1
|
El-Ghiaty MA, Alqahtani MA, El-Mahrouk SR, Isse FA, Alammari AH, El-Kadi AOS. Alteration of Hepatic Cytochrome P450 Expression and Arachidonic Acid Metabolism by Arsenic Trioxide (ATO) in C57BL/6 Mice. Biol Trace Elem Res 2025; 203:1000-1015. [PMID: 38758479 DOI: 10.1007/s12011-024-04225-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024]
Abstract
The success of arsenic trioxide (ATO) in acute promyelocytic leukemia has driven a plethora studies to investigate its efficacy in other malignancies. However, the inherent toxicity of ATO limits the expansion of its clinical applications. Such toxicity may be linked to ATO-induced metabolic derangements of endogenous substrates. Therefore, the primary objective of this study was to investigate the effect of ATO on the hepatic formation of arachidonic acid (AA) metabolites, hydroxyeicosatetraenoic acids (HETEs), as well as their most notable producing machinery, cytochrome P450 (CYP) enzymes. For this purpose, C57BL/6 mice were intraperitoneally injected with 8 mg/kg ATO for 6 and 24 h. Total RNA was extracted from harvested liver tissues for qPCR analysis of target genes. Hepatic microsomal proteins underwent incubation with AA, followed by identification/quantification of the produced HETEs. ATO downregulated Cyp2e1, while induced Cyp2j9 and most of Cyp4a and Cyp4f, and this has resulted in a significant increase in 17(S)-HETE and 18(R)-HETE, while significantly decreased 18(S)-HETE. Additionally, ATO induced Cyp4a10, Cyp4a14, Cyp4f13, Cyp4f16, and Cyp4f18, resulting in a significant elevation in 20-HETE formation. In conclusion, ATO altered hepatic AA metabolites formation through modulating the underlying network of CYP enzymes. Modifying the homeostatic production of bioactive AA metabolites, such as HETEs, may entail toxic events that can, at least partly, explain ATO-induced hepatotoxicity. Such modification can also compromise the overall body tolerability to ATO treatment in cancer patients.
Collapse
Affiliation(s)
- Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada
| | - Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada
| | - Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada
| | - Fadumo A Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada
| | - Ahmad H Alammari
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
2
|
El-Mahrouk SR, El-Ghiaty MA, Alqahtani MA, El-Kadi AOS. Arsenic Trioxide (ATO III) Induces NAD(P)H Quinone Oxidoreductase 1 (NQO1) Expression in Hepatic and Extrahepatic Tissues of C57BL/6 Mice. Chem Res Toxicol 2024; 37:2040-2051. [PMID: 39630573 DOI: 10.1021/acs.chemrestox.4c00422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Arsenic trioxide (ATOIII) has emerged as a potent therapeutic agent for acute promyelocytic leukemia (APL), yet its clinical application is often limited by significant adverse effects. This study investigates the molecular mechanisms underlying ATOIII's impact on cellular detoxification pathways, focusing on the regulation of NAD(P)H/quinone oxidoreductase (NQO1), a crucial enzyme in maintaining cellular homeostasis and cancer prevention. We explored ATOIII's effects on NQO1 expression in C57BL/6 mice and Hepa-1c1c7 cells, both independently and in combination with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a known NQO1 inducer. Our findings revealed that ATOIII significantly increased NQO1 expression in hepatic and extrahepatic tissues, as well as in Hepa-1c1c7 cells, at mRNA, protein, and activity levels. This upregulation occurred both in the presence and absence of TCDD. Mechanistically, we demonstrated that ATOIII promotes the nuclear translocation of both nuclear factor erythroid 2-related factor-2 (NRF2) and aryl hydrocarbon receptor (AHR) transcription factors. Furthermore, ATOIII exposure increased antioxidant response element (ARE)-driven reporter gene activity, indicating a transcriptional mechanism of NQO1 induction. Notably, gene silencing experiments confirmed the critical roles of both NRF2 and AHR in mediating ATOIII-induced NQO1 expression. In conclusion, ATOIII exposure is found to upregulate the NQO1 enzyme through a transcriptional mechanism via AHR- and NRF2- dependent mechanisms, offering valuable insights into its therapeutic mechanisms.
Collapse
Affiliation(s)
- Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
- Faculty of Pharmacy, Tanta University, Gharbia, Tanta 31111, Egypt
| | - Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
| | - Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
| |
Collapse
|
3
|
Li ZZ, He JY, Wu Q, Liu B, Bu LL. Recent advances in targeting myeloid-derived suppressor cells and their applications to radiotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:233-264. [PMID: 37438019 DOI: 10.1016/bs.ircmb.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterogenous immature myeloid cells with potent immune suppressive properties that not only constrain anti-tumor immune activation and functions, promote tumor progression, but also contribute to treatment resistance and tumor relapse. Targeting MDSCs may be a promising new cancer treatment method, but there is still a problem of low treatment efficiency. Combined application with radiotherapy may be a potential method to solve this problem. Drug delivery systems (DDSs) provide more efficient targeted drug delivery capability and can reduce the toxicity and side effects of drugs. Recent advance in DDSs targeting development, recruitment, differentiation, and elimination of MDSCs have shown promising effect in reversing immune inhibition and in overcoming radiotherapy resistance. In this review, we systematically summarized DDSs applied to target MDSCs for the first time, and classified and discussed it according to its different mechanisms of action. In addition, this paper also reviewed the biological characteristics of MDSCs and their role in the initiation, progression, and metastasis of cancer. Moreover, this review also summarizes the role of DDSs targeting MDSCs in radiosensitization. Finally, the future development of DDSs targeting MDSCs is also prospected.
Collapse
Affiliation(s)
- Zi-Zhan Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Jing-Yu He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Bing Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
4
|
El-Ghiaty MA, Alqahtani MA, El-Kadi AOS. Down-regulation of hepatic cytochromes P450 1A1 and 1A2 by arsenic trioxide (ATO) in vivo and in vitro: A role of heme oxygenase 1. Chem Biol Interact 2022; 364:110049. [PMID: 35872050 DOI: 10.1016/j.cbi.2022.110049] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/03/2022] [Accepted: 07/13/2022] [Indexed: 11/03/2022]
Abstract
Arsenic trioxide (ATO) has evolved from an environmental threat to a successful therapy for acute promyelocytic leukemia (APL) and probably for solid tumors in the future. However, its efficacy comes at a cost of multi-organ toxicity whose mechanism remains unresolved. Arsenicals have been reported to modulate cytochrome P450 1A (CYP1A) enzymes, thus modifying activation/detoxification of drugs/procarcinogens. Therefore, this study aimed to investigate the possible effects of ATO on CYP1A1 and CYP1A2, in absence and presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) using in vivo and in vitro models. For this purpose, C57BL/6 mice were intraperitoneally injected with 8 mg/kg ATO with or without 15 μg/kg TCDD for 6 and 24 h. Furthermore, HepG2 cells were treated with ATO (1, 5, and 10 μM) with or without 1 nM TCDD for 6 and 24 h. ATO significantly inhibited TCDD-mediated induction of CYP1A1/1A2 mRNA, protein, and activity in both models. ATO differentially modulated CYP1A1/1A2 basal levels in vivo. We also demonstrated that ATO downregulates CYP1A through inhibiting the transcriptional activation of its regulatory element at both basal and inducible levels. Additionally, ATO significantly induced mRNA and protein of heme oxygenase 1 (HMOX1) in vivo and in vitro. In HepG2 cells, inhibition of HMOX1 by tin (IV) mesoporphyrin (IX) (SnMP) resulted in a partial restoration of the TCDD-mediated induction of CYP1A1 activity that was inhibited by ATO co-exposure. Our findings show that ATO alters both constitutive and inducible CYP1A1/1A2 expressions through transcriptional and HMOX1-mediated post-translational mechanisms. This implies the possible involvement of ATO in clearance-related consequences for the substrates of these enzymes such as drug-drug interactions or suboptimal toxicant elimination.
