1
|
Chang J, Yan S, Geng Z, Wang Z. Inhibition of splicing factors SF3A3 and SRSF5 contributes to As 3+/Se 4+ combination-mediated proliferation suppression and apoptosis induction in acute promyelocytic leukemia cells. Arch Biochem Biophys 2023; 743:109677. [PMID: 37356608 DOI: 10.1016/j.abb.2023.109677] [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: 04/17/2023] [Revised: 05/28/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
The low-dose combination of Arsenite (As3+) and selenite (Se4+) has the advantages of lower biological toxicity and better curative effects for acute promyelocytic leukemia (APL) therapy. However, the underlying mechanisms remain unclear. Here, based on the fact that the combination of 2 μM A3+ plus 4 μM Se4+ possessed a stronger anti-leukemic effect on APL cell line NB4 as compared with each individual, we employed iTRAQ-based quantitative proteomics to identify a total of 58 proteins that were differentially expressed after treatment with As3+/Se4+ combination rather than As3+ or Se4+ alone, the majority of which were involved in spliceosome pathway. Among them, eight proteins stood out by virtue of their splicing function and significant changes. They were validated as being decreased in mRNA and protein levels under As3+/Se4+ combination treatment. Further functional studies showed that only knockdown of two splicing factors, SF3A3 and SRSF5, suppressed the growth of NB4 cells. The reduction of SF3A3 was found to cause G1/S cell cycle arrest, which resulted in proliferation inhibition. Moreover, SRSF5 downregulation induced cell apoptosis through the activation of caspase-3. Taken together, these findings indicate that SF3A3 and SRSF5 function as pro-leukemic factors and can be potential novel therapeutic targets for APL.
Collapse
Affiliation(s)
- Jiayin Chang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, PR China
| | - Shihai Yan
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, PR China
| | - Zhirong Geng
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, PR China.
| | - Zhilin Wang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, PR China.
| |
Collapse
|
2
|
Tabary M, Aryannejad A, Noroozi N, Tavangar SM, Mohammad Jafari R, Araghi F, Dadkhahfar S, Dehpour AR. Ivermectin Increases Random-Pattern Skin Flap Survival in Rats: The Novel Role of GABAergic System. J Surg Res 2021; 259:431-441. [PMID: 33069391 DOI: 10.1016/j.jss.2020.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ivermectin (IVM) was first used as an antiparasitic agent; however, the role of this drug evolved into a broad spectrum. Many mechanisms have been proposed, including interaction with the GABAergic system. Considering the presence of GABA receptor in the skin tissue and its role in ischemia-reperfusion I/R injury, we aimed to evaluate the effect of IVM through GABA receptors on random-pattern skin flap survival. METHODS Sixty Wistar male rats were used. Multiple doses of IVM (0.01, 0.05, 0.2, and 0.5 mg/kg) were injected intraperitoneally before the surgery. Baclofen (selective GABAB agonist) and bicuculline (selective GABAA antagonist) were administered in combination with IVM to assess the role of the GABAergic system. Histopathological evaluations, immunohistochemical staining, quantitative assessment of IL-1β and TNFα, and the expression of GABAA α1 subunit and GABAB R1 receptors were evaluated in the skin tissue. RESULTS IVM 0.05 mg/kg could significantly increase flap survival compared with the control group (P < 0.001). Subeffective dose of baclofen (0.1 mg/kg) had synergistic effect with the subeffective dose of IVM (0.01 mg/kg) (P < 0.001), whereas bicuculline 1 mg/kg reversed the effect of IVM (0.05 mg/kg) (P < 0.001). IVM 0.05 mg/kg could also decrease the IL-1β and TNFα levels and increase the expression of GABAA α1 subunit and GABAB R1 receptors in the flap tissue compared with the control group. CONCLUSIONS IVM could improve skin flap survival, probably mediated by the GABAergic pathway. Both GABAA and GABAB receptors are involved in this process. This finding may repurpose the use of old drug, "Ivermectin."
