1
|
Walker DM, Lazarova TI, Riesinger SW, Poirier MC, Messier T, Cunniff B, Walker VE. WR1065 conjugated to thiol-PEG polymers as novel anticancer prodrugs: broad spectrum efficacy, synergism, and drug resistance reversal. Front Oncol 2023; 13:1212604. [PMID: 37576902 PMCID: PMC10419174 DOI: 10.3389/fonc.2023.1212604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/27/2023] [Indexed: 08/15/2023] Open
Abstract
The lack of anticancer agents that overcome innate/acquired drug resistance is the single biggest barrier to achieving a durable complete response to cancer therapy. To address this issue, a new drug family was developed for intracellular delivery of the bioactive aminothiol WR1065 by conjugating it to discrete thiol-PEG polymers: 4-star-PEG-S-S-WR1065 (4SP65) delivers four WR1065s/molecule and m-PEG6-S-S-WR1065 (1LP65) delivers one. Infrequently, WR1065 has exhibited anticancer effects when delivered via the FDA-approved cytoprotectant amifostine, which provides one WR1065/molecule extracellularly. The relative anticancer effectiveness of 4SP65, 1LP65, and amifostine was evaluated in a panel of 15 human cancer cell lines derived from seven tissues. Additional experiments assessed the capacity of 4SP65 co-treatments to potentiate the anticancer effectiveness and overcome drug resistance to cisplatin, a chemotherapeutic, or gefitinib, a tyrosine kinase inhibitor (TKI) targeting oncogenic EGFR mutations. The CyQUANT®-NF proliferation assay was used to assess cell viability after 48-h drug treatments, with the National Cancer Institute COMPARE methodology employed to characterize dose-response metrics. In normal human epithelial cells, 4SP65 or 1LP65 enhanced or inhibited cell growth but was not cytotoxic. In cancer cell lines, 4SP65 and 1LP65 induced dose-dependent cytostasis and cytolysis achieving 99% cell death at drug concentrations of 11.2 ± 1.2 µM and 126 ± 15.8 µM, respectively. Amifostine had limited cytostatic effects in 11/14 cancer cell lines and no cytolytic effects. Binary pairs of 4SP65 plus cisplatin or gefitinib increased the efficacy of each partner drug and surmounted resistance to cytolysis by cisplatin and gefitinib in relevant cancer cell lines. 4SP65 and 1LP65 were significantly more effective against TP53-mutant than TP53-wild-type cell lines, consistent with WR1065-mediated reactivation of mutant p53. Thus, 4SP65 and 1LP65 represent a unique prodrug family for innovative applications as broad-spectrum anticancer agents that target p53 and synergize with a chemotherapeutic and an EGFR-TKI to prevent or overcome drug resistance.
Collapse
Affiliation(s)
- Dale M. Walker
- The Burlington HC Research Group, Inc., Jericho, VT, United States
| | | | | | - Miriam C. Poirier
- Carcinogen–DNA Interactions Section, Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Terri Messier
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Vernon E. Walker
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
2
|
Choy J, Swe EPP, Shearer A. Amifostine and rituximab in refractory immune thrombocytopaenia: A case series. Vox Sang 2021; 117:601-605. [PMID: 34939200 DOI: 10.1111/vox.13226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/02/2021] [Accepted: 11/09/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND OBJECTIVES Management of refractory immune thrombocytopaenia (ITP) can be challenging. Amifostine, a thiophosphate prodrug, induces megakaryocyte maturation. In 2010, Fan et al. published results for 21 Chinese splenectomized patients, aged 13-92, with steroid-refractory ITP. Nineteen patients (15 patients aged >18 years) achieved remission 2 months post-amifostine. This is the first publication utilizing amifostine and rituximab in refractory ITP. MATERIALS AND METHODS At the Cairns Hospital in Australia, we identified five patients treated with amifostine and rituximab for refractory ITP. Amifostine IV 400 mg once daily was administered 5 days/week for 5 weeks as tolerated. Rituximab was administered concurrently with/preceding amifostine based on clinician preference. Data were obtained through medical records and follow-up serology up to 5 years post-amifostine was available. RESULTS Three cases demonstrated durable responses up to 5 years' follow-up. One patient initially achieved remission but relapsed 1-year post-amifostine. One patient who did not have a splenectomy prior to amifostine did not respond. CONCLUSION Three out of five patients achieved durable responses with amifostine and rituximab. Although there is confounding by rituximab, given its established low durable response rate, it is likely that the excellent long-term responses achieved were a result of amifostine. Clinical trials with larger patient cohorts and further investigation are required to confirm the efficacy and mechanism of amifostine in ITP.
Collapse
Affiliation(s)
- Joleen Choy
- Department of Haematology, Queensland Health, Cairns Hospital, Cairns North, Queensland, Australia
| | - Ei Phyu Phyu Swe
- Department of Medicine, Queensland Health, Cairns Hospital, Cairns North, Queensland, Australia
| | - Andrew Shearer
- Department of Haematology, Queensland Health, Cairns Hospital, Cairns North, Queensland, Australia
| |
Collapse
|
3
|
King M, Joseph S, Albert A, Thomas TV, Nittala MR, Woods WC, Vijayakumar S, Packianathan S. Use of Amifostine for Cytoprotection during Radiation Therapy: A Review. Oncology 2019; 98:61-80. [PMID: 31846959 DOI: 10.1159/000502979] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 08/19/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Radiation therapy is a cornerstone of the therapeutic modalities used in modern oncology. However, it is sometimes limited in its ability to achieve optimal tumor control by radiation-induced normal tissue toxicity. In delivering radiation therapy, a balance must be achieved between maximizing the dose to the tumor and minimizing any injury to the normal tissues. Amifostine was the first Food and Drug Administration (FDA)-approved clinical radiation protector intended to reduce the impact of radiation on normal tissue, lessening its toxicity and potentially allowing for increased tumor dose/control. Despite being FDA-approved almost 20 years ago, Amifostine has yet to achieve widespread clinical use. SUMMARY A thorough review of Amifostine's development, mechanism of action, and current clinical status were conducted. A brief history of Amifostine is given, from its development at Walter Reid Institute of Research to its approval for clinical use. The mechanism of action of Amifostine is explored. The results of a complete literature review of all prospective randomized trials to date involving the use of Amifostine in radiation therapy are presented. The results are arranged by treatment site and salient findings discussed. Side effects and complications to consider in using Amifostine are reviewed. Key Messages: Amifostine has been explored as a radiation protectant in most radiation treatment sites. Studies have demonstrated efficacy of Amifostine in all treatment sites reviewed, but results are heterogeneous. The heterogeneity of studies looking at Amifostine as a clinical radiation protectant has precluded a definitive answer on its efficacy. Complicating its clinical use is its toxicity and delivery requirements. Amifostine has largely fallen out of use with the advent of intensity modulated radiation therapy (IMRT). However, side effects with IMRT remain a challenge and concern. The use of Amifostine in the IMRT era has been poorly explored and is worthy of future study.