Collapse
Affiliation(s)
- Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
5
|
Tissue Architecture Influences the Biological Effectiveness of Boron Neutron Capture Therapy in In Vitro/In Silico Three-Dimensional Self-Assembly Cell Models of Pancreatic Cancers. Cancers (Basel) 2021; 13:cancers13164058. [PMID: 34439214 PMCID: PMC8394840 DOI: 10.3390/cancers13164058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Boron neutron capture therapy (BNCT) is becoming one of the most promising radiotherapies for aggressive cancers, but the detailed cellular mechanisms of BNCT remain largely underexplored. Solid tumors are composed of heterogeneous cell populations, which create a 3-dimensional complicated microenvironment for tumor progression. To recapture the influences of the microenvironment on BNCT efficacy, we applied a self-assembly 3D cell culture system with two different types of pancreatic cancer cells. In contrast to previous findings with γ-ray exposure, we found that the 3D architecture of pancreatic tumor can facilitate the susceptibility of cancer cells to BNCT, as compared to 2D tissue structure; a computer simulation model was established to further confirm this unexpected finding. These outcomes can contribute to better understanding of the radiobiology of BNCT, and the developed models may facilitate the recent development in personalized radiotherapy. Abstract Pancreatic cancer is a leading cause of cancer death, and boron neutron capture therapy (BNCT) is one of the promising radiotherapy techniques for patients with pancreatic cancer. In this study, we evaluated the biological effectiveness of BNCT at multicellular levels using in vitro and in silico models. To recapture the phenotypic characteristic of pancreatic tumors, we developed a cell self-assembly approach with human pancreatic cancer cells Panc-1 and BxPC-3 cocultured with MRC-5 fibroblasts. On substrate with physiological stiffness, tumor cells self-assembled into 3D spheroids, and the cocultured fibroblasts further facilitated the assembly process, which recapture the influence of tumor stroma. Interestingly, after 1.2 MW neutron irradiation, lower survival rates and higher apoptosis (increasing by 4-fold for Panc-1 and 1.5-fold for BxPC-3) were observed in 3D spheroids, instead of in 2D monolayers. The unexpected low tolerance of 3D spheroids to BNCT highlights the unique characteristics of BNCT over conventional radiotherapy. The uptake of boron-containing compound boronophenylalanine (BPA) and the alteration of E-cadherin can partially contribute to the observed susceptibility. In addition to biological effects, the probability of induced α-particle exposure correlated to the multicellular organization was speculated to affect the cellular responses to BNCT. A Monte Carlo (MC) simulation was also established to further interpret the observed survival. Intracellular boron distribution in the multicellular structure and related treatment resistance were reconstructed in silico. Simulation results demonstrated that the physical architecture is one of the essential factors for biological effectiveness in BNCT, which supports our in vitro findings. In summary, we developed in vitro and in silico self-assembly 3D models to evaluate the effectiveness of BNCT on pancreatic tumors. Considering the easy-access of this 3D cell-assembly platform, this study may not only contribute to the current understanding of BNCT but is also expected to be applied to evaluate the BNCT efficacy for individualized treatment plans in the future.
Collapse
|
6
|
Abstract
Arsenic trioxide (ATO) is among the first-line chemotherapeutic drugs used in oncological practice. It has shown substantial efficacy in treating patients with relapsed or refractory acute promyelocytic leukaemia. The clinical use of ATO is hampered due to cardiotoxicity and hence many patients are precluded from receiving this highly effective treatment. An alternative to this would be to use any drug that can ameliorate the cardiotoxic effects and allow exploiting the full therapeutic potential of ATO, with considerable impact on cancer therapy. Generation of reactive oxygen species is involved in a wide range of human diseases, including cancer, cardiovascular, pulmonary and neurological disorders. Hence, agents with the ability to protect against these reactive species may be therapeutically useful. The present review focuses on the beneficial as well as harmful effects of arsenic and ATO, the mechanisms underlying ATO toxicity and the possible ways that can be adopted to circumvent ATO-induced toxicity.
Collapse
|
7
|
Saad-El-Din AA, El-Tanahy ZH, El-Sayed SN, Anees LM, Farroh HA. Study of electron spin resonance and viscosity for hemoglobin polymer after arsenic trioxide and gamma irradiation treatment. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2019. [DOI: 10.1016/j.jrras.2014.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Aisha A. Saad-El-Din
- Biophys., Lab. Rad. Phys. Dep. National Center of Radiation Research and Technology (NCRRT), AEA, P.O. Box 29, Madinat Nasr, Cairo, Egypt
| | - Zinab H. El-Tanahy
- Nucl. Phys. Dep. Faculty of Science, Al-Azhar University, Girls branch, Cairo, Egypt
| | - Suzan N. El-Sayed
- Solid Stat. Phys. Dep. Faculty of Science, Al-Azhar University, Girls branch, Cairo, Egypt
| | - Laila M. Anees
- Health Res. Dep. National Center of Radiation Research and Technology (NCRRT), AEA, P.O. Box 29, Madinat Nasr, Cairo, Egypt
| | - Hoda A. Farroh
- Health Res. Dep. National Center of Radiation Research and Technology (NCRRT), AEA, P.O. Box 29, Madinat Nasr, Cairo, Egypt
| |
Collapse
|
8
|
Xie L, Guo W, Tang X, Yang Y, Xu J. Effects of Arsenic Trioxide on Minor Progressive High-Grade Osteosarcoma of the Extremities Metastatic to the Lung: Results of 39 Patients Treated in a Single Institution. Case Rep Oncol 2016; 9:610-628. [PMID: 27920692 PMCID: PMC5118832 DOI: 10.1159/000448705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 07/26/2016] [Indexed: 01/06/2023] Open
Abstract
Patients who mildly progressed after first-line chemotherapy were administered arsenic trioxide (ATO) 5–10 mg intravenously daily. Thirty-nine patients were finally enrolled in the study, of whom 19 patients received first-line chemotherapy with ATO infusion while 20 patients did not. Progression-free survival at 4 months was 89.2 and 62.7% (p = 0.043) for the ATO group and the control group, respectively, while the 2-year overall survival was 61 and 16.4% (p = 0.032).