Collapse
Affiliation(s)
- Mohammadreza Tabary
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Armin Aryannejad
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nafise Noroozi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Araghi
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Dadkhahfar
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Chillè D, Aiello D, Grasso GI, Giuffrè O, Napoli A, Sgarlata C, Foti C. Complexation of As(III) by phosphonate ligands in aqueous fluids: Thermodynamic behavior, chemical binding forms and sequestering abilities. J Environ Sci (China) 2020; 94:100-110. [PMID: 32563473 DOI: 10.1016/j.jes.2020.03.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 06/11/2023]
Abstract
In recent years, the contamination of water by arsenic reached alarming levels in many countries of the world, attracting the interest of many researchers engaged in testing methodologies able to remove this harmful pollutant. An important aspect that must be taken into consideration is the possibility to find arsenic in different chemical forms which could require different approaches for its removal. At this aim, a speciation analysis appears to be crucial for better understanding the behavior of arsenic species in aqueous solutions, especially in presence of compounds with marked chelating properties. Phosphonates can be identified as good sequestering agents and, at this purpose, this manuscript intends to investigate the interaction of As(III) with three phosphonic acids derived from nitrilotriacetic acid (NTA) by replacements of one (N-(Phosphonomethyl) iminodiacetic acid, NTAP), two (N,N-Bis-(phosphonomethyl) glycine, NTA2P) and three (Nitrilotri(methylphosphonic acid), NTA3P) carboxylic groups with the same number of phosphonate groups. An in-depth potentiometric and calorimetric investigation allowed to determine speciation models featured by simple ML, MLHi and ML(OH) species. A complete thermodynamic characterization of the systems is reported together with the definition of coordination mode by mass spectrometry measurements. On the light of the speciation models, the possibility of using these ligands in arsenic removal techniques was assessed by determining the pL0.5 (the concentration of ligand able to remove the 50% of metal ion present in trace). All ligands show a good sequestering ability, in particular under the conditions of fresh water, following the trend NTA3P > NTA2P > NTAP.
Collapse
Affiliation(s)
- Donatella Chillè
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Donatella Aiello
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Giuseppa Ida Grasso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Ottavia Giuffrè
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Anna Napoli
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Carmelo Sgarlata
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Claudia Foti
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| |
Collapse
|
4
|
Khatami F, Larijani B, Heshmat R, Nasiri S, Haddadi-Aghdam M, Teimoori-Toolabi L, Tavangar SM. Hypermethylated RASSF1 and SLC5A8 promoters alongside BRAF V600E mutation as biomarkers for papillary thyroid carcinoma. J Cell Physiol 2020; 235:6954-6968. [PMID: 32017063 DOI: 10.1002/jcp.29591] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
Abstract
Circulating cell-free DNA (cfDNA) has been considered as a diagnostic source to track genetic and epigenetic alterations in cancer. We aimed to study mutation in addition to the methylation status in the promoter regions of RASSF1 and SLC5A8 genes in tissues and circulating free DNA samples of patients affected with papillary thyroid carcinoma (PTC) and thyroid nodules as controls. BRAFV600E mutation was studied by ARMS-scorpion real-time polymerase chain reaction method in 57 PTC and 45 thyroid nodule cases. Methylation status of RASSF1 and SLC5A8 promoter regions was analyzed by methylation-specific high-resolution melting curve analysis. BRAFV600E mutation was found in 39 (68.4%) out of 57 PTC tissue samples, while in 33 (49.1%) cases of cfDNA, this mutation was detected. The frequency of BRAFV600E mutation in cfDNA was significantly different between metastatic and nonmetastatic PTC cases (22 of 33 PTC cases vs. 5 of 34 thyroid nodule samples). Methylation levels of three promoter regions of SLC5A8 and proximal promoter region of RASSF1 was significantly different between PTC and thyroid nodule cases in both cfDNA and tissue DNA. In addition, the methylation status of these two genes in tissue DNA was reflected in methylation status observed in cfDNA. This study confirmed that BRAFV600E mutation is better for discrimination between papillary thyroid carcinoma and thyroid nodules. On the other hand, hypermethylation in the more proximal promoter regions to RASSF1 and SLC5A8 genes showed higher sensitivity and more acceptable specificity for this discrimination.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirzad Nasiri
- Departments of Surgery, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Haddadi-Aghdam
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed M Tavangar
- Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Miodragović Ð, Swindell EP, Waxali ZS, Bogachkov A, O'Halloran TV. Beyond Cisplatin: Combination Therapy with Arsenic Trioxide. Inorganica Chim Acta 2019; 496:119030. [PMID: 32863421 PMCID: PMC7453736 DOI: 10.1016/j.ica.2019.119030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Platinum drugs (cisplatin, oxaliplatin, and carboplatin) and arsenic trioxide are the only commercial inorganic non-radioactive anticancer drugs approved by the US Food and Drug Administration. Numerous efforts are underway to take advantage of the synergy between the anticancer activity of cisplatin and arsenic trioxide - two drugs with strikingly different mechanisms of action. These include co-encapsulation of the two drugs in novel nanoscale delivery systems as well as the development of small molecule agents that combine the activity of these two inorganic materials. Several of these new molecular entities containing Pt-As bonds have broad anticancer activity, are robust in physiological buffer solutions, and form stable complexes with biopolymers. This review summarizes results from a number of preclinical studies involving the combination of cisplatin and As2O3, co-encapsulation and nanoformulation efforts, and the chemistry and cytotoxicity of the first member of platinum anticancer agents with an arsenous acid moiety bound to the platinum(II) center: arsenoplatins.
