1
|
Al-Gizawiy MM, Wujek RT, Alhajala HS, Cobb JM, Prah MA, Doan NB, Connelly JM, Chitambar CR, Schmainda KM. Potent in vivo efficacy of oral gallium maltolate in treatment-resistant glioblastoma. Front Oncol 2024; 13:1278157. [PMID: 38288102 PMCID: PMC10822938 DOI: 10.3389/fonc.2023.1278157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Background Treatment-resistant glioblastoma (trGBM) is an aggressive brain tumor with a dismal prognosis, underscoring the need for better treatment options. Emerging data indicate that trGBM iron metabolism is an attractive therapeutic target. The novel iron mimetic, gallium maltolate (GaM), inhibits mitochondrial function via iron-dependent and -independent pathways. Methods In vitro irradiated adult GBM U-87 MG cells were tested for cell viability and allowed to reach confluence prior to stereotactic implantation into the right striatum of male and female athymic rats. Advanced MRI at 9.4T was carried out weekly starting two weeks after implantation. Daily oral GaM (50mg/kg) or vehicle were provided on tumor confirmation. Longitudinal MRI parameters were processed for enhancing tumor ROIs in OsiriX 8.5.1 (lite) with Imaging Biometrics Software (Imaging Biometrics LLC). Statistical analyses included Cox proportional hazards regression models, Kaplan-Meier survival plots, linear mixed model comparisons, and t-statistic for slopes comparison as indicator of tumor growth rate. Results In this study we demonstrate non-invasively, using longitudinal MRI surveillance, the potent antineoplastic effects of GaM in a novel rat xenograft model of trGBM, as evidenced by extended suppression of tumor growth (23.56 mm3/week untreated, 5.76 mm3/week treated, P < 0.001), a blunting of tumor perfusion, and a significant survival benefit (median overall survival: 30 days untreated, 56 days treated; P < 0.001). The therapeutic effect was confirmed histologically by the presence of abundant cytotoxic cellular swelling, a significant reduction in proliferation markers (P < 0.01), and vessel normalization characterized by prominent vessel pruning, loss of branching, and uniformity of vessel lumina. Xenograft tumors in the treatment group were further characterized by an absence of an invasive edge and a significant reduction in both, MIB-1% and mitotic index (P < 0.01 each). Transferrin receptor and ferroportin expression in GaM-treated tumors illustrated cellular iron deprivation. Additionally, treatment with GaM decreased the expression of pro-angiogenic markers (von Willebrand Factor and VEGF) and increased the expression of anti-angiogenic markers, such as Angiopoietin-2. Conclusion Monotherapy with the iron-mimetic GaM profoundly inhibits trGBM growth and significantly extends disease-specific survival in vivo.
Collapse
Affiliation(s)
- Mona M. Al-Gizawiy
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Robert T. Wujek
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hisham S. Alhajala
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jonathan M. Cobb
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Melissa A. Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ninh B. Doan
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jennifer M. Connelly
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Christopher R. Chitambar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
2
|
Tan S, Chen X, Liu W. Tumor-suppressive role of miR-139-5p in angiogenesis and tumorigenesis of ovarian cancer: Based on GEO microarray analysis and experimental validation. Cell Signal 2023; 109:110730. [PMID: 37244634 DOI: 10.1016/j.cellsig.2023.110730] [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] [Received: 03/07/2023] [Revised: 05/03/2023] [Accepted: 05/21/2023] [Indexed: 05/29/2023]
Abstract
This study clarified the possible molecular mechanisms by which the miR-139-5p/SOX4/TMEM2 axis affected angiogenesis and tumorigenesis of ovarian cancer (OC) based on GEO microarray datasets and experimental support. The expression of miR-139-5p and SOX4 was examined in clinical OC samples. Human umbilical vein endothelial cells (HUVECs) and human OC cell lines were included in vitro experiments. Tube formation assay was conducted in HUVECs. The expression of SOX4, SOX4, and VEGF in OC cells was identified using Western blot and immunohistochemistry. Luciferase assays were conducted to validate the targeting relationship between miR-139-5p and SOX4 and between SOX4 and TMEM2. A RIP assay assessed the binding of SOX4 and miR-139-5p. The impact of miR-139-5p and SOX4 on OC tumorigenesis in vivo was evaluated in nude mice. SOX4 was up-regulated, while miR-139-5p was down-regulated in OC tissues and cells. Ectopic miR-139-5p expression or SOX4 knockdown inhibited angiogenesis and tumorigenicity of OC. By targeting SOX4 in OC, miR-139-5p lowered VEGF expression, angiogenesis, and TMEM2 expression. The miR-139-5p/SOX4/TMEM2 axis also reduced VEGF expression and angiogenesis, which might curtail OC growth in vivo. Collectively, miR-139-5p represses VEGF expression and angiogenesis by targeting the transcription factor SOX4 and down-regulating TMEM2 expression, thereby impeding OC tumorigenesis.