Collapse
Affiliation(s)
- Lu Xie
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| | - Xiaodong Tang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| | - Yi Yang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| | - Jie Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| |
Collapse
|
9
|
Zhang L, Zhang Z, Mason RP, Sarkaria JN, Zhao D. Convertible MRI contrast: Sensing the delivery and release of anti-glioma nano-drugs. Sci Rep 2015; 5:9874. [PMID: 25962872 PMCID: PMC4428068 DOI: 10.1038/srep09874] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/19/2015] [Indexed: 12/27/2022] Open
Abstract
There is considerable interest in developing nanohybrids of imaging contrast agents and drugs for image-guided drug delivery. We have developed a strategy of utilizing manganese (Mn) to enhance the nano-encapsulation of arsenic trioxide (ATO). Formation of arsenite (As3+)-Mn precipitates in liposomes generates magnetic susceptibility effects, reflected as dark contrast on T2-weighted MRI. Intriguingly, following cell uptake, the As-Mn complex decomposes in response to low pH in endosome-lysosome releasing ionic As3+, the active form of ATO, and Mn2+, the T1 contrast agent that gives a bright signal. Glioblastoma (GBM) is well known for its high resistance to chemotherapy, e.g., temozolomide (TMZ). Building upon the previously established phosphatidylserine (PS)-targeted nanoplatform that has excellent GBM-targeting specificity, we now demonstrate the effectiveness of the targeted nanoformulated ATO for treating TMZ-resistant GBM cells and the ability of the convertible Mn contrast as a surrogate revealing the delivery and release of ATO.
Collapse
Affiliation(s)
- Liang Zhang
- Radiology, UT Southwestern Medical Center, Dallas, TX
| | | | - Ralph P Mason
- Radiology, UT Southwestern Medical Center, Dallas, TX
| | | | - Dawen Zhao
- Radiology, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
10
|
Boyko-Fabian M, Niehr F, Distel L, Budach V, Tinhofer I. Increased growth-inhibitory and cytotoxic activity of arsenic trioxide in head and neck carcinoma cells with functional p53 deficiency and resistance to EGFR blockade. PLoS One 2014; 9:e98867. [PMID: 24927258 PMCID: PMC4057125 DOI: 10.1371/journal.pone.0098867] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 05/08/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Mutations in the p53 gene are frequently observed in squamous cell carcinoma of the head and neck region (SCCHN) and have been associated with drug resistance. The potential of arsenic trioxide (ATO) for treatment of p53-deficient tumor cells and those with acquired resistance to cisplatin and cetuximab was determined. MATERIAL AND METHODS In a panel of 10 SCCHN cell lines expressing either wildtype p53, mutated p53 or which lacked p53 by deletion the interference of p53 deficiency with the growth-inhibitory and radiosensitizing potential of ATO was determined. The causal relationship between p53 deficiency and ATO sensitivity was evaluated by reconstitution of wildtype p53 in p53-deficient SCCHN cells. Interference of ATO treatment with cell cycle, DNA repair and apoptosis and its efficacy in cells with acquired resistance to cisplatin and cetuximab was evaluated. RESULTS Functional rather than structural defects in the p53 gene predisposed tumor cells to increased sensitivity to ATO. Reconstitution of wt p53 in p53-deficient SCCHN cells rendered them less sensitive to ATO treatment. Combination of ATO with irradiation inhibited clonogenic growth in an additive manner. The inhibitory effect of ATO in p53-deficient tumor cells was mainly associated with DNA damage, G2/M arrest, upregulation of TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) receptors and apoptosis. Increased activity of ATO was observed in cetuximab-resistant SCCHN cells whereas cisplatin resistance was associated with cross-resistance to ATO. CONCLUSIONS Addition of ATO to treatment regimens for p53-deficient SCCHN and tumor recurrence after cetuximab-containing regimens might represent an attractive strategy in SCCHN.
Collapse
Affiliation(s)
- Mariya Boyko-Fabian
- Translational Radiooncology Laboratory, Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Franziska Niehr
- Translational Radiooncology Laboratory, Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Luitpold Distel
- Department of Radiation Oncology, University Erlangen-Nuremberg, Erlangen, Germany
| | - Volker Budach
- Translational Radiooncology Laboratory, Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Ingeborg Tinhofer
- Translational Radiooncology Laboratory, Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
- * E-mail:
| |
Collapse
|
11
|
Florea AM, Büsselberg D. The two opposite facets of arsenic: toxic and anticancer drug. ACTA ACUST UNITED AC 2013. [DOI: 10.5339/jlghs.2013.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Arsenic compounds have been known and used for centuries but their effects in living organisms still represent a large unknown. Arsenic compounds have paradoxical effects: they are threatening to human health, especially upon long-term exposure that can induce the development of cancer; however, they are used as drugs against cancer. This review focuses on the effects shown by clinically and environmentally relevant arsenic compounds in living organisms with a focus on the calcium–apoptosis link.
Collapse
Affiliation(s)
- Ana-Maria Florea
- 1Universität Trier, Fachbereich VI Umwelttoxikologie, Universitätsring 15 54296 Trier, Germany
| | - Dietrich Büsselberg
- 2Weill Cornell Medical College in Qatar, Qatar Foundation – Education City, P.O. Box 24144, Doha, Qatar
| |
Collapse
|
12
|
Kothari S, Herzig G, Slone S, Herzig R. Multiple isolated extramedullary relapse of acute promyelocytic leukemia after allogeneic hematopoietic stem cell transplant: a case report and review of literature. SPRINGERPLUS 2013; 2:49. [PMID: 23476896 PMCID: PMC3586393 DOI: 10.1186/2193-1801-2-49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 02/09/2013] [Indexed: 12/12/2022]
Abstract
Isolated extramedullary disease (EMD) is uncommon, especially in acute promyelocytic leukemia (APL) after allogeneic hematopoietic stem cell transplantation (HSCT). We review the literature and present a 32 year old woman with APL who developed multiple EMDs after allogeneic HSCT within the calvarium, and later found to have various isolated lesions including femur, humerus and thoraco lumbar vertebrae. She was treated with local radiotherapy (XRT) to EMD lesions, all-trans retinoic acid, arsenic trioxide and donor lymphocyte infusion at different time points in her clinical course, without success. Out of reported cases in clinical setting as ours, average onset of isolated EMD is 25 months and median survival 14 months. Effective treatment of isolated EMD after HSCT is not yet clear, but ATO in combination with local XRT, tamibarotene and second HSCT have shown good results in some reported cases, but accumulation of more cases is needed to elucidate optimal therapy in such setting.