Collapse
Affiliation(s)
- Ðenana Miodragović
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Northeastern Illinois University, 5500 North St Louis Avenue, Chicago, Illinois 60625, United States
| | - Elden P Swindell
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Zohra Sattar Waxali
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Abraham Bogachkov
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Thomas V O'Halloran
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
6
|
Aghamir SMK, Heshmat R, Ebrahimi M, Ketabchi SE, Parichehreh Dizaji S, Khatami F. The Impact Of Succinate Dehydrogenase Gene (SDH) Mutations In Renal Cell Carcinoma (RCC): A Systematic Review. Onco Targets Ther 2019; 12:7929-7940. [PMID: 31579262 PMCID: PMC6771773 DOI: 10.2147/ott.s207460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 09/09/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Renal cell cancer (RCC) syndrome is linked to Krebs cycle compartments and their coding genes' alterations like succinate dehydrogenase genes (SDHx). Here we present a systematic review of the SDH genes' mutations and their impact on both RCC diagnosis and prognosis. METHODS This systematic review includes any study in which tissue samples of RCC are considered in correlation with the SDHx mutations, microsatellite instability (MSI), and protein expression. For this purpose, a systematic search of MEDLINE (PubMed), Scopus, Embase, and Web of Science databases was conducted and finally 5384 articles were recruited. All studies' content was checked to find the related ones which were 145 articles, which with data extraction were limited to nineteen. RESULTS The final selected nineteen studies investigating the SDHx role in RCC tumor genesis were included, among which fifteen were mutation analysis, three were just SDHx protein expression, and two were MSI and mutation analysis studies. A total of 432 RCC patients were reported by SDH mutations, and 64 patients with MSI and SDH expression change were reported in 514 surgically resected renal epithelial tumors. The most common mutation was the single nucleotide variant rs772551056 (c.137G>A) of SDHB. For SDHC, c.380A>G presented in 48 RCC patients, and for SDHA a novel germline mutation c.2T>C: p.M1T in an occasional case of gastrointestinal stromal tumor intricate with RCC. CONCLUSION RCC as an aggressive type of kidney cancer needs some biomarkers to be diagnosed exactly. It was shown recently that the succinate dehydrogenase gene variations can provide this diagnostic and prognostic biomarker. For this purpose, SDHB rs772551056 associated with its protein expression alterations can be taken into account. It is possible that a novel mutation of SDHA (c.2T>C: p.M1T) can provide evidence of GIST associated with RCC as well.