Collapse
Affiliation(s)
- Shu Tan
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Xiuwei Chen
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Wei Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, PR China.
| |
Collapse
|
3
|
Urso G, Boncu AG, Carrara N, Zaman DT, Malfassi L, Marcarini S, Minoli L, Pavesi S, Sala M, Scanziani E, Dolera M. Cranial Spinal Spreading of Canine Brain Gliomas after Hypofractionated Volumetric-Modulated Arc Radiotherapy and Concomitant Temozolomide Chemotherapy: A Four-Case Report. Vet Sci 2022; 9:541. [PMID: 36288154 PMCID: PMC9608067 DOI: 10.3390/vetsci9100541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
Gliomas are the second-most-common primary brain tumors in dogs. Surgery and radiotherapy are established treatment approaches with similar median survival time, whereas conventional chemotherapy is burdened by severe adverse effects. Spinal and leptomeningeal spread of gliomas have been described following radiotherapy treatment alone. The purpose of this study was to evaluate the outcome for four dogs with primary high-grade gliomas in the forebrain without evidence, at diagnosis, of neoplastic invasion along the spinal cord, that were treated with concomitant chemotherapy (temozolomide) and hypofractionated volumetric-modulated arc radiotherapy (VMAT-RT). Temozolomide was selected for its radiosensitive properties, and radiotherapy dose protocols of 37 Gy in 7 fractions or 42 Gy in 10 fractions were used. After an initial complete or partial response, tumors recurred across the cranial-spinal pathway. Post-mortem macroscopic examinations confirmed swollen spinal cord and hyperemic meningeal sleeve, with nodular lesions on the meningeal surface. Microscopically, infiltration of the spinal cord and meninges by neoplastic cells (with features of oligodendrogliomas) were observed. This work seems to suggest that the entire central nervous system should be investigated in diagnostic examinations of canine gliomas. Dose-escalation trials and/or spinal cord prophylaxis treatment could also be evaluated to prevent tumor progression.
Collapse
Affiliation(s)
- Gaetano Urso
- Azienda Socio Sanitaria Territoriale di Lodi, 26900 Lodi, Italy
| | | | - Nancy Carrara
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy
| | - Dragos-Teodor Zaman
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy
| | - Luca Malfassi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy
| | - Silvia Marcarini
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy
| | - Lucia Minoli
- Dipartimento di Scienze Veterinarie, Università degli Studi di Torino, 10095 Grugliasco, Italy
| | - Simone Pavesi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy
| | - Massimo Sala
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy
| | - Eugenio Scanziani
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 20122 Milano, Italy
- Mouse and Animal Pathology Lab (MAPLab), Università degli Studi di Milano–La Statale–Fondazione UniMi, 20122 Milano, Italy
| | - Mario Dolera
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy
| |
Collapse
|
4
|
Liu Y, Li X, Pen R, Zuo W, Chen Y, Sun X, Gou J, Guo Q, Wen M, Li W, Yu S, Liu H, Huang M. Targeted delivery of irinotecan to colon cancer cells using epidermal growth factor receptor-conjugated liposomes. Biomed Eng Online 2022; 21:53. [PMID: 35918704 PMCID: PMC9344698 DOI: 10.1186/s12938-022-01012-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 05/02/2022] [Indexed: 12/31/2022] Open
Abstract
Background CPT-11 (irinotecan) is one of the most efficient agents used for colorectal cancer chemotherapy. However, as for many other chemotherapeutic drugs, how to minimize the side effects of CPT-11 still needs to be thoroughly described. Objectives This study aimed to develop the CPT-11-loaded DSPE-PEG 2000 targeting EGFR liposomal delivery system and characterize its targeting specificity and therapeutic effect on colorectal cancer (CRC) cells in vitro and in vivo. Results The synthesized liposome exhibited spherical shapes (84.6 ± 1.2 nm to 150.4 nm ± 0.8 nm of estimated average sizes), good stability, sustained release, and enough drug loading (55.19%). For in vitro experiments, SW620 cells treated with CPT-11-loaded DSPE-PEG2000 targeting EGFR liposome showed lower survival extended level of intracellular ROS production. In addition, it generated an enhanced apoptotic cell rate by upregulating the protein expression of both cleaved-caspase-3 and cleaved-caspase-9 compared with those of SW620 cells treated with free CPT-11. Importantly, the xenograft model showed that both the non-target and EGFR-targeted liposomes significantly inhibited tumor growth compared to free CPT-11. Conclusions Compared with the non-target CPT-11-loaded DSPE-PEG2000 liposome, CPT-11-loaded DSPE-PEG2000 targeting EGFR liposome treatment showed much better antitumor activity in vitro in vivo. Thus, our findings provide new assets and expectations for CRC targeting therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12938-022-01012-8.