Collapse
Affiliation(s)
- Shalin Kothari
- Division of Blood and Bone Marrow Transplant, University of Louisville, James Graham Brown Cancer Center, 529 South Jackson Street, Louisville, KY 40202 USA
| | | | | | | |
Collapse
|
13
|
Arsenic Trioxide as a Vascular Disrupting Agent: Synergistic Effect with Irinotecan on Tumor Growth Delay in a CT26 Allograft Model. Transl Oncol 2013; 6:83-91. [PMID: 23418620 DOI: 10.1593/tlo.12322] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/22/2012] [Accepted: 11/25/2012] [Indexed: 11/18/2022] Open
Abstract
The mechanism of action of arsenic trioxide (ATO) has been shown to be complex, influencing numerous signal transduction pathways and resulting in a vast range of cellular effects. Among these mechanisms of action, ATO has been shown to cause acute vascular shutdown and massive tumor necrosis in a murine solid tumor model like vascular disrupting agent (VDA). However, relatively little is understood about this VDA-like property and its potential utility in developing clinical regimens. We focused on this VDA-like action of ATO. On the basis of the endothelial cell cytotoxicity assay and tubulin polymerization assay, we observed that higher concentrations and longer treatment with ATO reduced the level of α- and β-tubulin and inhibited the polymerization of tubulin. The antitumor action and quantitative tumor perfusion studies were carried out with locally advanced murine CT26 colon carcinoma grown in female BALB/c mice. A single injection of ATO intraperitoneally displayed central necrosis of the tumor tissue by 24 hours. T1-weighted dynamic contrast-enhanced magnetic resonance image revealed a significant decrease in tumor enhancement in the ATO-treated group. Similar to other VDAs, ATO treatment alone did not delay the progression of tumor growth; however, ATO treatment after injection of other cytotoxic agent (irinotecan) showed significant additive antitumor effect compared to control and irinotecan alone therapy. In summary, our data demonstrated that ATO acts as a VDA by means of microtubule depolymerization. It exhibits significant vascular shutdown activity in CT26 allograft model and enhances antitumor activity when used in combination with another cytotoxic chemotherapeutic agent.
Collapse
|
14
|
Alhasan MK, Liu L, Lewis MA, Magnusson J, Mason RP. Comparison of optical and power Doppler ultrasound imaging for non-invasive evaluation of arsenic trioxide as a vascular disrupting agent in tumors. PLoS One 2012; 7:e46106. [PMID: 23029403 PMCID: PMC3460997 DOI: 10.1371/journal.pone.0046106] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 08/28/2012] [Indexed: 01/13/2023] Open
Abstract
Small animal imaging provides diverse methods for evaluating tumor growth and acute response to therapy. This study compared the utility of non-invasive optical and ultrasound imaging to monitor growth of three diverse human tumor xenografts (brain U87-luc-mCherry, mammary MCF7-luc-mCherry, and prostate PC3-luc) growing in nude mice. Bioluminescence imaging (BLI), fluorescence imaging (FLI), and Power Doppler ultrasound (PD US) were then applied to examine acute vascular disruption following administration of arsenic trioxide (ATO). During initial tumor growth, strong correlations were found between manual caliper measured tumor volume and FLI intensity, BLI intensity following luciferin injection, and traditional B-mode US. Administration of ATO to established U87 tumors caused significant vascular shutdown within 2 hrs at all doses in the range 5 to 10 mg/kg in a dose dependant manner, as revealed by depressed bioluminescent light emission. At lower doses substantial recovery was seen within 4 hrs. At 8 mg/kg there was >85% reduction in tumor vascular perfusion, which remained depressed after 6 hrs, but showed some recovery after 24 hrs. Similar response was observed in MCF7 and PC3 tumors. Dynamic BLI and PD US each showed similar duration and percent reductions in tumor blood flow, but FLI showed no significant changes during the first 24 hrs. The results provide further evidence for comparable utility of optical and ultrasound imaging for monitoring tumor growth, More specifically, they confirm the utility of BLI and ultrasound imaging as facile assays of the vascular disruption in solid tumors based on ATO as a model agent.
Collapse
Affiliation(s)
| | | | | | - Jennifer Magnusson
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
15
|
Tian J, Zhao H, Nolley R, Reese SW, Young SR, Li X, Peehl DM, Knox SJ. Darinaparsin: solid tumor hypoxic cytotoxin and radiosensitizer. Clin Cancer Res 2012; 18:3366-76. [PMID: 22535156 DOI: 10.1158/1078-0432.ccr-11-3179] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Hypoxia is an important characteristic of the solid tumor microenvironment and constitutes a barrier for effective radiotherapy. Here, we studied the effects of darinaparsin (an arsenic cytotoxin) on survival and radiosensitivity of tumor cells in vitro under normoxia and hypoxia and in vivo using xenograft models, compared to effects on normal tissues. EXPERIMENTAL DESIGN The cytotoxicity and radiosensitization of darinaparsin were first tested in vitro in a variety of solid tumor cell lines under both normoxia and hypoxia and compared with arsenic trioxide (ATO, an arsenical with reported cytotoxic and radiosensitizing activities on tumor cells). The effects were then tested in mouse models of xenograft tumors derived from tumor cell lines and clinical tumor specimens. The potential mechanisms of darinaparsin effects, including reactive oxygen species (ROS) generation, cellular damage, and changes in global gene expression, were also investigated. RESULTS In comparison with ATO, darinaparsin had significantly higher in vitro cytotoxic and radiosensitizing activities against solid tumor cells under both normoxia and hypoxia. In vivo experiments confirmed these activities at doses that had no systemic toxicities. Importantly, darinaparsin did not radiosensitize normal bone marrow and actually radioprotected normal intestinal crypts. The darinaparsin-mediated antitumor effects under hypoxia were not dependent on ROS generation and oxidative damage, but were associated with inhibition of oncogene (RAS and MYC)-dependent gene expression. CONCLUSION Darinaparsin has significant and preferential cytotoxic and radiosensitizing effects on solid tumors as compared with normal cells. Darinaparsin may therefore increase the therapeutic index of radiation therapy and has near term translational potential.
Collapse
Affiliation(s)
- Junqiang Tian
- Department of Radiation Oncology and Urology, School of Medicine, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Su Y, Wang X, Xu W, Xue L, He C, Yang D, An R. Arsenic Trioxide Increases the Sensitivity of 786–0 Renal Carcinoma Cells to Radiotherapy. Cancer Invest 2012; 30:114-8. [DOI: 10.3109/07357907.2011.640652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
17
|
Vascular disrupting agent arsenic trioxide enhances thermoradiotherapy of solid tumors. JOURNAL OF ONCOLOGY 2012; 2012:934918. [PMID: 22272199 PMCID: PMC3261488 DOI: 10.1155/2012/934918] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 08/23/2011] [Accepted: 09/06/2011] [Indexed: 11/17/2022]
Abstract
Our previous studies demonstrated arsenic trioxide- (ATO-) induced selective tumor vascular disruption and augmentation of thermal or radiotherapy effect against solid tumors. These results suggested that a trimodality approach of radiation, ATO, and local hyperthermia may have potent therapeutic efficacy against solid tumors. Here, we report the antitumor effect of hypofractionated radiation followed by ATO administration and local 42.5 °C hyperthermia and the effects of cisplatin and thermoradiotherapy. We found that the therapeutic efficacy of ATO-based thermoradiotherapy was equal or greater than that of cisplatin-based thermoradiotherapy, and marked evidence of in vivo apoptosis and tumor necrosis were observed in ATO-treated tumors. We conclude that ATO-based thermoradiotherapy is a powerful means to control tumor growth by using vascular disruption to augment the effects of thermal and radiation therapy.