Collapse
Affiliation(s)
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Ebrahimi
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Somayeh Parichehreh Dizaji
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khatami
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Khatami F, Larijani B, Heshmat R, Nasiri S, Saffar H, Shafiee G, Mossafa A, Tavangar SM. Promoter Methylation of Four Tumor Suppressor Genes in Human Papillary Thyroid Carcinoma. IRANIAN JOURNAL OF PATHOLOGY 2019; 14:290-298. [PMID: 31754358 PMCID: PMC6824767 DOI: 10.30699/ijp.2019.94401.1922] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 07/27/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND & OBJECTIVE Papillary thyroid cancer (PTC) is considered to be the most common type of thyroid malignancies. Epigenetic alteration, in which the chromatin conformation and gene expression change without changing the sequence of DNA, can occur in some tumor suppressor genes and oncogenes. Methylation is the most common type of epigenetic alterations that can be an excellent indicator of PTC invasive behavior. METHODS In this research, we determined the promoter methylation status of four tumor suppressor genes (SLC5A8, RASSF1, MGMT, and DNMT1) and compared the results of 55 PTC cases with 40 goiter patients. For methylation, we used the methylation-sensitive high resolution melting (MS-HRM) assay technique. The resulting graphs of each run were compared with those of 0%, 50%, and 100% methylated controls. RESULTS Our data showed that the promoter methylation of SLC5A8, Ras association domain family member 1(RASSF1), and MGMT were significantly different between PTC tissue and goiter with P-value less than 0.05. The most significant differences were observed in RASSF1; 77.2% of hyper-methylated PTC patients versus 15.6% hyper-methylated goiter samples (P<0.001). CONCLUSION RASSF1 promoter methylation can be a PTC genetic marker. RASSF1 promoter methylation is under the impact of the methyltransferase genes (DNMT1 and MGMT), protein expression, and promoter methylation.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirzad Nasiri
- Department of Surgery, Tehran University of Medical Sciences, Shariati Hospital, Tehran, Iran
| | - Hiva Saffar
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Gita Shafiee
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azam Mossafa
- Department of Surgery, Tehran University of Medical Sciences, Shariati Hospital, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Khorsand-Rahimzadeh A, Khatami F, Sefidbakht S, Saffar H, Tavangar SM. Dyspnea as the First Manifestation of Silent Renal Cell Carcinoma. IRANIAN JOURNAL OF PATHOLOGY 2019; 14:87-93. [PMID: 31531106 PMCID: PMC6708564 DOI: 10.30699/ijp.14.1.87] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 12/24/2018] [Indexed: 11/29/2022]
Abstract
Renal cell carcinoma (RCC) accounts for only 3% of adult malignancies, and the lung is the most common site of metastasis of this tumor, which may be accompanied by pleural metastasis. However, solitary pleural involvement is very rare and its presentation with dyspnea as the first manifestation of RCC is extremely rare. We describe a 39-year-old male with episodes of dyspnea dating back 6 months prior to hospital admission. During paraclinical investigations, chest computed tomography (CT) demonstrated pleural effusion and multiple pulmonary nodules, raising the question of primary mesothelioma or metastasis from distant focus. Histopathology and immuno- histochemical examinations of pleural biopsy provided evidence of metastatic RCC of the clear cell type. Therefore, an abdominal contrast computed tomography (CT) was performed, revealing a 3 cm right renal mass, which was then removed by partial nephrectomy. Physicians and pathologists should be aware of unusual presentations of RCC with no symptoms attributable to the kidneys, including dyspnea as in our case.
Collapse
Affiliation(s)
| | - Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Salma Sefidbakht
- Dept. of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hiva Saffar
- Dept. of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Dept. of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Khatami F, Larijani B, Nikfar S, Hasanzad M, Fendereski K, Tavangar SM. Personalized treatment options for thyroid cancer: current perspectives. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:235-245. [PMID: 31571972 PMCID: PMC6750856 DOI: 10.2147/pgpm.s181520] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
Abstract
Thyroid cancer is one of the most common endocrine malignancies, with increasing incidence all over the world. In spite of good prognosis for differentiated thyroid carcinoma, for an unknown reason, about 5–10% of the patients, the cancer will show aggressive behavior, develop metastasis, and be refractory to treatment strategies like radioactive iodine. Regarding the genetic information, each thyroid cancer patient can be considered as an individual unique one, with unique genetic information. Contrary to standard chemotherapy drugs, target therapy components aim at one or more definite molecular pathway on cancer cells, so their selection is underlying patient’s genetic information. Nowadays, several mutations and rearrangements including BRAF, VEGF receptors, RET, and RET/PTC, KDR, KIT, PDGFRA, CD274, and JAK2 are taken into account for the therapeutic components like larotrectinib (TRK inhibitor), vemurafenib, sunitinib, sorafenib, selumetinib, and axitinib. With the new concept of personalized treatment of thyroid cancer diagnoses, planning treatment, finding out how well treatment will work, and estimating a prognosis has changed for the better over the last decade.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiarad Fendereski