Collapse
Affiliation(s)
- Yongwei Liu
- Department of Infection, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China.
| | - Xinghui Li
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Renqun Pen
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Wei Zuo
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Ya Chen
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Xiuying Sun
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Juhua Gou
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Qianwen Guo
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Maoling Wen
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Wuqi Li
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China
| | - Shuangjiang Yu
- Department of Neurosurgery, The First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, 400038, China
| | - Hao Liu
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.11 Changyuan St Square North Rd, Rongchang District, Chongqing, 402460, China.
| | - Min Huang
- Department of Digestion, The Affiliated Hospital of North Sichuan Medical College, No.1, MaoYuan South Rd, Shunqing District, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
5
|
Jannier S, Kemmel V, Sebastia Sancho C, Chammas A, Sabo AN, Pencreach E, Farace F, Chenard MP, Lhermitte B, Geoerger B, Aerts I, Frappaz D, Leblond P, André N, Ducassou S, Corradini N, Bertozzi AI, Guérin E, Vincent F, Velten M, Entz-Werle N. SFCE-RAPIRI Phase I Study of Rapamycin Plus Irinotecan: A New Way to Target Intra-Tumor Hypoxia in Pediatric Refractory Cancers. Cancers (Basel) 2020; 12:cancers12103051. [PMID: 33092063 PMCID: PMC7656302 DOI: 10.3390/cancers12103051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary More and more relapsing or refractory pediatric cancers are described to present hypoxic features linked to a worse outcome. Therefore, the aim of our phase I study RAPIRI was the targeting of the central node mTor/HIF-1α with rapamycin plus irinotecan and determine the appropriated dose of this combination. As expected, the tolerance was optimal across all dose levels and no maximum tolerated dose of both drugs was reached. The pharmacokinetics (PK) helped us to refine the doses to use in the future phase II trial and the importance of PK follow-up in such combination. We also confirmed in almost half of the interpretable patients for tumor response a non-progressive disease. All those observations additionally to the ancillary’s studies provide strong evidence to propose a next trial focusing on brain tumors and sarcomas and using biweekly 125 mg/m2 irinotecan dose with a PK follow-up and a rapamycin dose of 1.5 mg/m2/day, reaching a blood concentration above 10 µg/L. Abstract Hypoxic environment is a prognostic factor linked in pediatric cancers to a worse outcome, favoring tumor progression and resistance to treatments. The activation of mechanistic Target Of Rapamycin (mTor)/hypoxia inducible factor (HIF)-1α pathway can be targeted by rapamycin and irinotecan, respectively. Therefore, we designed a phase I trial associating both drugs in pediatric refractory/relapsing solid tumors. Patients were enrolled according to a 3 + 3 escalation design with ten levels, aiming to determine the MTD (maximum tolerated dose) of rapamycin plus irinotecan. Rapamycin was administered orally once daily in a 28-day cycle (1 to 2.5 mg/m2/day), associating biweekly intravenous irinotecan (125 to 240 mg/m2/dose). Toxicities, pharmacokinetics, efficacy analyses, and pharmacodynamics were evaluated. Forty-two patients, aged from 2 to 18 years, were included. No MTD was reached. Adverse events were mild to moderate. Only rapamycin doses of 1.5 mg/m2/day reached over time clinically active plasma concentrations. Tumor responses and prolonged stable disease were associated with a mean irinotecan area under the curve of more than 400 min.mg/L. Fourteen out of 31 (45.1%) patients had a non-progressive disease at 8 weeks. Most of them were sarcomas and brain tumors. For the phase II trial, we can then propose biweekly 125 mg/m2 irinotecan dose with a pharmacokinetic (PK) follow-up and a rapamycin dose of 1.5 mg/m2/day, reaching a blood concentration above 10 µg/L.
Collapse
Affiliation(s)
- Sarah Jannier
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (S.J.); (F.V.)
| | - Véronique Kemmel
- Laboratory of Biochemistry, University Hospital of Strasbourg, 67098 Strasbourg, France; (V.K.); (A.-N.S.); (E.G.)
- Laboratory of Pharmacology and Toxicology in Neurocardiology-EA7296, University of Strasbourg, 67000 Strasbourg, France
| | - Consuelo Sebastia Sancho
- Radiology Department, Pediatric Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (C.S.S.); (A.C.)
| | - Agathe Chammas
- Radiology Department, Pediatric Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (C.S.S.); (A.C.)
| | - Amelia-Naomie Sabo
- Laboratory of Biochemistry, University Hospital of Strasbourg, 67098 Strasbourg, France; (V.K.); (A.-N.S.); (E.G.)