Collapse
Affiliation(s)
- Maurice King
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Sanjay Joseph
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ashley Albert
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Toms V Thomas
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Mary R Nittala
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA,
| | - William C Woods
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Srinivasan Vijayakumar
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Satyaseelan Packianathan
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
4
|
In vitro study of doxorubicin-induced oxidative stress in spermatogonia and immature Sertoli cells. Toxicol Appl Pharmacol 2018; 348:32-42. [PMID: 29660436 DOI: 10.1016/j.taap.2018.04.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 02/02/2023]
Abstract
Pediatric chemotherapy treatments can impair long-term male fertility. Unfortunately, no fertility preservation solution is available for pre-pubertal boys. Studies suggest that doxorubicin, used against pediatric cancers, induces oxidative stress in the testis. However, the targeted testicular cell types remain unknown. The goal of this study was to determine whether doxorubicin can induce oxidative stress in rat spermatogonia (GC-6Spg) and immature Sertoli (Ser-W3) cell lines, and to assess their protection by antioxidants. Using the MTT assay, we have shown that doxorubicin induces a time- and dose-dependent cytotoxicity in these two cell lines, Ser-W3 being more sensitive than GC-6Spg. After 3 h of treatment, reactive oxygen species and nuclear 8-oxo-deoxyguanosine increase in Ser-W3, but not in GC-6Spg. Moreover, after 6 h of treatment, intracellular reduced glutathione levels decrease significantly in Ser-W3 cells. These results show that doxorubicin induces oxidative stress in the Ser-W3 cell line. However, a depletion in glutathione does not affect their survival, and supplementation only offers a weak protection after exposure to doxorubicin, suggesting that the glutathione system is not essential for Ser-W3 cell line's defense against doxorubicin. On the other hand, among four antioxidants selected from the literature, none reduces the cytotoxicity of doxorubicin in Ser-W3 cells. Together, our data suggest that oxidative stress may not be a major pathway for doxorubicin's cytotoxicity in GC-6Spg and Ser-W3 lines. This study provides new insights in the mechanisms by which chemotherapies affect the pre-pubertal testis, with the long-term goal to help improve the quality of life of pediatric cancer survivors.
Collapse
|
5
|
Kumar A, Choudhary S, Adhikari JS, Chaudhury NK. Sesamol ameliorates radiation induced DNA damage in hematopoietic system of whole body γ-irradiated mice. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:79-90. [PMID: 28766757 DOI: 10.1002/em.22118] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/24/2017] [Accepted: 06/29/2017] [Indexed: 05/27/2023]
Abstract
Ionizing radiation exposure is harmful and at high doses can lead to acute hematopoietic radiation syndrome. Therefore, agents that can protect hematopoietic system are important for development of radioprotector. Sesamol is a potential molecule for development of radioprotector due to its strong free radical scavenging and antioxidant properties. In the present study, sesamol was evaluated for its role in DNA damage and repair in hematopoietic system of γ-irradiated CB57BL/6 mice and compared with amifostine. C57BL/6 male mice were administered with sesamol 20 mg/kg (i.p.) followed by 2 Gy whole body irradiation (WBI) at 30 min. Mice were sacrificed at 0.5, 3, 24 h postirradiation; bone marrow, splenocytes, and peripheral blood lymphocytes were isolated to measure DNA damages and repair using alkaline comet,γ-H2AXand micronucleus assays. An increase in % of tail DNA was observed in all organs of WBI mice. Whereas in pre-administered sesamol reduced %DNA in tail (P ≤ 0.05). Sesamol has also reduced formation of radiation induced γ-H2AX foci after 0.5 h in these organs and further lowered to respective control values at 24 h of WBI. Similar reduction of % DNA in tail and γ-H2AX foci were observed with amifostine (P ≤ 0.05). Analysis of mnPCE frequency at 24 h has revealed similar extent of protection by sesamol and amifostine. Interestingly, both sesamol and amifostine, alone and with radiation, also increased the granulocytes count significantly compared to the control (P ≤ 0.05). These findings suggest that sesamol has strong potential to protect hematopoietic system by lowering radiation induced DNA damages and can prevent acute hematopoietic syndrome in mice. Environ. Mol. Mutagen. 59:79-90, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Arun Kumar
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Marg, Timarpur, Delhi, 110054, India
| | - Sandeep Choudhary
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Marg, Timarpur, Delhi, 110054, India
| | - Jawahar S Adhikari
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Marg, Timarpur, Delhi, 110054, India
| | - Nabo K Chaudhury
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Marg, Timarpur, Delhi, 110054, India
| |
Collapse
|
6
|
Gal Y, Mazor O, Falach R, Sapoznikov A, Kronman C, Sabo T. Treatments for Pulmonary Ricin Intoxication: Current Aspects and Future Prospects. Toxins (Basel) 2017; 9:E311. [PMID: 28972558 PMCID: PMC5666358 DOI: 10.3390/toxins9100311] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Ricin, a plant-derived toxin originating from the seeds of Ricinus communis (castor beans), is one of the most lethal toxins known, particularly if inhaled. Ricin is considered a potential biological threat agent due to its high availability and ease of production. The clinical manifestation of pulmonary ricin intoxication in animal models is closely related to acute respiratory distress syndrome (ARDS), which involves pulmonary proinflammatory cytokine upregulation, massive neutrophil infiltration and severe edema. Currently, the only post-exposure measure that is effective against pulmonary ricinosis at clinically relevant time-points following intoxication in pre-clinical studies is passive immunization with anti-ricin neutralizing antibodies. The efficacy of this antitoxin treatment depends on antibody affinity and the time of treatment initiation within a limited therapeutic time window. Small-molecule compounds that interfere directly with the toxin or inhibit its intracellular trafficking may also be beneficial against ricinosis. Another approach relies on the co-administration of antitoxin antibodies with immunomodulatory drugs, thereby neutralizing the toxin while attenuating lung injury. Immunomodulators and other pharmacological-based treatment options should be tailored according to the particular pathogenesis pathways of pulmonary ricinosis. This review focuses on the current treatment options for pulmonary ricin intoxication using anti-ricin antibodies, disease-modifying countermeasures, anti-ricin small molecules and their various combinations.