- Pediateric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Khatami F, Aghamir SMK, Tavangar SM. Oncometabolites: A new insight for oncology. Mol Genet Genomic Med 2019; 7:e873. [PMID: 31321921 PMCID: PMC6732276 DOI: 10.1002/mgg3.873] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The new aspect of oncometabolite can be an indicator of genetic and epigenetic change and cancer biomarker.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Khatami F, Larijani B, Nasiri S, Tavangar SM. Liquid Biopsy as a Minimally Invasive Source of Thyroid Cancer Genetic and Epigenetic Alterations. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:19-29. [PMID: 32351906 PMCID: PMC7175608 DOI: 10.22088/ijmcm.bums.8.2.19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/22/2019] [Indexed: 12/12/2022]
Abstract
In the blood of cancer patients, some nucleic acid fragments and tumor cells can be found that make it possible to trace tumor changes through a simple blood test called “liquid biopsy”. The main components of liquid biopsy are fragments of DNA and RNA shed by tumors into the bloodstream and circulate freely (ctDNAs and ctRNAs). Tumor cells which are shed into the blood (circulating tumor cells or CTCs), and exosomes that have been investigated for non-invasive detection and monitoring several tumors including thyroid cancer. Genetic and epigenetic alterations of a thyroid tumor can be a driver for tumor genesis or essential for tumor progression and invasion. Liquid biopsy can be real-time representative of such genetic and epigenetic alterations to trace tumors. In thyroid tumors, the circulating BRAF mutation is now taken into account for both thyroid cancer diagnosis and determination of the most effective treatment strategy. Several recent studies have indicated the ctDNA methylation pattern of some iodine transporters and DNA methyltransferase as a diagnostic and prognostic biomarker in thyroid cancer as well. There has been a big hope that the recent advances of genome sequencing together with liquid biopsy can be a game changer in oncology.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirzad Nasiri
- Departments of Surgery, Tehran University of Medical Sciences, Shariati Hospital, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Departments of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Khatami F, Teimoori-Toolabi L, Heshmat R, Nasiri S, Saffar H, Mohammadamoli M, Aghdam MH, Larijani B, Tavangar SM. Circulating ctDNA methylation quantification of two DNA methyl transferases in papillary thyroid carcinoma. J Cell Biochem 2019; 120:17422-17437. [PMID: 31127647 DOI: 10.1002/jcb.29007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/06/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022]
Abstract
Papillary thyroid cancer (PTC) is the most common type of cancer among thyroid malignancies. Tumor-related methylation of circulating tumor DNA (ctDNA) in plasma could represent tumor specific alterations can be considered as good biomarkers in circulating tumor cells. In this study, we studied the methylation status of seven promoter regions of two DNA methyl Transferases (MGMT and DNMT1) genes as the methylated ctDNA in plasma and tissue samples of patients with PTC and goiter patients as noncancerous controls. METHODS Both ctDNA and tissue genomic DNA of 57 PTC and 45 Goiter samples were isolated. After bisulfite modification, the methylation status was studied by Methylation-Sensitive High Resolution Melting (MS-HRM) assay technique. Four promoter regions of O6-methylguanine-DNA methyltransferase (MGMT) and three promoter regions of DNA methyltransferase 1 (DNMT1) were assessed. RESULTS From seven candidate promoter regions of two methyltrasferase coding genes, the methylation status of ctDNA within MGMT (a), MGMT (c), MGMT (d), and DNMT1 (b) were meaningfully different between PTC cases and controls. However, the most significant differences were seen in circulating ctDNA MGMT (c) which was hypermethylated in 25 (43.9 %) of patients with PTC vs 2 (4. 4 %) of goiter samples. Between two selected DNA methyl transferase, the methylation of MGMT as the maintenance methyltransferase was significantly higher in PTC cases than goiter controls (P-value < .001). The resulting areas under the receiver operating characteristic (ROC) curve were 0.78 for MGMT (d) for PTC versus goiter samples that can represent the overall ability of MGMT (d) methylation status to discriminate between PTC and goiter patients. CONCLUSION Among seven candidate regions of ctDNA the MGMT (c) and MGMT (d) showed higher sensitivity and specificity for PTC as a suitable candidates as biomarkers of PTC.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirzad Nasiri