- Laboratory of Pharmacology and Toxicology in Neurocardiology-EA7296, University of Strasbourg, 67000 Strasbourg, France
| | - Erwan Pencreach
- Oncobiology Platform, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 67098 Strasbourg, France;
| | - Françoise Farace
- «Circulating Tumor Cells» Translational Platform, Gustave Roussy, University of Paris-Saclay, 94800 Villejuif, France;
| | - Marie Pierre Chenard
- Pathology Department, University Hospital of Strasbourg, 67098 Strasbourg, France; (M.P.C.); (B.L.)
- Centre de Ressources Biologiques, University Hospital of Strasbourg, 67098 Strasbourg, France
| | - Benoit Lhermitte
- Pathology Department, University Hospital of Strasbourg, 67098 Strasbourg, France; (M.P.C.); (B.L.)
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, Department of Pediatric and Adolescent Oncology, Université Paris-Saclay, INSERM U1015, 94800 Villejuif, France;
| | - Isabelle Aerts
- Oncology Center SIREDO, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Didier Frappaz
- Pediatric Oncology Department, Léon Berard Institute, 69373 Lyon, France; (D.F.); (P.L.); (N.C.)
| | - Pierre Leblond
- Pediatric Oncology Department, Léon Berard Institute, 69373 Lyon, France; (D.F.); (P.L.); (N.C.)
- Pediatric Oncology Unit, Oscar Lambret Center, 59020 Lille, France
| | - Nicolas André
- Pediatric Onco-Hematology Unit, CHU La Timone, 13005 Marseille, France;
| | - Stephane Ducassou
- Pediatric Onco-Hematology Department, University Hospital of Bordeaux, 33000 Bordeaux, France;
| | - Nadège Corradini
- Pediatric Oncology Department, Léon Berard Institute, 69373 Lyon, France; (D.F.); (P.L.); (N.C.)
- Pediatric Oncology Unit, University Hospital of Nantes, 44093 Nantes, France
| | - Anne Isabelle Bertozzi
- Pediatric Onco-Hematology Department, University Hospital of Toulouse, 31059 Toulouse, France;
| | - Eric Guérin
- Laboratory of Biochemistry, University Hospital of Strasbourg, 67098 Strasbourg, France; (V.K.); (A.-N.S.); (E.G.)
| | - Florence Vincent
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (S.J.); (F.V.)
| | - Michel Velten
- Clinical Research Department, ICANS, 67200 Strasbourg, France;
| | - Natacha Entz-Werle
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France; (S.J.); (F.V.)
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets, Faculty of Pharmacy, 67401 Illkirch, France
- Correspondence: ; Tel.: +33-3-88-12-83-96
| |
Collapse
|
6
|
Dong J, Meng X, Li S, Chen Q, Shi L, Jiang C, Cai J. Risk of Adverse Vascular Events in Patients with Malignant Glioma Treated with Bevacizumab Plus Irinotecan: A Systematic Review and Meta-Analysis. World Neurosurg 2019; 130:e236-e243. [PMID: 31203059 DOI: 10.1016/j.wneu.2019.06.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Bevacizumab plus irinotecan is a new beneficial chemotherapy strategy for patients with malignant glioma. The purpose of this systematic review and meta-analysis was to comprehensively assess the risk of adverse vascular events in adults with malignant glioma treated with bevacizumab plus irinotecan. METHODS The Cochrane Library, Embase and PubMed were searched, and relevant trials were identified up to June 2018. Two investigators screened all titles and abstracts for possible inclusion and extracted data independently. Six studies were included, and 5 of them in the control group using bevacizumab alone or bevacizumab with temozolomide. Three systems were used to assess the quality of evidence and the level of recommendation. The Oxford Centre for Evidence-Based Medicine Levels of Evidence (2009) system was used to classify the evidence into 5 levels (classes I-V). The star system from the Newcastle-Ottawa Scale was used to assess methodological quality. The GRADE profiler was used to evaluate the overall body of evidence. RESULTS Our data show that bevacizumab plus irinotecan therapy does not significantly affect the risk of systemic adverse events (odds ratio [OR], 1.17; 95% confidence interval [CI], 0.43-3.18). Patients treated with bevacizumab plus irinotecan had a similar risk of hematotoxicity (OR, 1.06; 95% CI, 0.26-4.38), thrombocytopenia (OR, 1.07; 95% CI, 0.25-4.63), and hypertension (OR, 1.34; 95% CI, 0.28-6.36) compared with the control group (those treated without irinotecan). Thrombosis occurred more frequently in patients treated with bevacizumab plus irinotecan compared with the control group (OR, 3.23; 95% CI, 1.47-7.12). CONCLUSIONS The risk of systemic adverse events was not significantly different between patients with malignant glioma treated with bevacizumab plus irinotecan and the control group. The risks of hematotoxicity, thrombocytopenia, and hypertension were similar in the 2 groups. The risk of thrombosis was higher in patients treated with bevacizumab plus irinotecan. Monitoring for thrombosis and administering anticoagulant therapy as necessary merit promotion for patients with malignant glioma receiving treatment with bevacizumab plus irinotecan.