Collapse
Affiliation(s)
- Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Ohad Mazor
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| |
Collapse
|
7
|
Klubo-Gwiezdzinska J, Costello J, Jensen K, Patel A, Tkavc R, Van Nostrand D, Burman KD, Wartofsky L, Vasko V. Amifostine does not protect thyroid cancer cells in DNA damaging in vitro models. Endocr Connect 2017; 6:469-478. [PMID: 28790138 PMCID: PMC5597971 DOI: 10.1530/ec-17-0138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/08/2017] [Indexed: 11/08/2022]
Abstract
BACKGROUND Amifostine is a potent scavenger of reactive oxygen species that is used for the salivary gland protection during therapy with radioactive iodine for thyroid cancer. There are no data on the potential effect of amifostine on thyroid cancer cells. METHODS We investigated the effects of the active form of amifostine (WR-1065) on the response of thyroid cancer cells to treatment with DNA-damaging agents. WR-1065 was examined in human thyroid cancer cell lines (FTC133, TPC1, BCPAP and C643) and embryonic fibroblast cells NIH3T3. DNA damage was induced by exposure to H2O2 (0.1 mM), by treatment with the radiomimetic neocarzinostatin (NCS 250 ng/mL) and by γ-radiation (6 Gy). DNA damage, cell viability and apoptosis were examined. RESULTS We demonstrated the selective action of WR-1065 (0.1 mM), which prevented oxidative stress-induced DNA damage in fibroblasts, but did not protect thyroid cancer cells from DNA damage and apoptosis documented by caspase-3 and PARP cleavage after exposure to H2O2, NCS and γ-radiation. Prolonged exposure to WR-1065 (0.1 mM for 24 h) was toxic for thyroid cancer cells; this treatment decreased the number of viable cells by 8% in C643 cells, 47% in TPC cells, 92% in BCPAP cells and 82% in FTC 133 cells. The cytotoxic effects of WR-1065 were not associated with induction of apoptosis. CONCLUSIONS Our data show that amifostine has no protective effect on thyroid cancer cells against DNA-damaging agents in vitro and suggest that amifostine will not attenuate the efficacy of radioiodine treatment in patients with thyroid cancer.
Collapse
Affiliation(s)
- Joanna Klubo-Gwiezdzinska
- National Institute of HealthNIDDK, Office 10 Center Drive, Bethesda, Maryland, USA
- Division of EndocrinologyDepartment of Medicine, Medstar Washington Hospital Center, Washington Hospital Center, Northwest, Washington, District of Columbia, USA
| | - John Costello
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Kirk Jensen
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Aneeta Patel
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Rok Tkavc
- Department of PathologyUniformed Services University of the Health Sciences, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Douglas Van Nostrand
- Division of EndocrinologyDepartment of Medicine, Medstar Washington Hospital Center, Washington Hospital Center, Northwest, Washington, District of Columbia, USA
| | - Kenneth D Burman
- Division of EndocrinologyDepartment of Medicine, Medstar Washington Hospital Center, Washington Hospital Center, Northwest, Washington, District of Columbia, USA
| | - Leonard Wartofsky
- Division of EndocrinologyDepartment of Medicine, Medstar Washington Hospital Center, Washington Hospital Center, Northwest, Washington, District of Columbia, USA
| | - Vasyl Vasko
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
8
|
Zhang F, Yang B, Zhang K, Hou ML, Lu XC, Li YX. CCND1-BCL2 Gene Network: A direct target of Amifostine in human acute megakaryocytic leukemia cells. Chem Biol Drug Des 2017; 89:681-693. [PMID: 27762064 DOI: 10.1111/cbdd.12889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/19/2016] [Accepted: 09/23/2016] [Indexed: 11/30/2022]
Abstract
Amifostine, 2-(3-aminopropyl) aminoethyl phosphorothioate, is a broad-spectrum cytoprotective agent used to treat nuclear radiation and chemical weapon injuries. Recently, amifostine has been shown to have a profound biological influence on tumor cells. To examine the effects and mechanisms underlying the effects of amifostine on human acute megakaryocytic leukemia, we evaluated the efficacy of amifostine against Dami cells and observed a cell cycle arrest in G2 /M phase. Amifostine treatment also induced cell apoptosis of Dami cells which corresponds to formal studies. Through whole-genome microarray and bioinformatics analyses, we found that amifostine affected the gene expression of CCND1, BCL2, and CASP3 which revealed the mechanism amifostine acted on Dami cells. Thus, CCND1-BCL2 Gene Network is predicted to be a direct target of amifostine treating human acute megakaryocytic leukemia, which may provide a novel potential target for the therapy of several subtypes of human AML.