- Departments of Surgery, Tehran University of Medical Sciences, Shariati Hospital, Tehran, Iran
| | - Hiva Saffar
- Departments of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Mohammadamoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Departments of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Khatami F, Payab M, Sarvari M, Gilany K, Larijani B, Arjmand B, Tavangar SM. Oncometabolites as biomarkers in thyroid cancer: a systematic review. Cancer Manag Res 2019; 11:1829-1841. [PMID: 30881111 PMCID: PMC6395057 DOI: 10.2147/cmar.s188661] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Thyroid cancer (TC) is an important common endocrine malignancy, and its incidence has increased in the past decades. The current TC diagnosis and classification tools are fine-needle aspiration (FNA) and histological examination following thyroidectomy. The metabolite profile alterations of thyroid cells (oncometabolites) can be considered for current TC diagnosis and management protocols. METHODS This systematic review focuses on metabolite alterations within the plasma, FNA specimens, and tissue of malignant TC contrary to benign, goiter, or healthy TC samples. A systematic search of MEDLINE (PubMed), Scopus, Embase, and Web of Science databases was conducted, and the final 31 studies investigating metabolite biomarkers of TC were included. RESULTS A total of 15 targeted studies and 16 untargeted studies revealed several potential metabolite signatures of TC such as glucose, fructose, galactose, mannose, 2-keto-d-gluconic acid and rhamnose, malonic acid and inosine, cholesterol and arachidonic acid, glycosylation (immunoglobulin G [IgG] Fc-glycosylation), outer mitochondrial membrane 20 (TOMM20), monocarboxylate transporter 4 (MCT4), choline, choline derivatives, myo-/scyllo-inositol, lactate, fatty acids, several amino acids, cell membrane phospholipids, estrogen metabolites such as 16 alpha-OH E1/2-OH E1 and catechol estrogens (2-OH E1), and purine and pyrimidine metabolites, which were suggested as the TC oncometabolite. CONCLUSION Citrate was suggested as the first most significant biomarker and lactate as the second one. Further research is needed to confirm these biomarkers as the TC diagnostic oncometabolite.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran,
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sarvari
- Metabolomics and Genomics Research Center, Endocrinology and Metabolomics Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Gilany
- Metabolomics and Genomics Research Center, Endocrinology and Metabolomics Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Reproductive Biotechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, Acercr, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran,
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran,
- Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran,
| |
Collapse
|
14
|
Zaki Dizaji M, Ghaffari SH, Hosseini E, Alizadeh N, Rostami S, Momeny M, Alimoghaddam K, Ghavamzadeh A. Survivin isoform expression in arsenic trioxide-treated acute promyelocytic leukemia cell line and patients: The odd expression pattern of survivin-2α. Asia Pac J Clin Oncol 2016; 13:e21-e30. [PMID: 27770503 DOI: 10.1111/ajco.12589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/09/2015] [Accepted: 11/25/2015] [Indexed: 12/13/2022]
Abstract
AIM Survivin, an inhibitor of apoptosis protein, is overexpressed in most cancers and is associated with chemotherapy resistance, increased tumor recurrence and shorter patient survival. Several survivin splice variants have been described, and none of their expressions have been defined in acute promyelocytic leukemia (APL). METHODS Expression of the survivin gene isoforms (survivin, -2α, -2B, -ΔΕx3 and -3B) were analyzed in 50 peripheral blood and 19 bone marrow samples that were collected at different phases of the disease (diagnostic, remission and relapse) in APL patients treated with arsenic trioxide (ATO) as a front-line therapy. In addition, the human APL-derived cell line (NB4) was analyzed for the expression of survivin isoforms and capsase-3 in response to the ATO. RESULTS Survivin and its variants were overexpressed significantly in patient's bone marrow samples compared to peripheral blood or normal samples. Their expression was decreased after ATO treatment in both NB4 cells (except survivin-2α) and APL patients along with PML-RARα copy number reduction. Downregulation of survivin isoforms was associated with an increase in both caspase-3 gene expression and its enzymatic activity levels. In a patient who did not respond to ATO treatment, expression of survivin isoforms (except survivin-2α) were highly increased during the induction therapy. CONCLUSION Survivin isoforms are upregulated in APL patients, and their expression is diminished during the ATO treatment. In addition, overexpression of survivin and its variants (except survivin-2α) are associated with unfavorable results, suggesting that they may play an important role in mechanisms underlying the resistance of APL cells to ATO.