Collapse
|
18
|
Liu SK, Bham SAS, Fokas E, Beech J, Im J, Cho S, Harris AL, Muschel RJ. Delta-like ligand 4-notch blockade and tumor radiation response. J Natl Cancer Inst 2011; 103:1778-98. [PMID: 22010178 DOI: 10.1093/jnci/djr419] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The microenvironment plays an important role in regulating tumor response to radiotherapy. Ionizing radiation can disrupt tumor vasculature, and Notch pathway inhibition can interfere with functional angiogenesis. We explored the potential cooperativity between Notch inhibition and ionizing radiation in delaying tumor growth. METHODS Human colorectal carcinoma LS174T cells, which express the Notch ligand delta-like ligand 4 (DLL4), and human head and neck cancer FaDu cells, which do not, were grown as subcutaneous xenografts in nude mice. The mice were treated with dibenzazepine (DBZ), a γ-secretase inhibitor that blocks all Notch signaling, or a DLL4-specific blocking monoclonal antibody, alone or in combination with ionizing radiation (n = 5-10 mice per group), and response was assessed by tumor growth delay. Microbubble contrast Doppler ultrasound was used to measure tumor blood flow. Tumor Notch activity was monitored by in vivo bioluminescence from a Notch luciferase reporter. Vessel density was assessed using Chalkley vessel counting. All statistical tests were two-sided. RESULTS In LS174T xenografts, the average time for tumor volumes to reach four times the starting volume was longer for mice treated with the DLL4 monoclonal antibody than for mice treated with DBZ (16.4 vs 9.5 days, difference = 6.9 days, 95% confidence interval [CI] = 3.7 to 10.1 days, P < .001). Both Notch inhibitors suppressed tumor Notch activity within 24 hours of administration compared with vehicle (change in luciferase activity, vehicle vs DBZ: 103% vs 28%, difference = 75%, 95% CI = 39% to 109%, P = .002; vehicle vs DLL4 antibody: 172% vs 26%, difference = 146%, 95% CI = 86% to 205%, P < .001). Administration of the DLL4 antibody or DBZ after ionizing radiation resulted in a supra-additive growth delay compared with vehicle (vehicle vs DLL4 antibody + ionizing radiation: 6.8 vs 44.3 days, difference = 37.5 days, 95% CI = 32 to 43 days, P < .001; vehicle vs DBZ + ionizing radiation: 7.1 vs 24.4 days, difference = 17.3 days, 95% CI = 15.9 to 18.6 days, P < .001). Treatment of mice with the DLL4 antibody alone or in combination with ionizing radiation increased tumor vessel density but reduced tumor blood flow. Combination therapy with DLL4 antibody and ionizing radiation resulted in extensive tumor necrosis in LS174T xenografts and enhanced tumor growth delay in FaDu xenografts. CONCLUSION The combination of specific DLL4-Notch blockade and ionizing radiation impairs tumor growth by promoting nonfunctional tumor angiogenesis and extensive tumor necrosis, independent of tumor DLL4 expression.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Adenocarcinoma/blood supply
- Adenocarcinoma/metabolism
- Adenocarcinoma/radiotherapy
- Amyloid Precursor Protein Secretases/antagonists & inhibitors
- Animals
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/pharmacology
- Blood Flow Velocity
- Calcium-Binding Proteins
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/radiotherapy
- Cell Line, Tumor
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/radiotherapy
- Combined Modality Therapy
- Dibenzazepines/pharmacology
- Humans
- Hypopharyngeal Neoplasms/metabolism
- Hypopharyngeal Neoplasms/radiotherapy
- Immunoblotting
- Immunohistochemistry
- Intercellular Signaling Peptides and Proteins/immunology
- Luminescent Measurements
- Mice
- Mice, Nude
- Necrosis
- Neoplasms/blood supply
- Neoplasms/metabolism
- Neoplasms/radiotherapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/prevention & control
- Neovascularization, Pathologic/radiotherapy
- Radiation, Ionizing
- Receptors, Notch/antagonists & inhibitors
- Signal Transduction
- Transplantation, Heterologous
- Ultrasonography, Doppler
Collapse
Affiliation(s)
- Stanley K Liu
- DPhil, Molecular Oncology Unit, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Diepart C, Karroum O, Magat J, Feron O, Verrax J, Calderon PB, Grégoire V, Leveque P, Stockis J, Dauguet N, Jordan BF, Gallez B. Arsenic trioxide treatment decreases the oxygen consumption rate of tumor cells and radiosensitizes solid tumors. Cancer Res 2011; 72:482-90. [PMID: 22139377 DOI: 10.1158/0008-5472.can-11-1755] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Arsenic trioxide (As(2)O(3)) is an effective therapeutic against acute promyelocytic leukemia and certain solid tumors. Because As(2)O(3) inhibits mitochondrial respiration in leukemia cells, we hypothesized that As(2)O(3) might enhance the radiosensitivity of solid tumors by increasing tumor oxygenation [partial pressure of oxygen (pO(2))] via a decrease in oxygen consumption. Two murine models of radioresistant hypoxic cancer were used to study the effects of As(2)O(3). We measured pO(2) and the oxygen consumption rate in vivo by electron paramagnetic resonance oximetry and (19)fluorine-MRI relaxometry. Tumor perfusion was assessed by Patent blue staining. In both models, As(2)O(3) inhibited mitochondrial respiration, leading to a rapid increase in pO(2). The decrease in oxygen consumption could be explained by an observed decrease in glutathione in As(2)O(3)-treated cells, as this could increase intracellular reactive oxygen species that can disrupt mitochondrial membrane potential. When tumors were irradiated during periods of As(2)O(3)-induced augmented oxygenation, radiosensitivity increased by 2.2-fold compared with control mice. Notably, this effect was abolished when temporarily clamped tumors were irradiated. Together, our findings show that As(2)O(3) acutely increases oxygen consumption and radiosensitizes tumors, providing a new rationale for clinical investigations of As(2)O(3) in irradiation protocols to treat solid tumors.