Collapse
Affiliation(s)
- Feng Zhang
- National Engineering Laboratory for Drug gable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, China.,Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Bo Yang
- Department of Geriatric Hematology, Chinese PLA General Hospital, Beijing, China
| | - Kailiang Zhang
- Schools of Stomatology, Lanzhou University, Lanzhou, China
| | - Mei-Ling Hou
- National Engineering Laboratory for Drug gable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, China
| | - Xue-Chun Lu
- Department of Geriatric Hematology, Chinese PLA General Hospital, Beijing, China
| | - Yu-Xin Li
- National Engineering Laboratory for Drug gable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, China
| |
Collapse
|
9
|
Chen Q, Chen L, Kong D, Shao J, Wu L, Zheng S. Dihydroartemisinin alleviates bile duct ligation-induced liver fibrosis and hepatic stellate cell activation by interfering with the PDGF-βR/ERK signaling pathway. Int Immunopharmacol 2016; 34:250-258. [PMID: 27038258 DOI: 10.1016/j.intimp.2016.03.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 03/10/2016] [Accepted: 03/10/2016] [Indexed: 01/29/2023]
Abstract
Liver fibrosis represents a frequent event following chronic insult to trigger wound healing responses in the liver. Activation of hepatic stellate cells (HSCs), which is a pivotal event during liver fibrogenesis, is accompanied by enhanced expressions of a series of marker proteins and pro-fibrogenic signaling molecules. Artemisinin, a powerful antimalarial medicine, is extracted from the Chinese herb Artemisia annua L., and can inhibit the proliferation of cancer cells. Dihydroartemisinin (DHA), the major active metabolite of artemisinin, is able to attenuate lung injury and fibrosis. However, the effect of DHA on liver fibrosis remains unclear. The aim of this study was to investigate the effect of DHA on bile duct ligation-induced injury and fibrosis in rats. DHA improved the liver histological architecture and attenuated collagen deposition in the fibrotic rat liver. Experiments in vitro showed that DHA inhibited the proliferation of HSCs and arrested the cell cycle at the S checkpoint by altering several cell-cycle regulatory proteins. Moreover, DHA reduced the protein expressions of a-SMA, α1 (I) collagen and fibronectin, being associated with interference of the platelet-derived growth factor β receptor (PDGF-βR)-mediated ERK pathway. These data collectively revealed that DHA relieved liver fibrosis possibly by targeting HSCs via the PDGF-βR/ERK pathway. DHA may be a therapeutic antifibrotic agent for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Qin Chen
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Lianyun Chen
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Desong Kong
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; Department of Science, Technology and Education, the Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Jiangjuan Shao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Li Wu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
10
|
Singh VK, Fatanmi OO, Wise SY, Newman VL, Romaine PLP, Seed TM. THE POTENTIATION OF THE RADIOPROTECTIVE EFFICACY OF TWO MEDICAL COUNTERMEASURES, GAMMA-TOCOTRIENOL AND AMIFOSTINE, BY A COMBINATION PROPHYLACTIC MODALITY. RADIATION PROTECTION DOSIMETRY 2016; 172:302-310. [PMID: 27542813 PMCID: PMC5444681 DOI: 10.1093/rpd/ncw223] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This study was designed to evaluate the possible potentiation of survival protection afforded by relatively low-dose amifostine prophylaxis against total body irradiation in combination with a protective, less toxic agent, gamma-tocotrienol (GT3). Mice were administered amifostine and/or GT3, then exposed to 9.2 Gy 60Co γ-irradiation and monitored for survival for 30 days. To investigate cytokine stimulation, mice were administered amifostine or GT3; serum samples were collected and analyzed for cytokines. Survival studies show single treatments of GT3 or amifostine significantly improved survival, compared to the vehicle, and combination treatments resulted in significantly higher survival compared to single treatments. In vivo studies with GT3 confirmed prior work indicating GT3 induces granulocyte colony-stimulating factor (G-CSF). This approach, the prophylactic combination of amifostine and GT3, which act through different mechanisms, shows promise and should be investigated further as a potential countermeasure for acute radiation syndrome.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Oluseyi O Fatanmi
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Stephen Y Wise
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Victoria L Newman
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Patricia L P Romaine
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | | |
Collapse
|
11
|
Miller TW, Soto-Pantoja DR, Schwartz AL, Sipes JM, DeGraff WG, Ridnour LA, Wink DA, Roberts DD. CD47 Receptor Globally Regulates Metabolic Pathways That Control Resistance to Ionizing Radiation. J Biol Chem 2015; 290:24858-74. [PMID: 26311851 DOI: 10.1074/jbc.m115.665752] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Indexed: 11/06/2022] Open
Abstract
Modulating tissue responses to stress is an important therapeutic objective. Oxidative and genotoxic stresses caused by ionizing radiation are detrimental to healthy tissues but beneficial for treatment of cancer. CD47 is a signaling receptor for thrombospondin-1 and an attractive therapeutic target because blocking CD47 signaling protects normal tissues while sensitizing tumors to ionizing radiation. Here we utilized a metabolomic approach to define molecular mechanisms underlying this radioprotective activity. CD47-deficient cells and cd47-null mice exhibited global advantages in preserving metabolite levels after irradiation. Metabolic pathways required for controlling oxidative stress and mediating DNA repair were enhanced. Some cellular energetics pathways differed basally in CD47-deficient cells, and the global declines in the glycolytic and tricarboxylic acid cycle metabolites characteristic of normal cell and tissue responses to irradiation were prevented in the absence of CD47. Thus, CD47 mediates signaling from the extracellular matrix that coordinately regulates basal metabolism and cytoprotective responses to radiation injury.
