1
|
Gunasekara W, Sachindra J, Madhushika MT, Liyanage P, Lekamwasam S. Cimetidine repurposed as a potential immunomodulatory agent against colorectal carcinoma: A systematic review. J Oncol Pharm Pract 2024; 30:930-936. [PMID: 38592456 DOI: 10.1177/10781552241247007] [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: 04/10/2024]
Abstract
OBJECTIVE To determine the survival benefit and immunomodulatory effects of cimetidine pre-, peri- or post-operatively in patients with colorectal cancer (CRC). METHODS A systematic review was conducted using PubMed and Cochrane Library to retrieve randomized control trials (RCTs) that investigated the effects of cimetidine on survival and immunomodulation via improvement in tumor infiltrating lymphocytes (TILs) and peripheral blood lymphocytes. The review was carried out in accordance with the extended Preferred Reporting Items for Systematic Reviews and Meta-analyses. RESULTS Four studies with the total of 267 patients were included in this systematic review. Treatment duration varied from 5 days to 1 year. Two studies reported a significant TIL response in the resected specimens after administering cimetidine, while one RCT showed an escalation of CD3, CD4 and CD57 lymphocytes in peripheral blood compared to the baseline following cimetidine treatment (p < 0.01). Of the three trials that examined the effects of cimetidine on survival, only two studies revealed significant survival benefit while the remaining study only showed a trend towards survival benefit. CONCLUSION Repurposing of existing drugs like cimetidine has a potential to offer a survival benefit by acting as an immunomodulatory agent in patients undergoing curative resection for CRC. However, the heterogeneity seen in current studies and the evolvement of adjunctive therapies for CRC warrant large-scale, well-designed prospective RCTs to establish the efficacy of cimetidine in CRC.
Collapse
Affiliation(s)
- Wwm Gunasekara
- Department of Pharmacology, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - Jlaa Sachindra
- Department of Pharmacology, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - M T Madhushika
- Department of Pharmacology, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - Plgc Liyanage
- Department of Pharmacology, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - S Lekamwasam
- Department of Medicine, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| |
Collapse
|
2
|
Adelakun SA, Ogunlade B, Akintunde OW, Omilachi VO. Long-term exposure to cimetidine induced gonado-toxicity in male rats: Modulating role of Ocimum gratissimum. Rev Int Androl 2022; 20 Suppl 1:S2-S16. [PMID: 35101366 DOI: 10.1016/j.androl.2020.10.012] [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: 02/29/2020] [Revised: 08/01/2020] [Accepted: 10/17/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Available evidence suggests that cimetidine is a reproductive toxicant that induces sexual and testicular dysfunction. Ocimum gratissimum (OG) is globally consumed for medicinal and nutritional purposes. To determine the modulating role of aqueous leaf extract of Ocimum gratissimum on cimetidine-induced gonado-toxicity, sexually mature male rats were randomized into four groups of six (n=6) rats each. Group A: control given 2ml distilled water. Group B received 500mg/kg body weight (bwt) of OG extract, Group C received 50mg/kg bwt cimetidine, and group D received 50mg/kg bwt of cimetidine+500mg/kg bwt OG extract once daily for 8 weeks via gastric gavage. Parameters tested include sperm parameters, testosterone (TT), luteinizing hormone (LH), follicle stimulating hormone (FSH) and prolactin, testicular alkaline phosphatase (ALP), acid phosphatase (ACP), lactate dehydrogenase (LDH), protein, cholesterol, glycogen, sexual behavioural parameters, and testicular histology. RESULTS There were depletions in the seminiferous epithelium, decreased sperm quality, TT, LH, and FSH, testicular enzymes, protein, cholesterol, glycogen, and sexual behaviour increase in animals treated with cimetidine only compared to control. OG restored and improved sexual behaviour and libido as evident from increased frequencies of mount, intromission, ejaculation, and ejaculatory latency. Mount latencies, intromission, post-ejaculation, and prolactin were significantly decreased. The significantly decreased testicular activities of ALP, ACP, LDH and protein, cholesterol, glycogen concentrations, TT, LH and FSH were increased by OG administration. CONCLUSION Ocimum gratissimum attenuated the deleterious effects of cimetidine on the testis, protected the seminiferous epithelium, restored, and boosted sexual competence, and promoted spermatogenesis.
Collapse
Affiliation(s)
- Sunday Aderemi Adelakun
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria; Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| | - Babatunde Ogunlade
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Olalekan Wasiu Akintunde
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Victoria Ojima Omilachi
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
3
|
Duarte D, Vale N. Combining repurposed drugs to treat colorectal cancer. Drug Discov Today 2021; 27:165-184. [PMID: 34592446 DOI: 10.1016/j.drudis.2021.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 07/19/2021] [Accepted: 09/22/2021] [Indexed: 02/08/2023]
Abstract
The drug development process, especially of antineoplastic agents, has become increasingly costly and ineffective. Drug repurposing and drug combination are alternatives to de novo drug development, being low cost, rapid, and easy to apply. These strategies allow higher efficacy, decreased toxicity, and overcoming of drug resistance. The combination of antineoplastic agents is already being applied in cancer therapy, but the combination of repurposed drugs is still under-explored in pre- and clinical development. In this review, we provide a set of pharmacological concepts focusing on drug repurposing for treating colorectal cancer (CRC) and that are relevant for the application of new drug combinations against this disease.