Collapse
Affiliation(s)
- Caroline Diepart
- Biomedical Magnetic Resonance Group, Louvain Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhao W, Lu X, Yuan Y, Liu C, Yang B, Hong H, Wang G, Zeng F. Effect of size and processing method on the cytotoxicity of realgar nanoparticles in cancer cell lines. Int J Nanomedicine 2011; 6:1569-77. [PMID: 21845047 PMCID: PMC3152475 DOI: 10.2147/ijn.s21373] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this study, the effects of the size and Chinese traditional processing (including elutriation, water cleaning, acid cleaning, alkali cleaning) on realgar nanoparticles (RN)-induced antitumor activity in human osteosarcoma cell lines (MG-63) and hepatoma carcinoma cell lines (HepG-2) were investigated. The human normal liver cell line (L-02) was used as control. RN was prepared by high-energy ball milling technology. The results showed that with the assistance of sodium dodecyl sulfate, the size of realgar could be reduced to 127 nm after 12 hours’ ball milling. The surface charge was decreased from 0.83 eV to −17.85 eV and the content of As2O3 clearly increased. Except for elutriation, the processing methods did not clearly change the size of the RN, but the content of As2O3 was reduced dramatically. In vitro MTT tests indicated that in the two cancer cell lines, RN cytotoxicity was more intense than that of the coarse realgar nanoparticles, and cytotoxicity was typically time- and concentration-dependent. Also, RN cytotoxicities in the HepG-2 and L-02 cells all increased with increasing milling time. Due to the reduction of the As2O3 content, water cleaning, acid cleaning, and alkali cleaning decreased RN cytotoxicity in HepG-2, but RN after elutriation, with the lowest As2O3 (3.5 mg/g) and the smallest size (109.3 nm), showed comparable cytotoxicity in HepG-2 to RN without treatment. Meanwhile, RN-induced cytotoxicity in L-02 cells was clearly reduced. Therefore, it can be concluded that RN may provide a strong antiproliferation effect in the MG-63 and HepG-2 cells. Elutriation processing is a suitable approach to limit the dangerous side-effects of As2O3, while maintaining the effectiveness of RN.
Collapse
Affiliation(s)
- Weizhong Zhao
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhao D, Jiang Y, Dong X, Liu Z, Qu B, Zhang Y, Ma N, Han Q. Arsenic trioxide reduces drug resistance to adriamycin in leukemic K562/A02 cells via multiple mechanisms. Biomed Pharmacother 2011; 65:354-8. [PMID: 21775093 DOI: 10.1016/j.biopha.2011.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Accepted: 04/07/2011] [Indexed: 11/30/2022] Open
Abstract
The present study aimed to study the mechanisms by which low dose arsenic trioxide (As(2)O(3)) reduces multidrug resistance. The potential influence of As(2)O(3) on cytotoxicity was examined by methyl thiazolyl tetrazolium (MTT) assay and the intracellular mean fluorescence intensity (MFI) of Adriamycin (ADM) was examined by flow cytometry. The gene expression of mdr1 mRNA was determined by RT-PCR. The change of cellular expression levels of drug resistant-related proteins, including P-gp, bcl-2, Topo-II, and GST-π, were measured by Western-blotting or immunocytochemistry assay. Data showed As(2)O(3) at non-cytotoxic concentration (2μM) significantly increased the cytotoxicity of ADM on K562/A02 cells. Cotreatment of As(2)O(3) and ADM significantly increased the ADM MFI than ADM alone (P<0.01). Following pretreatment of K562/A02 cells with As(2)O(3), the expression of Topo-II was increased while the expression of GST-π and bcl-2 was decreased. No obvious alternation of expression of mdr1 mRNA or P-gp was observed. Thus, low dose As(2)O(3) partially reduced drug resistance to ADM in K562/A02 cells via multiple mechanisms, which selectively inhibited the efflux pump GST-π but not P-gp, as well as modulated the expression of MDR-related proteins such as Topo-II and bcl-2, in line with previous studies. In conclusions: The effect of As(2)O(3) on reducing MDR may have wide clinical application in chemotherapy regimens for leukemia.
Collapse
Affiliation(s)
- Dianfeng Zhao
- Department of Hematologic Neoplasms, the First Affiliated Hospital, Jilin University, 71 Xinmin Avenue, Changchun, 130021, China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Yeh KY, Chang JWC, Li YY, Wang CH, Wang HM. Tumor growth inhibition of metastatic nasopharyngeal carcinoma cell lines by low dose of arsenic trioxide via alteration of cell cycle progression and induction of apoptosis. Head Neck 2010; 33:734-42. [PMID: 20737493 DOI: 10.1002/hed.21535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2010] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Although arsenic trioxide (ATO) has displayed anticancer activity against primary nasopharyngeal carcinoma (NPC), its efficacy in metastatic NPC deserved further investigation because the biological/therapeutic difference in cancer cells probably exists between primary and distant sites. METHODS Two human metastatic NPC cell lines (NPC-BM1 and NPC-BM2) were investigated. We measured cellular proliferation, cell cycle, and apoptotic extent of BM1 and BM2 cells treated with ATO in vitro. Furthermore, we evaluated the tumor growth after ATO treatment in vivo. RESULTS Low-dose ATO treatment is sufficient to induce an antiproliferative effect, alter the cell cycle, and increase apoptosis in BM1 and BM2 cells. BM1 tumor growth in a xenograft model with low-dose and short-schedule (1 mg/kg/day, intraperitoneal injection for 5 consecutive days) of ATO treatment significantly slowed in vivo. CONCLUSION ATO at low dose seems to be an encouraging schedule for palliative treatment of metastatic NPC.
Collapse
Affiliation(s)
- Kun-Yun Yeh
- Division of Hemato-oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung & Chang Gung University, College of Medicine, Taiwan, Providence of China.
| | | | | | | | | |
Collapse
|
23
|
Preparation, characterization, in vivo and in vitro studies of arsenic trioxide Mg-Fe ferrite magnetic nanoparticles. Acta Pharmacol Sin 2009; 30:1688-93. [PMID: 19960013 DOI: 10.1038/aps.2009.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
AIM MgFe(2)O(4) magnetic nanoparticle composed of As(2)O(3) (As(2)O(3)-MNPs) were prepared and their in vitro and in vivo characteristics were studied. METHODS The solvent-displacement method was applied for preparation of the nanoparticle using Poly-D,L-lactic-co-glycolic acid(PLGA). The characteristics studies of the products included magnetic response, morphology (transmission electron microscopy and scanning electron microscopy), entrapment efficiency, drug loading, particle sizes, zeta potential, in vitro drug release and tissue magnetic targeting. Nanoparticle cytotoxicity to Saos-2 cells was investigated using the MTT assay. To guide the external magnetic field in the liver, the concentration of As(2)O(3) in the liver and kidney was measured using an atomic fluorescence spectrometer after injecting As(2)O(3)-MNPs into the caudal veins of mice. RESULTS The As(2)O(3)-MNPs were approximately spherical. The average diameter, drug loading, entrapment efficiency and zeta potential of As(2)O(3)-MNPs were 109.9 nm, 10.08%, 82.16%, and -14.33 mV, respectively. The specific saturation magnetism was 8.65 emu/g. In vivo, the concentration of As(2)O(3) in the liver was significantly higher than that in the non-magnetic group. While the concentration of As(2)O(3) in the kidney was lower than that in the non-magnetic group. The C(max) in liver tissue in the magnetic group was 30.65 microg/g, which was 4.17 times the drug concentration in the same group in kidney tissue (7.35 microg/g) and 2.88 times the concentration of drug (10.66 microg/g) in the liver tissue of the non-magnetic group. CONCLUSION The PLGA polymer-loaded magnetic nanoparticle composed of arsenic trioxide can be magnetically targeted well and applied in biomedicine.