Collapse
Affiliation(s)
- Thomas W Miller
- From the Laboratory of Pathology and Paradigm Shift Therapeutics, Rockville, Maryland 20852, and
| | - David R Soto-Pantoja
- From the Laboratory of Pathology and Departments of Cancer Biology and Hypertension and Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | | | | | - William G DeGraff
- Radiation Biology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Lisa A Ridnour
- Radiation Biology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - David A Wink
- Radiation Biology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | | |
Collapse
|
12
|
|
13
|
Ormsby RJ, Lawrence MD, Blyth BJ, Bexis K, Bezak E, Murley JS, Grdina DJ, Sykes PJ. Protection from radiation-induced apoptosis by the radioprotector amifostine (WR-2721) is radiation dose dependent. Cell Biol Toxicol 2014; 30:55-66. [DOI: 10.1007/s10565-014-9268-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 01/09/2014] [Indexed: 12/11/2022]
|
14
|
Vendramini V, Robaire B, Miraglia SM. Amifostine-doxorubicin association causes long-term prepubertal spermatogonia DNA damage and early developmental arrest. Hum Reprod 2012; 27:2457-66. [DOI: 10.1093/humrep/des159] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
15
|
Xu Y, Parmar K, Du F, Price BD, Sun Y. The radioprotective agent WR1065 protects cells from radiation damage by regulating the activity of the Tip60 acetyltransferase. INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2011; 2:295-302. [PMID: 22187663 PMCID: PMC3242426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 09/28/2011] [Indexed: 05/31/2023]
Abstract
BACKGROUND The aminothiol WR1065 is a highly effective free radical scavenger which can protect cells from the cytotoxic effects of ionizing radiation. Currently, WR1065 is used clinically to protect patients from radiation injury occurring during radiation therapy protocols. However, it is becoming increasingly clear that WR1065 can alter radiosensitivity through a mechanism which is independent of its ability to function as a free radical scavenger. Here, we examined the ability of WR1065 to directly regulate signaling pathways involved in the DNA damage response. METHODOLOGY The ability of WR1065 to enhance the survival of irradiated bone marrow cells and primary cultures was established. DNA damage signaling was monitored by measuring activation of the ATM kinase by western blot analysis and activation of Tip60 using an in vitro acetylation assay. Tip60 function was abrogated by expression of a catalytically inactive Tip60, and the effect on radiosensitivity evaluated. PRINCIPAL FINDINGS Treatment of cells with WR1065 led to a small but significant increase in the kinase activity of ATM. Further, WR1065 robustly activated the Tip60 acetyltransferase, which is a key upstream regulator of the ATM kinase. In addition, WR1065 directly activated the acetyltransferase activity of purified Tip60 in vitro, indicating a direct interaction between WR1065 and Tip60. Finally, cells with reduced levels of Tip60 activity exhibited a significant reduction in radioprotection by WR1065. CONCLUSIONS Direct regulation of Tip60's acetyltransferase activity by WR1065 makes a significant contribution to the radioprotective effects of WR1065. Activators of Tip60 may therefore make effective clinical radioprotectors.
Collapse
Affiliation(s)
- Ye Xu
- Department of Radiation Oncology, Dana-Farber Cancer InstituteHarvard Medical School, 450 Brookline Avenue, Boston, MA 02115USA
| | - Kalindi Parmar
- Department of Radiation Oncology, Dana-Farber Cancer InstituteHarvard Medical School, 450 Brookline Avenue, Boston, MA 02115USA
| | - Fengxia Du
- Disease Genomics and Individualized Medicine Key Lab, Beijing Institute of GenomicsChinese Academy of Sciences, No 7 Beitucheng West Road, Chaoyang District, Beijing 100029PR China
| | - Brendan D Price
- Department of Radiation Oncology, Dana-Farber Cancer InstituteHarvard Medical School, 450 Brookline Avenue, Boston, MA 02115USA
| | - Yingli Sun
- Disease Genomics and Individualized Medicine Key Lab, Beijing Institute of GenomicsChinese Academy of Sciences, No 7 Beitucheng West Road, Chaoyang District, Beijing 100029PR China
| |
Collapse
|
16
|
Saavedra MM, Henríquez-Hernández LA, Lara PC, Pinar B, Rodríguez-Gallego C, Lloret M. Amifostine modulates radio-induced apoptosis of peripheral blood lymphocytes in head and neck cancer patients. JOURNAL OF RADIATION RESEARCH 2010; 51:603-607. [PMID: 20699603 DOI: 10.1269/jrr.10030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Head and neck cancer is treated mainly with surgery and radiotherapy. Xerostomia and mucositis are common adverse effects of radiation therapy. One of the strategies aimed at decreasing radiation toxicity is the use of radioprotective agents, such as amifostine. We previously reported that radio induced apoptosis of peripheral blood lymphocytes was statistically associated with normal tissue toxicity in the form of severe xerostomia. The aim of the present study was to explore the effects of amifostine on the radiation-induced apoptosis of peripheral blood lymphocytes from patients suffering head and neck cancer. Eighteen consecutive patients with squamous cell carcinoma of the head and neck were included in the study. Peripheral blood lymphocytes were isolated before and after the treatment with amifostine. Then, cells were irradiated at 0, 1, 2 and 8 Gy during 24 hours. Apoptosis was measured by flow cytometry using annexin V/propidium iodide. As expected, radio-induced apoptosis values fitted to a semi logarithmic equation as follows: RIA = β ln(Gy) + α. The administration of amifostine prior to radiation therapy modulates radio-induced apoptosis of peripheral blood lymphocytes: 13.68 vs. 13.37 (P = 0.027), 19.11 vs. 17.64 (P = 0.001) and 30.70 vs. 28.84 (P = 0.001), before and after the administration of the drug for 1, 2 and 8 Gy respectively. α and β decreased significantly after the administration of the drug: 13.58 vs. 12.99 (P = 0.009) and 8.21 vs. 7.53 (P = 0.017), respectively. Our results provide new information about the biological actions of amifostine in vivo.