Collapse
Affiliation(s)
- Diana Duarte
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| |
Collapse
|
4
|
Ginosar Y, Bromberg Z, Nachmanson N, Ariel I, Skarzinski G, Hagai L, Elchalal U, Shapiro J, Abramovitch R. Chronic hypoxia in pregnant mice impairs the placental and fetal vascular response to acute hypercapnia in BOLD-MRI hemodynamic response imaging. Placenta 2021; 110:29-38. [PMID: 34116499 DOI: 10.1016/j.placenta.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 04/29/2021] [Accepted: 05/25/2021] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Brief hypercapnic challenge causes acute placental hypoperfusion with fetal brain sparing on BOLD-MRI. We hypothesize that this non-invasive imaging strategy can distinguish between normal pregnancy and chronic placental hypoperfusion (using the maternal hypoxia model). METHODS Eighteen pregnant female ICR mice were randomized to three groups: normoxia, late-onset hypoxia (12%O2;E13.5-17.5) and early-onset hypoxia (12%O2;E10.5-17.5). On E17.5, animals were imaged in a 4.7-T Bruker-Biospec MRI scanner. Fast coronal True-FISP was performed to identify organs of interest (placenta and fetal heart, liver and brain). BOLD-MRI was performed at baseline and during a 4-min hypercapnic challenge (5%CO2). %-change in placental and fetal signal was analyzed from T2*-weighted gradient echo MR images. Following MRI, fetuses and placentas were harvested, weighed and immuno-stained. RESULTS In normoxic mice, hypercapnia caused reduction in BOLD-MRI signal in placenta (-44% ± 7%; p < 0.0001), fetal liver (-32% ± 7%; p < 0.0001) and fetal heart (-54% ± 12%; p < 0.002), with relative fetal brain sparing (-12% ± 5%; p < 0.0001). These changes were markedly attenuated in both hypoxia groups. Baseline fetal brain/placenta SI ratio was highest in normoxic mice (1.14 ± 0.017) and reduced with increasing duration of hypoxia (late-onset hypoxia: 1.00 ± 0.026; early-onset hypoxia: 0.91 ± 0.016; p = 0.02). Both hypoxic groups exhibited fetal growth restriction with prominent placental glycogen-containing cells, particularly in early-onset hypoxia. There was increased fetal neuro- and intestinal-apoptosis in early-onset hypoxia only. CONCLUSIONS BOLD-MRI with brief hypercapnic challenge distinguished between normoxia and both hypoxia groups, while fetal neuroapoptosis was only observed after early-onset hypoxia. This suggests that BOLD-MRI with hypercapnic challenge can identify chronic fetal asphyxia before the onset of irreversible brain injury.
Collapse
Affiliation(s)
- Yehuda Ginosar
- Department of Anesthesiology and Critical Care Medicine, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel; Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA; The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel.
| | - Zohar Bromberg
- The Goldyne Savad Institute of Gene Therapy and MRI Laboratory, Human Biology Research Center, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Nathalie Nachmanson
- The Goldyne Savad Institute of Gene Therapy and MRI Laboratory, Human Biology Research Center, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Ilana Ariel
- Perinatal Pathology Unit, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Galina Skarzinski
- Perinatal Pathology Unit, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Lital Hagai
- Medical Student, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Uriel Elchalal
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Joel Shapiro
- Department of Anesthesiology and Critical Care Medicine, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Rinat Abramovitch
- The Goldyne Savad Institute of Gene Therapy and MRI Laboratory, Human Biology Research Center, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Ein Karem, And the Faculty of Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
5
|
Galisteo A, Jannus F, García-García A, Aheget H, Rojas S, Lupiañez JA, Rodríguez-Diéguez A, Reyes-Zurita FJ, Quílez del Moral JF. Diclofenac N-Derivatives as Therapeutic Agents with Anti-Inflammatory and Anti-Cancer Effect. Int J Mol Sci 2021; 22:ijms22105067. [PMID: 34064702 PMCID: PMC8151993 DOI: 10.3390/ijms22105067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/20/2022] Open
Abstract
A series of diclofenac N-derivatives (2, 4, 6, 8c, 9c, 10a-c) were synthesized in order to test their anti-cancer and anti-inflammatory effects. The anticarcinogen activity has been assayed against three cancer cell lines: HT29, human colon cancer cells; Hep-G2, human hepatic cells; and B16-F10, murine melanoma cells. First, we determined the cytotoxicity of the different compounds, finding that the most effective compound was compound 8c against all cell lines and both compounds 4 and 6 in human Hep-G2 and HT29 cell lines. Compounds 4 and 8c were selected for the percentage of apoptosis determination, cell cycle distribution, and mitochondrial membrane potential measure because these products presented the lowest IC50 values in two of the three cancer cell lines assayed (B16-F10 and HepG2), and were two of the three products with lowest IC50 in HT29 cell line. Moreover, the percentages of apoptosis induction were determined for compounds 4 and 8c, showing that the highest values were between 30 to 60%. Next, the effects of these two compounds were observed on the cellular cycle, resulting in an increase in the cell population in G2/M cell cycle phase after treatment with product 8c, whereas compound 4 increased the cells in phase