Collapse
|
24
|
Park SG, Jung JJ, Won HJ, Kang MS, Seo SK, Choi IW, Eun CK, Ahn KJ, Park CW, Lee SW, Lew YS, Bae IJ, Choi IH. Tetra-arsenic oxide (Tetras) enhances radiation sensitivity of solid tumors by anti-vascular effect. Cancer Lett 2009; 277:212-7. [DOI: 10.1016/j.canlet.2008.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 12/05/2008] [Accepted: 12/08/2008] [Indexed: 11/28/2022]
|
25
|
Fröhlich E, Czarnocka B, Brossart P, Wahl R. Antitumor effects of arsenic trioxide in transformed human thyroid cells. Thyroid 2008; 18:1183-93. [PMID: 19014326 DOI: 10.1089/thy.2008.0114] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND To improve radioiodine treatment of metastasized differentiated thyroid carcinomas, substances that increase iodide uptake are needed. Many tumors are not responsive to retinoic acid as a differentiating agent. Therefore, identification of other differentiating substances is needed. Arsenic trioxide (ATO) was investigated for its potential to increase iodide uptake. METHODS The action of ATO on proliferation, differentiation, and apoptosis was evaluated in follicular and papillary thyroid carcinoma cell lines. To get insight into the mode of action of ATO, coincubations with inhibitors of the phosphoinositide 3 (PI3) kinase pathway (V-Akt Murine Thymoma Viral Oncogene Homolog 1, Akt inhibitors) were performed; glutathione (GSH) levels were determined, as well as synergistic effects of ATO with inhibitors of GSH metabolism, inductors of oxidative stress. As a potential additional target of the pleiotropic action of ATO, its effect on glucose uptake was investigated. The expression of sodium iodide symporter, pendrin, phospho-Akt, and glucose transporter 1 was studied to reveal a potential effect of ATO on the transcription of specific genes. RESULTS ATO reduced proliferation, increased iodide uptake and apoptosis, and, as an additional new mechanism, decreased glucose uptake in transformed thyrocytes. The pharmacological reduction of the amount of reduced GSH was effective in enhancing the differentiating action of ATO, whereas the combination of ATO with Akt-1 inhibitors reduced cell number but did not increase differentiation. CONCLUSIONS Our study suggests a new therapeutic option for postoperative treatment of radioiodine nonresponsive differentiated thyroid carcinomas.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/drug therapy
- Adenocarcinoma, Follicular/metabolism
- Adenocarcinoma, Follicular/pathology
- Adenocarcinoma, Follicular/radiotherapy
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Arsenic Trioxide
- Arsenicals/pharmacology
- Biological Transport, Active/drug effects
- Carcinoma, Papillary/drug therapy
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/pathology
- Carcinoma, Papillary/radiotherapy
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/metabolism
- Glucose/metabolism
- Glutathione/metabolism
- Humans
- Iodides/metabolism
- Iodine Radioisotopes/therapeutic use
- Oncogene Protein v-akt/antagonists & inhibitors
- Oxides/pharmacology
- Phosphatidylinositol 3-Kinases/metabolism
- Radiation Tolerance
- Reactive Oxygen Species/metabolism
- Thyroid Neoplasms/drug therapy
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/radiotherapy
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Department of Endocrinology, Metabolism, Nephrology, and Clinical Chemistry, University of Tuebingen, Tuebingen, Germany
| | | | | | | |
Collapse
|
26
|
Kumar P, Gao Q, Ning Y, Wang Z, Krebsbach PH, Polverini PJ. Arsenic trioxide enhances the therapeutic efficacy of radiation treatment of oral squamous carcinoma while protecting bone. Mol Cancer Ther 2008; 7:2060-9. [PMID: 18645016 DOI: 10.1158/1535-7163.mct-08-0287] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Therapeutic radiation is commonly used in the treatment of squamous cell carcinoma of the oral cavity and pharynx. Despite the proven efficacy of this form of anticancer therapy, high-dose radiation treatment is invariably associated with numerous unwanted side effects. This is particularly true for bone, in which radiation treatment often leads to osteoradionecrosis. The aim of this study was to investigate if treatment with arsenic trioxide (As(2)O(3)) could enhance the antitumor effect of radiotherapy whereas minimizing the destructive effects of radiation on bone. As(2)O(3) treatment induced a dose-dependent (1-20 mumol/L) inhibition of endothelial and tumor cell (OSCC-3 and UM-SCC-74A) survival and significantly enhanced radiation-induced endothelial cell and tumor cell death. In contrast, As(2)O(3) treatment (0.5-7.5 mumol/L) induced the proliferation of osteoblasts and also protected osteoblasts against radiation-induced cell death. Furthermore, As(2)O(3) treatment was able to significantly enhance radiation-induced inhibition of endothelial cell tube formation and tumor cell colony formation. To test the effectiveness of As(2)O(3) and radiation treatment in vivo, we used a severe combined immunodeficiency mouse model that has a bone ossicle and tumor growing side by side subcutaneously. Animals treated with As(2)O(3) and radiation showed a significant inhibition of tumor growth, tumor angiogenesis, and tumor metastasis to the lungs as compared with As(2)O(3) treatment or radiation treatment alone. In contrast, As(2)O(3) treatment protected bone ossicles from radiation-induced bone loss. These results suggest a novel strategy to enhance the therapeutic efficacy of radiation treatment while protecting bone from the adverse effects of therapeutic radiation.
Collapse
Affiliation(s)
- Pawan Kumar
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 North University Avenue, Room no. 5205, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Florea AM, Splettstoesser F, Büsselberg D. Arsenic trioxide (As2O3) induced calcium signals and cytotoxicity in two human cell lines: SY-5Y neuroblastoma and 293 embryonic kidney (HEK). Toxicol Appl Pharmacol 2007; 220:292-301. [PMID: 17376498 DOI: 10.1016/j.taap.2007.01.022] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Revised: 01/08/2007] [Accepted: 01/29/2007] [Indexed: 10/23/2022]
Abstract
Arsenic trioxide (As(2)O(3)) has anticancer properties; however, its use also leads to neuro-, hepato- or nephro-toxicity, and therefore, it is important to understand the mechanism of As(2)O(3) toxicity. We studied As(2)O(3) influence on intracellular calcium ([Ca(2+)](i)) homeostasis of human neuroblastoma SY-5Y and embryonic kidney cells (HEK 293). We also relate the As(2)O(3) induced [Ca(2+)](i) modifications with cytotoxicity. We used Ca(2+) sensitive dyes (fluo-4 and rhod-2) combined with laser scanning microscopy or fluorescence activated cell sorting to measure Ca(2+) changes during the application of As(2)O(3) and we approach evaluation of cytotoxicity. As(2)O(3) (1 microM) increased [Ca(2+)](i) in SY-5Y and HEK 293 cells. Three forms of [Ca(2+)](i)-elevations were found: (1) steady-state increases, (2) transient [Ca(2+)](i)-elevations and (3) Ca(2+)-spikes. [Ca(2+)](i) modifications were independent from extracellular Ca(2+) but dependent on internal calcium stores. The effect was not reversible. Inositol triphosphate (IP(3)) and ryanodine (Ry) receptors are involved in regulation of signals induced by As(2)O(3). 2-APB and dantrolene significantly reduced the [Ca(2+)](i)-rise (p<0.001, t-test) but did not completely abolish [Ca(2+)](i)-elevation or spiking. This indicates that other Ca(2+) regulating mechanisms are involved. In cytotoxicity tests As(2)O(3) significantly reduced cell viability in both cell types. Staining with Hoechst 33342 showed occurrence of apoptosis and DNA damage. Our data suggest that [Ca(2+)](i) is an important messenger in As(2)O(3) induced cell death.