Collapse
Affiliation(s)
- Maite M Saavedra
- Radiotherapy Oncology Service, Hospital Universitario de Gran Canaria, Dr. Negrín, Spain
| | | | | | | | | | | |
Collapse
|
17
|
Dedieu S, Canron X, Rezvani HR, Bouchecareilh M, Mazurier F, Sinisi R, Zanda M, Moenner M, Bikfalvi A, North S. The cytoprotective drug amifostine modifies both expression and activity of the pro-angiogenic factor VEGF-A. BMC Med 2010; 8:19. [PMID: 20334641 PMCID: PMC2859403 DOI: 10.1186/1741-7015-8-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 03/24/2010] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Amifostine (WR-2721, delivered as Ethyol) is a phosphorylated aminothiol compound clinically used in addition to cis-platinum to reduce the toxic side effects of therapeutic treatment on normal cells without reducing their efficacy on tumour cells. Its mechanism of action is attributed to the free radical scavenging properties of its active dephosphorylated metabolite WR-1065. However, amifostine has also been described as a potent hypoxia-mimetic compound and as a strong p53 inducer; both effects are known to potently modulate vascular endothelial growth factor (VEGF-A) expression. The angiogenic properties of this drug have not been clearly defined. METHODS Cancer cell lines and endothelial cells were used in culture and treated with Amifostine in order to study (i) the expression of angiogenesis related genes and proteins and (ii) the effects of the drug on VEGF-A induced in vitro angiogenesis. RESULTS We demonstrated that the treatment of several human cancer cell lines with therapeutical doses of WR-1065 led to a strong induction of different VEGF-A mRNA isoforms independently of HIF-1alpha. VEGF-A induction by WR-1065 depends on the activation of the eIF2alpha/ATF4 pathway. This up-regulation of VEGF-A mRNA was accompanied by an increased secretion of VEGF-A proteins fully active in stimulating vascular endothelial cells (EC). Nevertheless, direct treatment of EC with amifostine impaired their ability to respond to exogenous VEGF-A, an effect that correlated to the down-regulation of VEGFR-2 expression, to the reduction in cell surface binding of VEGF-A and to the decreased phosphorylation of the downstream p42/44 kinases. CONCLUSIONS Taken together, our results indicate that amifostine treatment modulates tumour angiogenesis by two apparently opposite mechanisms - the increased VEGF-A expression by tumour cells and the inhibition of EC capacity to respond to VEGF-A stimulation.
Collapse
Affiliation(s)
- S Dedieu
- Inserm, U920, Talence, F-33400, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Lialiaris T, Kotsiou E, Pouliliou S, Kareli D, Makrinou H, Kouskoukis A, Papachristou F, Koukourakis M. Cytoprotective activity of amifostine on cultured human lymphocytes exposed to irinotecan. Food Chem Toxicol 2009; 47:2445-9. [DOI: 10.1016/j.fct.2009.06.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/18/2009] [Accepted: 06/29/2009] [Indexed: 10/20/2022]
|
19
|
Prieto González EA, Fuchs AG, Sánchez GS. Amifostine (WR2721) confers DNA protection to in vivo cisplatin-treated murine peripheral blood leukocytes. Dose Response 2009; 7:234-46. [PMID: 19809542 DOI: 10.2203/dose-response.08-026.prieto] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Amifostine [S-2-3-aminopropyl amino ethyl phosphorotioic acid], a modulator agent for antineoplastic drugs involved in free radicals generation has given controversial results in cisplatin treated leukocytes in vitro. We have evaluated the amifostine protection over leukocytes in vivo, using comet assay. Groups of five OF1 male mice were given one of three doses of amifostine (56, 105 and 200 mg/Kg) after a cisplatin single injection (10 mg/Kg). Serum malonyldialdehyde levels, catalase and superoxide dismutase activity were also evaluated. Amifostine showed significant DNA protection (p< 0.01) at the two lower doses evaluated. Malonyldialdehyde decreased in all amifostine treatments with respect to cisplatin while antioxidant enzyme activities remained unchanged. However, DNA migration increased with the highest amifostine dose; in fact highest dose of amifostine did no protect damage caused by cisplatin this result have implications on amifostine treatment schedules in clinical practice.
Collapse
Affiliation(s)
- E A Prieto González
- Centro de Altos Estudios en Ciencias de la Salud, Universidad Abierta Interamericana, Ave. Montes de Oca. No. 745, Buenos Aires Capital, Federal Argentina.
| | | | | |
Collapse
|
20
|
Grochová D, Šmardová J. The antimutagenic and cytoprotective effects of amifostine: the role of p53. J Appl Biomed 2007. [DOI: 10.32725/jab.2007.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
21
|
Pataer A, Fanale MA, Roth JA, Swisher SG, Hunt KK. Induction of apoptosis in human lung cancer cells following treatment with amifostine and an adenoviral vector containing wild-type p53. Cancer Gene Ther 2006; 13:806-14. [PMID: 16628227 DOI: 10.1038/sj.cgt.7700960] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenoviral delivery of the p53 gene is a potential therapeutic approach for the treatment of lung cancer. Furthermore, amifostine is a cytoprotective agent and recent reports have described its potentiation of chemotherapy's antitumor activity in lung cancer. Therefore, we wished to investigate the ability of amifostine both alone and in combination with p53-based therapy to induce apoptosis, and to understand the mechanisms by which this apoptosis occurs. Using p53 null and wild-type p53 human lung cancer cells and normal human bronchial epithelial cells, we evaluated the effects of amifostine on proliferation and apoptosis. We then analyzed Adp53 in combination with amifostine and performed isobologram analysis. Expression of p53, p21(WAF1), Bax, Bak, bcl-2, as well as total and phosphorylated Cdc2 in the absence and presence of olomoucine, a phosphorylated Cdc2 kinase inhibitor, was then determined. Amifostine-induced apoptosis in human lung cancer cells in a dose-dependent fashion. The combination of amifostine and Adp53 significantly enhanced, with a supra-additive effect, the inhibition of proliferation of lung cancer cells. This enhancement of apoptosis by amifostine was associated with activation of p53 and dephosphorylation of Cdc2 proteins. Notably, olomoucine effectively prevented amifostine and/or Adp53-induced Cdc2 kinase activation and subsequent apoptosis. Our data shows that amifostine alone can induce apoptosis of human lung cancer cells, and that the combination of Adp53 with amifostine resulted in significantly higher levels of apoptosis. In addition, it appears that Cdc2 kinase plays an important role in the induction of apoptosis by amifostine and Adp53.