G0/G1, by possible differentiation process induction. Finally, to determine the possible apoptosis mechanism triggered by these compounds, mitochondrial potential was evaluated, indicating the possible activation of extrinsic apoptotic mechanism. On the other hand, we studied the anti-inflammatory effects of these diclofenac (DCF) derivatives on lipopolysaccharide (LPS) activated RAW 264.7 macrophages-monocytes murine cells by inhibition of nitric oxide (NO) production. As a first step, we determined the cytotoxicity of the synthesized compounds, as well as DCF, against these cells. Then, sub-cytotoxic concentrations were used to determine NO release at different incubation times. The greatest anti-inflammatory effect was observed for products 2, 4, 8c, 10a, 10b, and 9c at 20 µg·mL-1 concentration after 48 h of treatment, with inhibition of produced NO between 60 to 75%, and a concentration that reduces to the 50% the production of NO (IC50 NO) between 2.5 to 25 times lower than that of DCF. In this work, we synthesized and determined for the first time the anti-cancer and anti-inflammatory potential of eight diclofenac N-derivatives. In agreement with the recent evidences suggesting that inflammation may contribute to all states of tumorigenesis, the development of these new derivatives capable of inducing apoptosis and anti-inflammatory effects at very low concentrations represent new effective therapeutic strategies against these diseases.
Collapse
Affiliation(s)
- Alberto Galisteo
- Department of Organic Chemistry, Institute of Biotechnology, University of Granada, 18071 Granada, Spain;
| | - Fatin Jannus
- Department of Biochemistry and Molecular Biology, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (F.J.); (J.A.L.)
| | - Amalia García-García
- Department of Inorganic Chemistry, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (A.G.-G.); (S.R.); (A.R.-D.)
| | - Houssam Aheget
- Centre for Genomics and Oncological Research, GENYO, C/Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain;
| | - Sara Rojas
- Department of Inorganic Chemistry, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (A.G.-G.); (S.R.); (A.R.-D.)
| | - José A. Lupiañez
- Department of Biochemistry and Molecular Biology, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (F.J.); (J.A.L.)
| | - Antonio Rodríguez-Diéguez
- Department of Inorganic Chemistry, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (A.G.-G.); (S.R.); (A.R.-D.)
| | - Fernando J. Reyes-Zurita
- Department of Biochemistry and Molecular Biology, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (F.J.); (J.A.L.)
- Correspondence: (F.J.R.-Z.); (J.F.Q.d.M.); Tel.: +34-958243252 (F.J.R.-Z.); +34-958243185 (J.F.Q.d.M.)
| | - José F. Quílez del Moral
- Department of Organic Chemistry, Institute of Biotechnology, University of Granada, 18071 Granada, Spain;
- Correspondence: (F.J.R.-Z.); (J.F.Q.d.M.); Tel.: +34-958243252 (F.J.R.-Z.); +34-958243185 (J.F.Q.d.M.)
| |
Collapse
|
6
|
Messer JG, Castillo EJ, Abraham AM, Jiron JM, Israel R, Yarrow JF, Thomas S, Reynolds MC, Wnek RD, Jorgensen M, Wanionok N, Van Poznak C, Bhattacharyya I, Kimmel DB, Aguirre JI. Anti-vascular endothelial growth factor antibody monotherapy causes destructive advanced periodontitis in rice rats (Oryzomys palustris). Bone 2020; 130:115141. [PMID: 31707108 PMCID: PMC6941430 DOI: 10.1016/j.bone.2019.115141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/15/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Angiogenesis inhibitors (AgI) are commonly used in combination chemotherapy protocols to treat cancer, and have been linked to osteonecrosis of the jaw (ONJ). However, it is unknown if AgI therapy alone is sufficient to induce ONJ. We have previously established an ONJ model in rice rats with localized periodontitis that receive zoledronic acid (ZOL). The purpose of this study was to use this model to determine the role of anti-vascular endothelial growth factor A (anti-VEGF) antibody treatment of rice rats with localized maxillary periodontitis. We hypothesized that rice rats with localized maxillary periodontitis given anti-VEGF monotherapy will develop oral lesions that resemble ONJ, defined by exposed, necrotic alveolar bone. METHODS At age 4 weeks, 45 male rice rats were randomized into three groups (n = 15): 1) VEH (saline), 2) ZOL (80 μg/kg body weight, intravenously once monthly), and 3) anti-VEGF (5 mg B20-4.1.1/kg body weight, subcutaneously twice weekly). After 24 weeks, rats were euthanized, jaws were excised and a high-resolution photograph of each quadrant was taken to assign a severity grade based on gross appearance. Jaws were then fixed, scanned by MicroCT, decalcified and sectioned for histopathologic and immunohistochemical analyses. RESULTS 40-80% of the rats in the three groups developed gross oral lesions. 50% of ZOL rats developed ONJ. In contrast, 80% of the anti-VEGF rats developed destructive advanced periodontitis that was characterized by extreme alveolar bone loss and fibrosis. Anti-VEGF rats never developed exposed, necrotic bone. Furthermore, only anti-VEGF rats developed mild to severe mandibular periodontitis. Compared to VEH rats, more T-cells were found in periodontal lesions of anti-VEGF rats and more cells of the monocyte lineage were found in ONJ lesions of ZOL rats. CONCLUSIONS Anti-VEGF monotherapy administered to a validated rodent model of ONJ caused a destructive advanced form of periodontitis that differed significantly from ONJ.