Collapse
Affiliation(s)
- Ana-Maria Florea
- Institut für Physiologie, Universitätsklinikum, Universität Duisburg Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | |
Collapse
|
28
|
Kai T, Kimura H, Shiga Y, Ogawa K, Sato H, Maruyama Y. Recurrent extramedullary relapse of acute promyelocytic leukemia after allogeneic stem cell transplantation: successful treatment by arsenic trioxide in combination with local radiotherapy. Int J Hematol 2006; 83:337-40. [PMID: 16757435 DOI: 10.1532/ijh97.05167] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Isolated extramedullary relapse is rare in patients with acute promyelocytic leukemia (APL) after allogeneic stem cell transplantation (SCT), and an optimal therapy for it has not been established. We describe a patient with APL who developed serially occurring extramedullary disease (EMD) after SCT. We confirmed that EMD had arisen from the recipient's APL blasts by detecting t(15;17) and PML/RARalpha from the tumor cell suspension. The patient displayed EMD 4 times at different sites. Administration of all-trans retinoic acid with local radiotherapy and with chemotherapy for the first to third EMDs resulted in regression of the tumors. However, these regimens did not prevent the subsequent occurrence of new EMD. For the fourth EMD, intravenous administration of arsenic trioxide followed by local radiotherapy resulted in the disappearance of EMD, and no further EMD has developed to date. In the present case, the bone marrow was in morphologic and molecular remission during the course of recurrent EMD. The accumulation of detailed cases is needed to elucidate the pathogenesis, predisposing factors, and optimal therapy for EMD in APL after SCT.
Collapse
MESH Headings
- Antineoplastic Agents/administration & dosage
- Arsenic Trioxide
- Arsenicals/administration & dosage
- Chromosomes, Human, Pair 15/genetics
- Chromosomes, Human, Pair 17/genetics
- Combined Modality Therapy
- Female
- Humans
- Leukemia, Promyelocytic, Acute/diagnostic imaging
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/therapy
- Middle Aged
- Neoplasm Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Oxides/administration & dosage
- Radiography
- Recurrence
- Remission Induction
- Sarcoma, Myeloid/diagnostic imaging
- Sarcoma, Myeloid/genetics
- Sarcoma, Myeloid/therapy
- Stem Cell Transplantation/adverse effects
- Translocation, Genetic/genetics
- Transplantation, Homologous
- Tretinoin/administration & dosage
Collapse
Affiliation(s)
- Tatsuyuki Kai
- Hematology, Kita-Fukushima Medical Center, Fukushima, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Ning S, Knox SJ. Optimization of combination therapy of arsenic trioxide and fractionated radiotherapy for malignant glioma. Int J Radiat Oncol Biol Phys 2006; 65:493-8. [PMID: 16563655 DOI: 10.1016/j.ijrobp.2005.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Revised: 12/07/2005] [Accepted: 12/09/2005] [Indexed: 11/17/2022]
Abstract
PURPOSE The primary objective was to optimize the combined treatment regimen using arsenic trioxide (ATO) and fractionated radiotherapy for the treatment of malignant glioma. METHODS AND MATERIALS Nude mice with human glioma xenograft tumors were treated with fractionated local tumor radiation of 250 cGy/fraction/day and 5 mg/kg ATO for 5-10 days. RESULTS Time course experiments demonstrated that maximal tumor growth delay occurred when ATO was administered between 0 and 4 h after radiation. The combination treatment of ATO and radiation synergistically inhibited tumor growth and produced a tumor growth delay time of 13.2 days, compared with 1.4 days and 6.5 days for ATO and radiation alone (p < 0.01), respectively. The use of concurrent therapy of radiation and ATO initially, followed by ATO as maintenance therapy, was superior to the use of preloading with ATO before combined therapy and produced a tumor growth delay time of 22.7 days as compared with 11.7 days for the ATO preloading regimen (p < 0.01). The maintenance dose of ATO after concurrent therapy was effective and important for continued inhibition of tumor growth. CONCLUSIONS The combined use of fractionated radiation and ATO is effective for the treatment of glioma xenograft tumors. ATO was most effective when administered 0-4 h after radiation without pretreatment with ATO. These results have important implications for the optimization of treatment regimen using ATO and fractionated radiotherapy for the treatment of brain tumors.
Collapse
Affiliation(s)
- Shoucheng Ning
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, CA 94305-1245, USA
| | | |
Collapse
|
30
|
Stanton CR, Thibodeau R, Lankowski A, Shaw JR, Hamilton JW, Stanton BA. Arsenic Inhibits CFTR-Mediated Chloride Secretion by Killifish (Fundulus heteroclitus) Opercular Membrane. Cell Physiol Biochem 2006; 17:269-78. [PMID: 16791002 DOI: 10.1159/000094139] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Killifish are euryhaline teleosts that normally experience rapid changes in the salinity of the swim water. Acclimation to seawater is mediated by cortisol, which by activating glucocorticoid receptors, upregulates CFTR mediated Cl- secretion in the gill and operculum. Arsenic, a toxic metalloid that naturally occurs in the aquatic environment, has been shown to disrupt glucocorticoid hormone-mediated regulation of genes. Because little is known about the effects of environmentally relevant levels of arsenic on ion channels and salt homeostasis, studies were conducted to examine the effects of arsenic on the ability of killifish to acclimate to increased salinity. Arsenic in the swim water or administered by intraperitoneal injection prevented acclimation. To determine if arsenic blocked acclimation by inhibiting CFTR mediated Cl- secretion (Isc), opercular membranes were isolated and mounted in Ussing chambers and the effects of arsenic on Isc were measured. Arsenic (24 hr exposure) reduced Isc in opercular membranes isolated from salt water acclimated killifish. In addition, arsenic acutely (5-10 minutes) and reversibly inhibited Isc with an IC50 = 4.1 microM (305 ppb) when applied to the apical (seawater) side of the operculum, but not when added to the basolateral side of the operculum. Arsenic (4 microM for 60 minutes) also reduced mitochondrial respiration. Thus, environmentally relevant levels of arsenic block acclimation to seawater in killifish by reversibly inhibiting CFTR-mediated Cl- secretion by the opercular membrane, in part by inhibiting mitochondrial respiration.
Collapse
Affiliation(s)
- Caitlin R Stanton
- Department of Physiology, Dartmouth Medical School, Hanover, Germany
| | | | | | | | | | | |
Collapse
|