Collapse
Affiliation(s)
- Jiawei Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Siyi Li
- Department of Mining and Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Qun Chen
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lei Shi
- Department of Health Management, School of Public Health, Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
7
|
Natale G, Bocci G. Does metronomic chemotherapy induce tumor angiogenic dormancy? A review of available preclinical and clinical data. Cancer Lett 2018; 432:28-37. [PMID: 29885517 DOI: 10.1016/j.canlet.2018.06.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/11/2018] [Accepted: 06/03/2018] [Indexed: 02/08/2023]
Abstract
Tumor dormancy is the ability of cancer cells to survive in a non-proliferating state. This condition can depend on three main mechanisms: cell cycle arrest (quiescence or cell dormancy), immunosurveillance (immunologic dormancy), or lack of functional blood vessels (angiogenic dormancy). In particular, under angiogenic dormancy, cancer cell proliferation is counterbalanced by apoptosis owing to poor vascularization, impeding tumor mass expansion beyond a microscopic size, with an asymptomatic and non-metastatic state. Tumor vasculogenic or non-angiogenic switch is essential to promote escape from tumor dormancy, leading to tumor mass proliferation and metastasis. In avascular lesions angiogenesis process results blocked from the equilibrium between pro- and anti-angiogenic factors, such as vascular endothelial growth factor (VEGF) and thrombospondin-1 (TSP-1), respectively. The angiogenic switch mainly depends on the disruption of this balance, in favor of pro-angiogenic factors, and on the recruitment of circulating endothelial progenitors (CEPs) that promote the formation of new blood vessels. Metronomic chemotherapy, the regular intake of doses able to sustain low but active concentrations of chemotherapeutic drugs during protracted time periods, is an encouraging therapeutic approach that has shown to upregulate anti-angiogenic factors such as TSP-1 and decline pro-angiogenic factors such as VEGF, suppressing the proangiogenic cells such as CEPs. In this perspective, metronomic chemotherapy may be one of the available therapeutic approaches capable to modulate favorably the angiogenic tumor dormancy, but further research is essential to better define this particular characteristic.
Collapse
Affiliation(s)
- Gianfranco Natale
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, and Museo di Anatomia Umana ''Filippo Civinini'', Università di Pisa, Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy.
| |
Collapse
|
8
|
Resistance to metronomic chemotherapy and ways to overcome it. Cancer Lett 2017; 400:311-318. [PMID: 28259819 DOI: 10.1016/j.canlet.2017.02.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/20/2017] [Accepted: 02/22/2017] [Indexed: 12/24/2022]
Abstract
Therapeutic resistance is amongst the major determinants of cancer mortality. Contrary to initial expectations, antivascular therapies are equally prone to inherent or acquired resistance as other cancer treatment modalities. However, studies into resistance to vascular endothelial growth factor pathway inhibitors revealed distinct mechanisms of resistance compared to conventional cytotoxic therapy. While some of these novel mechanisms of resistance also appear to be functional regarding metronomic chemotherapy, herein we summarize available evidence for mechanisms of resistance specifically described in the context of metronomic chemotherapy. Numerous preclinically identified molecular targets and pathways represent promising avenues to overcome resistance and enhance the benefits achieved with metronomic chemotherapy eventually. However, there are considerable challenges to clinically translate the preclinical findings.