Collapse
Affiliation(s)
- J G Messer
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - E J Castillo
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - A M Abraham
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - J M Jiron
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - R Israel
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - J F Yarrow
- Research Service, VA Medical Center, Gainesville, FL, United States of America; Division of Endocrinology, Diabetes, and Metabolism, University of Florida College of Medicine, Gainesville, FL, United States of America.
| | - S Thomas
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - M C Reynolds
- Research Service, VA Medical Center, Gainesville, FL, United States of America
| | - R D Wnek
- Research Service, VA Medical Center, Gainesville, FL, United States of America
| | - M Jorgensen
- Department of Pediatrics, College of Medicine, UF, United States of America.
| | - N Wanionok
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America
| | - C Van Poznak
- University of Michigan, Ann Arbor, MI, United States of America.
| | - I Bhattacharyya
- Department of Oral & Maxillofacial Diagnostic Sciences, College of Dentistry, UF, United States of America.
| | - D B Kimmel
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - J I Aguirre
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| |
Collapse
|
7
|
Nowak-Sliwinska P, Scapozza L, Ruiz i Altaba A. Drug repurposing in oncology: Compounds, pathways, phenotypes and computational approaches for colorectal cancer. Biochim Biophys Acta Rev Cancer 2019; 1871:434-454. [PMID: 31034926 PMCID: PMC6528778 DOI: 10.1016/j.bbcan.2019.04.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 02/08/2023]
Abstract
The strategy of using existing drugs originally developed for one disease to treat other indications has found success across medical fields. Such drug repurposing promises faster access of drugs to patients while reducing costs in the long and difficult process of drug development. However, the number of existing drugs and diseases, together with the heterogeneity of patients and diseases, notably including cancers, can make repurposing time consuming and inefficient. The key question we address is how to efficiently repurpose an existing drug to treat a given indication. As drug efficacy remains the main bottleneck for overall success, we discuss the need for machine-learning computational methods in combination with specific phenotypic studies along with mechanistic studies, chemical genetics and omics assays to successfully predict disease-drug pairs. Such a pipeline could be particularly important to cancer patients who face heterogeneous, recurrent and metastatic disease and need fast and personalized treatments. Here we focus on drug repurposing for colorectal cancer and describe selected therapeutics already repositioned for its prevention and/or treatment as well as potential candidates. We consider this review as a selective compilation of approaches and methodologies, and argue how, taken together, they could bring drug repurposing to the next level.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland; Translational Research Center in Oncohaematology, University of Geneva, Rue Michel Servet 1, 1211 Geneva 4, Switzerland.
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland
| | - Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, 1211 Geneva 4, Switzerland
| |
Collapse
|
8
|
Beltrame FL, Sasso-Cerri E. Vitamin B 12-induced spermatogenesis recovery in cimetidine-treated rats: effect on the spermatogonia number and sperm concentration. Asian J Androl 2018; 19:567-572. [PMID: 27568998 PMCID: PMC5566851 DOI: 10.4103/1008-682x.182397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The H2-receptor antagonist cimetidine is an antiulcer drug also used for the treatment of cancer due to its antiangiogenic effect. However, this drug has caused structural changes in the seminiferous tubules. Vitamin B12 has been used as a therapeutic agent for the treatment of male infertility. The supplementation of rats with vitamin B12 during cimetidine treatment has recovered the damaged seminiferous tubules, but how this vitamin restores the seminiferous epithelium has not been clarified. In this study, we evaluated whether vitamin B12 improves the number of spermatogonia, spermatocytes, and sperm concentration in cimetidine-treated rats. Adult male rats were treated for 50 days as follows: cimetidine group received 100 mg kg−1 b.w. of cimetidine, cimetidine-B12 group received cimetidine and 3 μg of vitamin B12-hydroxocobalamin, B12 group received only 3 μg of vitamin, and control group received saline. Sperm concentration was calculated and historesin-embedded testes sections were used for the quantitative analyses of spermatogonia (A; In/B) and spermatocytes. TUNEL method and PCNA immunofluorescence were performed. Cimetidine caused a significant reduction in sperm concentration. TUNEL-positive spermatogonia and spermatocytes were correlated to a significant reduction in the number of these cells. In cimetidine-B12 group, sperm concentration was higher than cimetidine group and a significant increase in the number of spermatogonia (stages II–VI) was correlated to a high incidence of PCNA-immunolabeled spermatogonia and spermatocytes. The results show that the supplementation of rats with vitamin B12 during cimetidine treatment increases sperm concentration and exerts a potential effect in the recovery of spermatogonia and spermatocytes.