Collapse
Affiliation(s)
- A Pataer
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
22
|
Ivanova T, Han Y, Son HJ, Yun YS, Song JY. Antimutagenic effect of polysaccharide ginsan extracted from Panax ginseng. Food Chem Toxicol 2006; 44:517-21. [PMID: 16242229 DOI: 10.1016/j.fct.2005.08.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2005] [Revised: 06/28/2005] [Accepted: 08/30/2005] [Indexed: 11/16/2022]
Abstract
Ginsan is a polysaccharide extracted from the roots of Panax ginseng, and it has earlier been reported to have an immunostimulatory effect. In the present study, the frequency of micronucleated polychromatic erythrocytes (MNPCE) was assessed in the bone marrow of C57BL/6 male mice treated with ginsan [100, 200 or 300 mg/kg body weight (b.w.)] or amifostine (200mg/kg b.w.) 30 min before as well as 15 min after 1.5 Gy of gamma-irradiation. Ginsan and amifostine did not alter the frequency of MNPCE of control mice (P>0.05), showing that they are non-mutagenic per se; gamma-irradiation induced a statistically significant (P<0.001) increase of MNPCE and decrease of PCE/NCE ratio (P<0.001) compared to control group. However, ginsan applied 30 min before or 15 min after irradiation reduced MNPCE in a dose-dependent manner. Amifostine (200mg/kg b.w.) did not reduce radiation-induced MNPCE, but stimulated erythropoiesis, when administered before irradiation. Based on the above results, radioprotective effect of ginsan can be partially attributed to reduction of radiation-induced genotoxicity.
Collapse
Affiliation(s)
- Tatiana Ivanova
- Medical Radiological Research Center, Russian Academy of Medical Sciences, Obninsk 249036, Russia
| | | | | | | | | |
Collapse
|
23
|
Dai D, Holmes AM, Nguyen T, Davies S, Theele DP, Verschraegen C, Leslie KK. A potential synergistic anticancer effect of paclitaxel and amifostine on endometrial cancer. Cancer Res 2005; 65:9517-24. [PMID: 16230417 DOI: 10.1158/0008-5472.can-05-1613] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although paclitaxel is one of the most effective chemotherapeutic agents, its usefulness is still limited in advanced and recurrent endometrial cancer. Amifostine protection of normal tissues against the side effects of chemotherapeutic agents has been clinically proven in cancer patients; however, its application in endometrial cancer has not been fully evaluated. We have investigated the use of paclitaxel and amifostine in controlling the growth of poorly differentiated endometrial cancer cells, Hec50co, in vitro and in vivo. Our studies show that amifostine had direct anticancer effects on endometrial cancer cells in vitro by arresting the cell cycle at the G1 phase and inducing apoptosis. Amifostine also inhibited s.c. tumor growth in athymic mice. Paclitaxel IC50 value was reduced from 14 to 2 nmol/L with pretreatment of a single dose of 178 micromol/L of amifostine for 72 hours. Amifostine also synergized with paclitaxel in the arrest of the cell cycle at the G2-M phase and in the induction of apoptosis. This two-drug regimen inhibited s.c. tumor growth as well as improved mouse survival significantly more than paclitaxel alone. Amifostine also significantly improved paclitaxel-induced cytotoxic effects on peripheral blood profiles. Our studies show that amifostine has direct anticancer effects on endometrial cancer. Our data have also shown a potential anticancer synergy between amifostine and paclitaxel in vitro and in vivo, whereas amifostine maintained a protective role in peripheral blood profiles. The dual specificity of amifostine action should be further investigated.
Collapse
Affiliation(s)
- Donghai Dai
- Reproductive Molecular Biology Laboratory, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology.
| | | | | | | | | | | | | |
Collapse
|
24
|
Ozcan Arican G. Cytoprotective effects of amifostine and cysteamine on cultured normal and tumor cells treated with paclitaxel in terms of mitotic index and 3H-thymidine labeling index. Cancer Chemother Pharmacol 2005; 56:221-9. [PMID: 15838658 DOI: 10.1007/s00280-004-0954-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2004] [Accepted: 11/05/2004] [Indexed: 11/27/2022]
Abstract
PURPOSE In this study, the cytotoxic effects of paclitaxel (PAC) in normal and tumor cells were established, and the cytoprotective effects of amifostine (AMI) and cysteamine (CYS) against this cytotoxicity were examined. METHODS Tumor cell lines used in this study were L-strain cells of mouse subcutaneous origin and human cervix carcinoma-derived HeLa cells. Mouse embryonic fibroblasts (MEFs) were used as the normal cell line. Results of the experiments were evaluated in terms of the mitotic index and the 3H-thymidine labeling index. PAC concentrations of 6 and 12 microg/ml were applied to the cells for 1-10 days either alone or in combination with 1 mug/ml of AMI and CYS. RESULTS In terms of the above parameters, statistically significant effects were not seen in cultures of any of the cell lines treated with 1 microg/ml of AMI or CYS alone. In contrast, both concentrations of PAC caused increasing cytotoxic effects with increasing treatment time (P < .001). The cytotoxic effect of PAC appeared as mitotic phase accumulation (G2/M blockage) and a subsequent decline in the synthesis phase. HeLa cells were very sensitive to PAC treatment, whereas MEF cells were quite resistant compared with tumor cells. In cells treated with combined drugs to investigate the cytoprotective effects of AMI and CYS on normal and tumor cell lines, PAC continued to show cytotoxic effects in tumor cells, but this effect was reduced in the normal cells. CONCLUSIONS AMI and CYS did not protect tumor cells against the cytotoxic effects of PAC, but protection was observed in normal cells. Furthermore, the protection provided by AMI was stronger than that provided by CYS.