Collapse
|
9
|
Takano S, Ishikawa E, Matsuda M, Sakamoto N, Akutsu H, Yamamoto T, Matsumura A. The anti-angiogenic role of soluble-form VEGF receptor in malignant gliomas. Int J Oncol 2016; 50:515-524. [PMID: 28000842 DOI: 10.3892/ijo.2016.3810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/21/2016] [Indexed: 11/05/2022] Open
|
10
|
Tianqin G, Chunlei C, Jingjing W. Synergistic Anti-glioma Effects in Vitro and in Vivo of Enediyne Antibiotic Neocarzinostatin and Paclitaxel via Enhanced Growth Delay and Apoptosis-Induction. Biol Pharm Bull 2016; 39:1623-1630. [DOI: 10.1248/bpb.b16-00285] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
11
|
Osuka S, Takano S, Watanabe S, Ishikawa E, Yamamoto T, Matsumura A. Valproic acid inhibits angiogenesis in vitro and glioma angiogenesis in vivo in the brain. Neurol Med Chir (Tokyo) 2013; 52:186-93. [PMID: 22522328 DOI: 10.2176/nmc.52.186] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antiangiogenic strategy is promising for malignant glioma. Histone deacetylase inhibitors (HDACIs) are unique anticancer agents that exhibit antiangiogenic effects. The in vitro and in vivo antiangiogenic effects of HDACIs, valproic acid (VPA), were investigated in malignant glioma in the brain. In vitro, VPA preferentially inhibited endothelial cell proliferation compared to glioma cell proliferation at the optimum concentration in a dose-dependent manner. VPA reduced vascular endothelial growth factor (VEGF) secretion of glioma cells in a dose-dependent manner under both normoxic and hypoxic conditions. VPA was also found to inhibit tube formation in the angiogenesis assay. In vivo, treatment with VPA combined with irinotecan reduced the number of vessels expressing factor VIII in the brain tumor model. VPA inhibits glioma angiogenesis by direct (inhibition of endothelial cell proliferation and tube formation) and indirect (decreased secretion of VEGF by glioma cells) mechanisms. These data suggest a potential role for VPA as an adjuvant therapy for patients with malignant glioma.
Collapse
Affiliation(s)
- Satoru Osuka
- Department of Neurosurgery, Institute of Clinical Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | |
Collapse
|
12
|
Jarzabek MA, Huszthy PC, Skaftnesmo KO, McCormack E, Dicker P, Prehn JH, Bjerkvig R, Byrne AT. In Vivo Bioluminescence Imaging Validation of a Human Biopsy–Derived Orthotopic Mouse Model of Glioblastoma Multiforme. Mol Imaging 2013. [DOI: 10.2310/7290.2012.00029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Monika A. Jarzabek
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| | - Peter C. Huszthy
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| | - Kai O. Skaftnesmo
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| | - Emmet McCormack
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| | - Patrick Dicker
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| | - Jochen H.M. Prehn
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| | - Rolf Bjerkvig
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| | - Annette T. Byrne
- From the Department of Physiology and Medical Physics, Centre for Systems Medicine, and PHS Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; NorLux Neuro-oncology Laboratory, Department of Biomedicine and Institute of Medicine, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway; and University College Dublin, Conway Institute, Belfield, Dublin, Ireland
| |
Collapse
|
13
|
Semenza GL. Cancer-stromal cell interactions mediated by hypoxia-inducible factors promote angiogenesis, lymphangiogenesis, and metastasis. Oncogene 2012; 32:4057-63. [PMID: 23222717 DOI: 10.1038/onc.2012.578] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/09/2012] [Indexed: 12/13/2022]
Abstract
Interactions between cancer cells and stromal cells, including blood vessel endothelial cells (BECs), lymphatic vessel endothelial cells (LECs), bone marrow-derived angiogenic cells (BMDACs) and other bone marrow-derived cells (BMDCs) play important roles in cancer progression. Intratumoral hypoxia, which affects both cancer and stromal cells, is associated with a significantly increased risk of metastasis and mortality in many human cancers. Recent studies have begun to delineate the molecular mechanisms underlying the effect of intratumoral hypoxia on cancer progression. Reduced O2 availability induces the activity of hypoxia-inducible factors (HIFs), which activate the transcription of target genes encoding proteins that play important roles in many critical aspects of cancer biology. Included among these are secreted factors, including angiopoietin 2, angiopoietin-like 4, placental growth factor, platelet-derived growth factor B, stem cell factor (kit ligand), stromal-derived factor 1, and vascular endothelial growth factor. These factors are produced by hypoxic cancer cells and directly mediate functional interactions with BECs, LECs, BMDACs and other BMDCs that promote angiogenesis, lymphangiogenesis, and metastasis. In addition, lysyl oxidase (LOX) and LOX-like proteins, which are secreted by hypoxic breast cancer cells, remodel extracellular matrix in the lungs, which leads to BMDC recruitment and metastatic niche formation.
Collapse
Affiliation(s)
- G L Semenza
- Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
14
|
Waterhouse DN, Yapp D, Verreault M, Anantha M, Sutherland B, Bally MB. Lipid-based nanoformulation of irinotecan: dual mechanism of action allows for combination chemo/angiogenic therapy. Nanomedicine (Lond) 2012; 6:1645-54. [PMID: 22077466 DOI: 10.2217/nnm.11.140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A number of studies have outlined the antiangiogenic effects of cytotoxic agents when administered frequently at low doses. These studies suggest that the effect of the cytotoxic agent is on the vasculature within the tumor and it is assumed that there is little or negligible cytotoxicity. Liposomal drug delivery systems have the ability to provide a dual mechanism of activity where tumor accumulation can deliver high local concentrations of the drug at the site of action with concomitant slow release of the drug from carriers in the blood compartment that results in antivascular effects, similar to that achieved when dosing frequently at low levels. Although this dual mechanism of activity may be linked to other lipid nanoparticle formulations of anticancer drugs, this article summarizes the evidence supporting direct (cytotoxic) and indirect (antivascular) actions of a liposomal formulation of irinotecan.