Collapse
Affiliation(s)
- Flávia L Beltrame
- Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Estela Sasso-Cerri
- Department of Morphology, Laboratory of Histology and Embryology, Dental School - São Paulo State University (UNESP/FOAr), Araraquara, São Paulo, Brazil
| |
Collapse
|
9
|
Sun YZ, Liu L, Cai N, Liu NN. Anti-angiogenic effect of rapamycin in mouse oxygen-induced retinopathy is mediated through suppression of HIF-1alpha/VEGF pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10167-10175. [PMID: 31966350 PMCID: PMC6965774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/16/2017] [Indexed: 06/10/2023]
Abstract
Hypoxia-inducible factor-1alpha (HIF-1α)-vascular endothelial growth factor (VEGF) pathway, a downstream of mammalian target of rapamycin (mTOR), plays a major role in the formation of pathological retinal angiogenesis. Rapamycin (RAPA), a highly specific inhibitor of mTOR, is widely used in cancer studies for its antiangiogenic activity. However, the inhibitory effects of RAPA on the HIF-1α-VEGF pathway in retinal tissues were rarely researched. The study aimed to investigate the efficacy and potential mechanisms of RAPA in inhibiting retinal neovascularization. Human umbilical vein endothelial cells (HUVECs) were treated with hypoxia and in the presence of different concentrations of RAPA. RAPA was injected intraperitoneally in oxygen-induced retinopathy (OIR) C57BL/6 mice from postnatal day 12 (P12) to P17. The proliferation of HUVECs, the protein and mRNA expressions of HIF-1α and VEGF were evaluated in HUVECs or OIR mice using MTT assay, ELISA, immunohistochemistry, Western-blot and real-time PCR. Histological methods were used to count blood vessel profiles in the inner retina. RAPA inhibited HUVECs proliferation and retinal neovascularization by reducing protein and mRNA expressions of HIF-1α and VEGF. RAPA suppresses hypoxia-induced HUVECs cell proliferation and pathological ocular angiogenesis through a mechanism linked to the targeting of HIF-1α/VEGF signaling.
Collapse
Affiliation(s)
- Yi Zhou Sun
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University Shenyang, Liaoning Province, China
| | - Lei Liu
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University Shenyang, Liaoning Province, China
| | - Na Cai
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University Shenyang, Liaoning Province, China
| | - Ning Ning Liu
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University Shenyang, Liaoning Province, China
| |
Collapse
|
10
|
Zhang Y, Davis C, Shah S, Hughes D, Ryan JC, Altomare D, Peña MMO. IL-33 promotes growth and liver metastasis of colorectal cancer in mice by remodeling the tumor microenvironment and inducing angiogenesis. Mol Carcinog 2016; 56:272-287. [PMID: 27120577 DOI: 10.1002/mc.22491] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/23/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023]
Abstract
Liver metastasis is the major cause of death from colorectal cancer (CRC). Understanding its mechanisms is necessary for timely diagnosis and development of effective therapies. Interleukin-33 (IL-33) is an IL-1 cytokine family member that uniquely functions as a cytokine and nuclear factor. It is released by necrotic epithelial cells and activated innate immune cells, functioning as an alarmin or an early danger signal. Its role in invoking type 2 immune response has been established; however, it has contrasting roles in tumor development and metastasis. We identified IL-33 as a potently upregulated cytokine in a highly metastatic murine CRC cell line and examined its role in tumor growth and metastasis to the liver. IL-33 was transgenically expressed in murine and human adenocarcinoma and carcinoma cell lines and their growth and spontaneous metastasis to the liver were assessed in orthotopic models of CRC in wild-type C57Bl/6 and Il33 knockout mice. The results showed that increased expression of IL-33 in CRC cells enhanced their tumor take, growth, and liver metastasis. Tumor- rather than host-derived IL-33 induced the enhanced recruitment of CD11b+ GR1+ and CD11b+ F4/80+ myeloid cells to remodel the tumor microenvironment by increased expression of mobilizing cytokines, and tumor angiogenesis by activating endothelial cells. IL-33 expression was elevated in patient tumor tissues, induced early in adenoma development, and activated by pro-inflammatory cytokines derived from the tumor microenvironment. The data suggest that tumor-derived IL-33 modulates the tumor microenvironment to potently promote colon carcinogenesis and liver metastasis, underscoring its potential as a therapeutic target. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| | - Celestia Davis
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Sapana Shah
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| | - Daniel Hughes
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| | | | - Diego Altomare
- College of Pharmacy, Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, South Carolina
| | - Maria Marjorette O Peña
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
11
|
Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-diclofenac as an anti-cancer agent. Ecancermedicalscience 2016; 10:610. [PMID: 26823679 PMCID: PMC4720497 DOI: 10.3332/ecancer.2016.610] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Diclofenac (DCF) is a well-known and widely used non-steroidal anti-inflammatory drug (NSAID), with a range of actions which are of interest in an oncological context. While there has long been an interest in the use of NSAIDs in chemoprevention, there is now emerging evidence that such drugs may have activity in a treatment setting. DCF, which is a potent inhibitor of COX-2 and prostaglandin E2 synthesis, displays a range of effects on the immune system, the angiogenic cascade, chemo- and radio-sensitivity and tumour metabolism. Both pre-clinical and clinical evidence of these effects, in multiple cancer types, is assessed and summarised and relevant mechanisms of action outlined. Based on this evidence the case is made for further clinical investigation of the anticancer effects of DCF, particularly in combination with other agents - with a range of possible multi-drug and multi-modality combinations outlined in the supplementary materials accompanying the main paper.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | | | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc; Newton MA 02459, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
12
|
Rajendran R, Liang J, Tang MYA, Henry B, Chuang KH. Optimization of arterial spin labeling MRI for quantitative tumor perfusion in a mouse xenograft model. NMR IN BIOMEDICINE 2015; 28:988-997. [PMID: 26104980 DOI: 10.1002/nbm.3330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 03/18/2015] [Accepted: 04/22/2015] [Indexed: 06/04/2023]
Abstract
Perfusion is an important biomarker of tissue function and has been associated with tumor pathophysiology such as angiogenesis and hypoxia. Arterial spin labeling (ASL) MRI allows noninvasive and quantitative imaging of perfusion; however, the application in mouse xenograft tumor models has been challenging due to the low sensitivity and high perfusion heterogeneity. In this study, flow-sensitive alternating inversion recovery (FAIR) ASL was optimized for a mouse xenograft tumor. To assess the sensitivity and reliability for measuring low perfusion, the lumbar muscle was used as a reference region. By optimizing the number of averages and inversion times, muscle perfusion as low as 32.4 ± 4.8 (mean ± standard deviation) ml/100 g/min could be measured in 20 min at 7 T with a quantification error of 14.4 ± 9.1%. Applying the optimized protocol, heterogeneous perfusion ranging from 49.5 to 211.2 ml/100 g/min in a renal carcinoma was observed. To understand the relationship with tumor pathology, global and regional tumor perfusion was compared with histological staining of blood vessels (CD34), hypoxia (CAIX) and apoptosis (TUNEL). No correlation was observed when the global tumor perfusion was compared with these pathological parameters. Regional analysis shows that areas of high perfusion had low microvessel density, which was due to larger vessel area compared with areas of low perfusion. Nonetheless, these were not correlated with hypoxia or apoptosis. The results suggest that tumor perfusion may reflect certain aspect of angiogenesis, but its relationship with other pathologies needs further investigation.
Collapse
Affiliation(s)
- Reshmi Rajendran
- Magnetic Resonance Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
| | - Jieming Liang
- Magnetic Resonance Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
| | - Mei Yee Annie Tang
- Magnetic Resonance Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
| | - Brian Henry
- Translational Medicine Research Centre, MSD, Singapore
| | - Kai-Hsiang Chuang
- Magnetic Resonance Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
- Clinical Imaging Research Centre, National University of Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
13
|
Laufer S, Mazuz A, Nachmansson N, Fellig Y, Corn BW, Bokstein F, Bashat DB, Abramovitch R. Monitoring brain tumor vascular heamodynamic following anti-angiogenic therapy with advanced magnetic resonance imaging in mice. PLoS One 2014; 9:e115093. [PMID: 25506833 PMCID: PMC4266643 DOI: 10.1371/journal.pone.0115093] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/18/2014] [Indexed: 11/18/2022] Open
Abstract
Advanced MR imaging methods have an essential role in classification, grading, follow-up and therapeutic management in patients with brain tumors. With the introduction of new therapeutic options, the challenge for better tissue characterization and diagnosis increase, calling for new reliable non-invasive imaging methods. In the current study we evaluated the added value of a combined protocol of blood oxygen level dependent (BOLD) imaging during hyperoxic challenge (termed hemodynamic response imaging (HRI)) in an orthotopic mouse model for glioblastoma under anti-angiogenic treatment with B20-4.1.1, an anti-VEGF antibody. In glioblastoma tumors, the elevated HRI indicated progressive angiogenesis as further confirmed by histology. In the current glioblastoma model, B20-treatment caused delayed tumor progression with no significant changes in HRI yet with slightly reduced tumor vascularity as indicated by histology. Furthermore, fewer apoptotic cells and higher proliferation index were detected in the B20-treated tumors compared to control-treated tumors. In conclusion, HRI provides an easy, safe and contrast agent free method for the assessment of the brain hemodynamic function, an additionally important clinical information.