Collapse
Affiliation(s)
- Gül Ozcan Arican
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
25
|
Mann K, Hainaut P. Aminothiol WR1065 induces differential gene expression in the presence of wild-type p53. Oncogene 2005; 24:3964-75. [PMID: 15750621 DOI: 10.1038/sj.onc.1208563] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The aminothiol WR1065 exerts selective cytoprotective effects in normal cells compared to cancer cells and has clinical applications for the protection of normal cells in cancer patients undergoing radio- or chemotherapy. There is evidence that p53 is activated in response to WR1065. To examine the effects of WR1065 on the signalling pathways controlled by p53, isogeneic human colon carcinoma cell lines (HCT116) differing only in the presence or absence of wild-type p53 were used. Treatment with WR1065 resulted in G1 cell cycle arrest in the p53-positive cell line but not in the p53-negative cell line. Long-term exposure resulted in minimal apoptosis of either cell line. Changes in gene expression in p53-positive or -negative cells treated with WR1065 were examined using commercial human stress and cancer gene arrays (Clontech Atlas arrays). Genes found to be specifically upregulated in a p53-dependent manner included coproporphyrinogen oxidase, ICErel-II cysteine protease, macrophage inhibitory cytokine-1 (also known as placental transforming growth factor beta), S100A4, and Waf1/p21. However, most proapoptotic genes typically upregulated by p53 in response to DNA damage were not activated. These studies show that WR1065 specifically modulates a subset of p53 target genes in a colon carcinoma cell line, consistent with the observation that this agent elicits essentially p53-dependent, cell cycle arrest responses.
Collapse
Affiliation(s)
- Kristine Mann
- Department of Biological Sciences and Biomedical Program, University of Alaska, 3211 Providence Drive, Anchorage, AK 99508, USA.
| | | |
Collapse
|
26
|
Vellón L, González-Cid M, Nebel MDC, Larripa I. Additive apoptotic effect of STI571 with the cytoprotective agent amifostine in K-562 cell line. Cancer Chemother Pharmacol 2005; 55:602-8. [PMID: 15700189 DOI: 10.1007/s00280-004-0952-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Accepted: 07/30/2004] [Indexed: 10/25/2022]
Abstract
PURPOSE To study the apoptotic effect of the 2-phenylaminopyrimidine derivative STI571 in combination with antioxidant agents on K-562 cell line derived from a Philadelphia chromosome-positive chronic myeloid leukemia patient. MATERIALS AND METHODS K-562 (BCR/ABL+), U-937, and HL60 (BCR/ABL-) leukemic cell lines were incubated with STI571 and the antioxidant agents catalase, glutathione, superoxide dismutase, and amifostine (AMI). Apoptotic effect was analyzed by morphological and flow cytometric criteria. RESULTS STI571 at concentrations higher than 0.25 mumol L(-1) produced apoptosis (P<0.05) in K-562 cells only after treatment for 72 h. At the mentioned concentrations, STI571 also induced an increase in the loss of mitochondrial transmembrane potential from 24.6 to 40%. Combination of STI571 (0.5 micromol L(-1)) with antioxidant agents showed that the cytoprotective agent AMI (0.75 mg mL(-1)) produced an additive effect in the proapoptotic activity of STI571 in K-562 cells at nuclear (58.8%+/-2.0 vs. 28.9%+/-3.3) and mitochondrial (53.3%+/-3.6 vs. 29.5%+/-1.2) levels. CONCLUSIONS Our results show that only AMI in combination with STI571, at submicromolar concentration, has an additive effect in K-562 cell line, and it does not have severe toxic effects on Philadelphia chromosome negative cells.
Collapse
Affiliation(s)
- Luciano Vellón
- Depto de Genética, Instituto de Investigaciones Hematológicas Mariano R. Castex, Academia de Medicina, J.A. Pacheco de Melo 3081, 1425 Buenos Aires, Argentina
| | | | | | | |
Collapse
|
27
|
Wang ZW, Zhou JM, Huang ZS, Yang AP, Liu ZC, Xia YF, Zeng YX, Zhu XF. Aloe polysaccharides mediated radioprotective effect through the inhibition of apoptosis. JOURNAL OF RADIATION RESEARCH 2004; 45:447-454. [PMID: 15613791 DOI: 10.1269/jrr.45.447] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Polysaccharides from aloe are always considered an effective radioprotector on irradiation-induced skin damage. The aim of this study was to determine if aloe polysaccharides (AP) have radioprotective effects on normal human cells in vitro and mouse survival in vivo and to explore the mechanism. Pretreatment with 50 microg/ml AP could improve the surviving fraction at 2 Gy (SF2) of three normal cell lines 293, ECV304, and C. liver from 41.5%, 46.5%, and 40.9% to 49.4%, 72.1%, and 89.1%, respectively. AP could also reduce the apoptotic rate of C. liver cells from 9.5% and 43.0% to 2.2% and 10.9% 48 h and 72 h after 2 Gy irradiation, respectively. Western blot analysis showed that pretreatment with AP could block the upregulation of pro-apoptotic p53, Bax, and Bad and the downregulation of Bcl-2 by irradiation. AP could lower thymocyte apoptosis of mice in vivo after 6 Gy irradiation and abrogate the cell cycle perturbation. Fifty mg/kg of AP treatment for 30 min before 7.5 Gy irradiation provided the best radioprotective effect and improved the 30-day survival rate of mice to 86.0%, from 10.0%. AP exerted radioprotective effects in vitro and in vivo through an inhibition of apoptosis.
Collapse
Affiliation(s)
- Zong-Wei Wang
- Department of Pharmacology, Guangzhou University of Traditional Chinese Medicine School of Pharmaceutical Science, Guangzhou, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|