Collapse
Affiliation(s)
- Dawn N Waterhouse
- BC Cancer Agency, Department of Experimental Therapeutics, 675 West 10th Avenue, Vancouver BC Canada, V5Z 1L3.
| | | | | | | | | | | |
Collapse
|
15
|
Guérin E, Raffelsberger W, Pencreach E, Maier A, Neuville A, Schneider A, Bachellier P, Rohr S, Petitprez A, Poch O, Moras D, Oudet P, Larsen AK, Gaub MP, Guenot D. In vivo topoisomerase I inhibition attenuates the expression of hypoxia-inducible factor 1α target genes and decreases tumor angiogenesis. Mol Med 2012; 18:83-94. [PMID: 22033674 DOI: 10.2119/molmed.2011.00120] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 10/10/2011] [Indexed: 02/04/2023] Open
Abstract
Topoisomerase I is a privileged target for widely used anticancer agents such as irinotecan. Although these drugs are classically considered to be DNA-damaging agents, increasing evidence suggests that they might also influence the tumor environment. This study evaluates in vivo cellular and molecular modifications induced by irinotecan, a topoisomerase I-directed agent, in patient-derived colon tumors subcutaneously implanted in athymic nude mice. Irinotecan was given intraperitoneally at 40 mg/kg five times every 5 d, and expression profiles were evaluated at d 25 in tumors from treated and untreated animals. Unexpectedly, the in vivo antitumor activity of irinotecan was closely linked to a downregulation of hypoxia-inducible factor-1α (HIF1A) target genes along with an inhibition of HIF1A protein accumulation. The consequence was a decrease in tumor angiogenesis leading to tumor size stabilization. These results highlight the molecular basis for the antitumor activity of a widely used anticancer agent, and the method used opens the way for mechanistic studies of the in vivo activity of other anticancer therapies.
Collapse
Affiliation(s)
- Eric Guérin
- EA 4438 Physiopathologie et Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Albertsson P, Lennernäs B, Norrby K. Low-dosage metronomic chemotherapy and angiogenesis: topoisomerase inhibitors irinotecan and mitoxantrone stimulate VEGF-A-mediated angiogenesis. APMIS 2011; 120:147-56. [PMID: 22229270 PMCID: PMC3321228 DOI: 10.1111/j.1600-0463.2011.02830.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Albertsson P, Lennernäs B, Norrby K. Low-dosage metronomic chemotherapy and angiogenesis: topoisomerase inhibitors irinotecan and mitoxantrone stimulate VEGF-A-mediated angiogenesis. APMIS 2011. Metronomic chemotherapy with cytotoxic agents has been shown to inhibit angiogenesis and, consequently, tumor growth by targeting vascular endothelial cells (ECs). In these regimens, anti-tumor activities additional to anti-angiogenesis may operate. Moreover, chemotherapy typically generates reactive oxygen species in targeted ECs, which can affect angiogenesis. The aim of the present study was to assess the systemic effect of low-dosage metronomic treatment with either irinotecan or mitoxantrone on angiogenesis induced by VEGF-A. Angiogenesis was induced in normal adult rat mesentery by intraperitoneal injection of a low dosage of VEGF-A. Thereafter, irinotecan and mitoxantrone were infused separately continuously at minimally toxic dosages for 14 consecutive days via a subcutaneous osmotic minipump. Angiogenesis was assessed in terms of objective and quantitative variables using morphologic and computerized image analyses. Irinotecan or mitoxantrone significantly stimulated angiogenesis, with ironotecan increasing angiogenesis by 104%, when compared with the vehicle-treated animals. Low-dosage metronomic chemotherapy with irinotecan or mitoxantrone stimulates angiogenesis in the normal mesentery of rats, probably by inducing low-level oxidative stress in the targeted ECs. Whether or not this pertains to tumor angiogenesis may be difficult to confirm, as several anti-tumor modes may operate during low-dosage metronomic chemotherapy.