Collapse
Affiliation(s)
- Shlomi Laufer
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ahinoam Mazuz
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Nathalie Nachmansson
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yakov Fellig
- Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | - Felix Bokstein
- Neuro-Oncology Service. Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Dafna Ben Bashat
- The Functional Brain Center, The Wohl Institute for Advanced Imaging, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Rinat Abramovitch
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
14
|
Komar-Stossel C, Gross E, Dery E, Corchia N, Meir K, Fried I, Abramovitch R. TL-118 and gemcitabine drug combination display therapeutic efficacy in a MYCN amplified orthotopic neuroblastoma murine model--evaluation by MRI. PLoS One 2014; 9:e90224. [PMID: 24603724 PMCID: PMC3946152 DOI: 10.1371/journal.pone.0090224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 01/31/2014] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB) is the most common extra-cranial pediatric solid tumor with up to 50% of NB patients classified as having high-risk disease with poor long-term survival rates. The poor clinical outcome and aggressiveness of high-risk NB strongly correlates with enhanced angiogenesis, suggesting anti-angiogenic agents as attractive additions to the currently insufficient therapeutics. TL-118, a novel drug combination has been recently developed to inhibit tumor angiogenesis. In the current study, we used the SK-N-BE (2) cell line to generate orthotopic NB tumors in order to study the combinational therapeutic potential of TL-118 with either Gemcitabine (40 mg/kg; IP) or Retinoic acid (40 mg/kg; IP). We show that TL-118 treatment (n = 9) significantly inhibited tumor growth, increased cell apoptosis, reduced proliferation and extended mouse survival. Moreover, the reciprocal effect of TL-118 and Gemcitabine treatment (n = 10) demonstrated improved anti-tumor activity. The synergistic effect of these drugs in combination was more effective than either TL or Gemcitabine alone (n = 9), via significantly reduced cell proliferation (p<0.005), increased apoptosis (p<0.05) and significantly prolonged survival (2-fold; p<0.00001). To conclude, we demonstrate that the novel drug combination TL-118 has the ability to suppress the growth of an aggressive NB tumor. The promising results with TL-118 in this aggressive animal model may imply that this drug combination has therapeutic potential in the clinical setting.
Collapse
Affiliation(s)
- Chani Komar-Stossel
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel; MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Eitan Gross
- Pediatric Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Elia Dery
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel; MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Nathalie Corchia
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel; MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Karen Meir
- Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Iris Fried
- Pediatric Hemato-Oncology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Rinat Abramovitch
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel; MRI/MRS lab HBRC, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
15
|
Kekelidze M, D’Errico L, Pansini M, Tyndall A, Hohmann J. Colorectal cancer: Current imaging methods and future perspectives for the diagnosis, staging and therapeutic response evaluation. World J Gastroenterol 2013; 19:8502-8514. [PMID: 24379567 PMCID: PMC3870495 DOI: 10.3748/wjg.v19.i46.8502] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/06/2013] [Accepted: 11/19/2013] [Indexed: 02/06/2023] Open
Abstract
In the last 10 years the mortality rate of colorectal cancer (CRC) has decreased by more than 20% due to the rising developments in diagnostic techniques and optimization of surgical, neoadjuvant and palliative therapies. Diagnostic methods currently used in the evaluation of CRC are heterogeneous and can vary within the countries and the institutions. This article aims to discuss in depth currently applied imaging modalities such as virtual computed tomography colonoscopy, endorectal ultrasound, computed tomography (CT) and magnetic resonance imaging (MRI) in the diagnosis of CRC. Special focus is put on the potential of recent diagnostic developments as diffusion weighted imaging MRI, MRI biomarkers (dynamic enhanced MRI), positron emission tomography with 2-(fluorine-18)-fluoro-2-deoxy-D-glucose (FDG-PET) combined with computed tomography (PET/CT) and new hepatobiliary MRI contrast agents. The precise role, advantage and disadvantages of these modalities are evaluated controversially in local staging, metastatic spread and treatment monitoring of CRC. Finally, the authors will touch upon the future perspectives in functional imaging evaluating the role of integrated FDG-PET/CT with perfusion CT, MRI spectroscopy of primary CRC and hepatic transit time analysis using contrast enhanced ultrasound and MRI in the detection of liver metastases. Validation of these newer imaging techniques may lead to significant improvements in the management of patients with colorectal cancer.
