hernández
- Diabetes Research Unit, Endocrinology Division, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
50
|
Conti E, Carrozza C, Capoluongo E, Volpe M, Crea F, Zuppi C, Andreotti F. Insulin-like growth factor-1 as a vascular protective factor. Circulation 2005; 110:2260-5. [PMID: 15477425 DOI: 10.1161/01.cir.0000144309.87183.fb] [Citation(s) in RCA: 178] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Elena Conti
- Department of Cardiovascular Diseases, Institute of Cardiology, Catholic University, Medical School, Via Todi 60, 00181 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|