Collapse
Affiliation(s)
- Per Albertsson
- Department of Oncology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | | |
Collapse
|
17
|
Kavitha CV, Agarwal C, Agarwal R, Deep G. Asiatic acid inhibits pro-angiogenic effects of VEGF and human gliomas in endothelial cell culture models. PLoS One 2011; 6:e22745. [PMID: 21826202 PMCID: PMC3149605 DOI: 10.1371/journal.pone.0022745] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 07/03/2011] [Indexed: 01/05/2023] Open
Abstract
Malignant gliomas are one of the most devastating and incurable tumors. Sustained excessive angiogenesis by glioma cells is the major reason for their uncontrolled growth and resistance toward conventional therapies resulting in high mortality. Therefore, targeting angiogenesis should be a logical strategy to prevent or control glioma cell growth. Earlier studies have shown that Asiatic Acid (AsA), a pentacyclic triterpenoid, is effective against glioma and other cancer cells; however, its efficacy against angiogenesis remains unknown. In the present study, we examined the anti-angiogenic efficacy of AsA using human umbilical vein endothelial cells (HUVEC) and human brain microvascular endothelial cells (HBMEC). Our results showed that AsA (5–20 µM) inhibits HUVEC growth and induces apoptotic cell death by activating caspases (3 and 9) and modulating the expression of apoptosis regulators Bad, survivin and pAkt-ser473. Further, AsA showed a dose-dependent inhibition of HUVEC migration, invasion and capillary tube formation, and disintegrated preformed capillary network. AsA also inhibited the VEGF-stimulated growth and capillary tube formation by HUVEC and HBMEC. Next, we analyzed the angiogenic potential of conditioned media collected from human glioma LN18 and U87-MG cells treated with either DMSO (control conditioned media, CCM) or AsA 20 µM (AsA20 conditioned media, AsA20CM). CCM from glioma cells significantly enhanced the capillary tube formation in both HUVEC and HBMEC, while capillary tube formation in both endothelial cell lines was greatly compromised in the presence of AsA20CM. Consistent with these results, VEGF expression was lesser in AsA20CM compared to CCM, and indeed AsA strongly inhibited VEGF level (both cellular and secreted) in glioma cells. AsA also showed dose-dependent anti-angiogenic efficacy in Matrigel plug assay, and inhibited the glioma cells potential to attract HUVEC/HBMEC. Overall, the present study clearly showed the strong anti-angiogenic potential of AsA and suggests its usefulness against malignant gliomas.
Collapse
Affiliation(s)
- Chandagirikoppal V. Kavitha
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Denver, Colorado, United States of America
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Denver, Colorado, United States of America
- University of Colorado Cancer Center, Aurora, Colorado, United States of America
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Denver, Colorado, United States of America
- University of Colorado Cancer Center, Aurora, Colorado, United States of America
| | - Gagan Deep
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Denver, Colorado, United States of America
- University of Colorado Cancer Center, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
18
|
Choijamts B, Naganuma Y, Nakajima K, Kawarabayashi T, Miyamoto S, Tachibana K, Emoto M. Metronomic irinotecan chemotherapy combined with ultrasound irradiation for a human uterine sarcoma xenograft. Cancer Sci 2011; 102:452-9. [PMID: 21205068 PMCID: PMC11158250 DOI: 10.1111/j.1349-7006.2010.01807.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 11/08/2010] [Accepted: 11/12/2010] [Indexed: 11/30/2022] Open
Abstract
Metronomic chemotherapy is the frequent administration of low doses of chemotherapeutic agents targeting tumor-associated endothelial cells. We examined the efficacy of metronomic irinotecan combined with low-intensity ultrasound (US) in human uterine sarcoma and evaluated its antiangiogenesis mechanism by measuring the circulating endothelial progenitor cells (CEP), a surrogate marker of angiogenesis. A human uterine sarcoma cell line, FU-MMT-3, was used in the present study because this tumor is one of the most malignant neoplasms of human solid tumors and it also has a high angiogenesis property. The combination of low-dose irinotecan and US irradiation significantly inhibited the tube formation of HUVEC and vascular endothelial growth factor expression of tumor cells in vitro. The FU-MMT-3 xenografts in nude mice were treated using US at a low intensity (2.0 w/cm(2), 1 MHz) for 4 min three times per week each after the intraperitoneal administration of irinotecan; this treatment was continued for 5 weeks. The tumor vascularity was assessed by contrast-enhanced color Doppler US in real time. The combination treatment significantly inhibited the mobilization of CEP and intratumoral vascularity compared with the control. This combination therapy showed a significant reduction in tumor volume, resulting in a significant prolongation of survival, in comparison with each treatment alone. These results suggest that the effect of metronomic chemotherapy for human uterine sarcoma was accelerated by US irradiation in vivo and this combination might therefore be potentially effective for new cancer therapy.
Collapse
Affiliation(s)
- Batsuren Choijamts
- Departments of Obstetrics and Gynecology, School of Medicine, Fukuoka University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|