Collapse
|
16
|
Edrei Y, Freiman M, Sklair-Levy M, Tsarfaty G, Gross E, Joskowicz L, Abramovitch R. Quantitative functional MRI biomarkers improved early detection of colorectal liver metastases. J Magn Reson Imaging 2013; 39:1246-53. [PMID: 24006217 DOI: 10.1002/jmri.24270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 05/16/2013] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To implement and evaluate the performance of a computerized statistical tool designed for robust and quantitative analysis of hemodynamic response imaging (HRI) -derived maps for the early identification of colorectal liver metastases (CRLM). MATERIALS AND METHODS CRLM-bearing mice were scanned during the early stage of tumor growth and subsequently during the advanced-stage. Three experienced radiologists marked various suspected-foci on the early stage anatomical images and classified each as either highly certain or as suspected tumors. The statistical model construction was based on HRI maps (functional-MRI combined with hypercapnia and hyperoxia) using a supervised learning paradigm which was further trained either with the advanced-stage sets (late training; LT) or with the early stage sets (early training; ET). For each group of foci, the classifier results were compared with the ground-truth. RESULTS The ET-based classification significantly improved the manual classification of the highly certain foci (P < 0.05) and was superior compared with the LT-based classification (P < 0.05). Additionally, the ET-based classification, offered high sensitivity (57-63%), accompanied with high positive predictive value (>94%) and high specificity (>98%) for suspected-foci. CONCLUSION The ET-based classifier can strengthen the radiologist's classification of highly certain foci. Additionally, it can aid in classifying suspected-foci, thus enabling earlier intervention which can often be lifesaving.
Collapse
Affiliation(s)
- Yifat Edrei
- The Goldyne Savad Institute for Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
17
|
Serum proteomics in biomedical research: a systematic review. Appl Biochem Biotechnol 2013; 170:774-86. [PMID: 23609910 DOI: 10.1007/s12010-013-0238-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 04/11/2013] [Indexed: 12/22/2022]
Abstract
Proteins that are important indicators of physiological or pathological states may contribute to the early diagnosis of disease, which may provide a basis for identifying the underlying mechanism of disease development. Serum, contains an abundance of proteins, offers an easy and inexpensive approach for disease detection and possesses a high potential to revolutionize the diagnostics. These differentially expressed proteins in serum have become an important role to monitoring the state for disease. Availability of emerging proteomic techniques gives optimism that serum can eventually be placed as a biomedium for clinical diagnostics. Advancements have benefited biomarker research to the point where serum is now recognized as an excellent diagnostic medium for the detection of disease. Comprehensive proteome of human serum fluid with high accuracy and availability has the potential to open new doors for disease biomarker discovery and for disease diagnostics, providing insights useful for future study. Thus, this review presents an overview of the value of serum as a credible diagnostic tool, and we aim to summarize the proteomic technologies currently used for global analysis of serum proteins and to elaborate on the application of serum proteomics to the discovery of disease biomarkers, and discuss some of the critical challenges and perspectives for this emerging field.
Collapse
|
18
|
Breuer S, Maimon O, Appelbaum L, Peretz T, Hubert A. TL-118-anti-angiogenic treatment in pancreatic cancer: a case report. Med Oncol 2013; 30:585. [PMID: 23609193 DOI: 10.1007/s12032-013-0585-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/13/2013] [Indexed: 01/05/2023]
Abstract
Pancreatic cancer is one of the most aggressive malignant tumors. In recent years, little progress has been made in understanding and treatment of the disease. The two most commonly used chemotherapy drugs approved for the treatment of pancreatic cancer are gemcitabine and fluorouracil. Anti-angiogenic treatment is one of the current promising approaches in cancer translational research. Its aim is to inhibit development of new blood vessels and thereby prevent further tumor growth. We present a first description of a pancreatic cancer patient treated with standard chemotherapy and TL-118, an anti-angiogenic combination of 4 drugs that target non-overlapping aspects of the angiogenic process (Provided by Tiltan Pharma Ltd for compassionate use). Our patient, treated with standard chemotherapy combined with TL-118, was diagnosed about 16 months ago and is still considered progression free, while being treated with that combination. Moreover, when the treatment with TL-118 was stopped, there was a clear elevation of tumor marker which dropped again with the renewal of TL-118. This effect was not achieved by gemcitabine treatment alone. Recently, a phase II clinical trial of TL-118 for pancreatic cancer patients that have not yet been treated with chemotherapy was initiated. (Tiltan Pharma Ltd). The study objective is to evaluate the efficacy, safety, and tolerability of TL-118 in gemcitabine-treated metastatic pancreatic cancer patients. This report describes a new approach in treating pancreatic cancer, enabling patients to obtain a longer progression-free survival using this new anti-angiogenic drug combination, added on standard chemotherapy.
Collapse
Affiliation(s)
- Shani Breuer
- Sharett Institute of Oncology, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| | | | | | | | | |
Collapse
|