Collapse
Affiliation(s)
- Majid Zaki Dizaji
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Hosseini
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Alizadeh
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrbano Rostami
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamran Alimoghaddam
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Assis PA, De Figueiredo-Pontes LL, Lima ASG, Leão V, Cândido LA, Pintão CT, Garcia AB, Saggioro FP, Panepucci RA, Chahud F, Nagler A, Falcão RP, Rego EM. Halofuginone inhibits phosphorylation of SMAD-2 reducing angiogenesis and leukemia burden in an acute promyelocytic leukemia mouse model. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:65. [PMID: 26099922 PMCID: PMC4486128 DOI: 10.1186/s13046-015-0181-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 06/11/2015] [Indexed: 01/09/2023]
Abstract
Background Halofuginone (HF) is a low-molecular-weight alkaloid that has been demonstrated to interfere with Metalloproteinase-2 (MMP-2) and Tumor Growth Factor-β (TGF-β) function and, to present antiangiogenic, antiproliferative and proapoptotic properties in several solid tumor models. Based on the fact that high levels of Vascular Endothelial Growth Factor (VEGF) and increased angiogenesis have been described in acute myeloid leukemia and associated with disease progression, we studied the in vivo effects of HF using an Acute Promyelocytic Leukemia (APL) mouse model. Methods NOD/SCID mice were transplanted with leukemic cells from hCG-PML/RARA transgenic mice (TM) and treated with HF 150 μg/kg/day for 21 days. The leukemic infiltration and the percentage of VEGF+ cells were evaluated by morphology and flow cytometry. The effect of HF on the gene expression of several pro- and antiangiogenic factors, phosphorylation of SMAD2 and VEGF secretion was assessed in vitro using NB4 and HUVEC cells. Results HF treatment resulted in hematological remission with decreased accumulation of immature cell and lower amounts of VEGF in BM of leukemic mice. In vitro, HF modulated gene expression of several pro- and antiangiogenic factors, reduced VEGF secretion and phosphorylation of SMAD2, blocking TGF-β-signaling. Conclusion Taken together, our results demonstrate that HF inhibits SMAD2 signaling and reduces leukemia growth and angiogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0181-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patricia A Assis
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Lorena L De Figueiredo-Pontes
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Ana Silvia G Lima
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Vitor Leão
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Larissa A Cândido
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Carolina T Pintão
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Aglair B Garcia
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Fabiano P Saggioro
- Pathology Department, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Rodrigo A Panepucci
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Fernando Chahud
- Pathology Department, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Arnon Nagler
- Hematology Division and Cord Blood Bank, Chaim Sheba Medical Center, Tel Aviv University, Tel Hashomer, 6997801, Israel.
| | - Roberto P Falcão
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| | - Eduardo M Rego
- Hematology and Oncology Divisions of the Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049900, Brazil.
| |
Collapse
|
16
|
|
17
|
Dayyani F, Kantarjian H, O'Brien S, Pierce S, Jones D, Faderl S, Garcia-Manero G, Cortes J, Ravandi F. Outcome of therapy-related acute promyelocytic leukemia with or without arsenic trioxide as a component of frontline therapy. Cancer 2010; 117:110-5. [PMID: 20803607 DOI: 10.1002/cncr.25585] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 06/15/2010] [Accepted: 07/20/2010] [Indexed: 01/10/2023]
Abstract
BACKGROUND Patients with therapy-related acute promyelocytic leukemia (t-APL) have been commonly exposed to topoisomerase inhibitors and may potentially benefit from induction regimens omitting anthracyclines. METHODS Retrospective analysis of the outcomes of 29 patients with t-APL who were either treated with arsenic trioxide (ATO) and all-trans-retinoic acid (ATRA) or with standard ATRA plus anthracycline-based chemotherapy was performed. RESULTS Prior therapy included chemotherapy alone, radiation alone, or a combination of the 2 in 19%, 33%, and 47% of patients, respectively. The combination of ATO and ATRA (n = 19) for induction resulted in a similar remission rate compared with ATRA plus chemotherapy (n = 10) (89% vs 70%; P = .35). The median overall survival for the patients treated with ATRA plus ATO was not reached compared with that for patients treated with ATRA plus chemotherapy (161 weeks; P = .79). CONCLUSIONS In this cohort of t-APL patients, outcomes with ATO and ATRA appeared to be comparable to anthracycline-containing induction regimens. This combination may be preferable in t-APL patients to avoid any risk of anthracycline-induced toxicities.
Collapse
Affiliation(s)
- Farshid Dayyani
- Department of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Mandegary A, Hosseini R, Ghaffari SH, Alimoghaddam K, Rostami S, Ghavamzadeh A, Ghahremani MH. The expression of p38, ERK1 and Bax proteins has increased during the treatment of newly diagnosed acute promyelocytic leukemia with arsenic trioxide. Ann Oncol 2010; 21:1884-1890. [PMID: 20164150 DOI: 10.1093/annonc/mdq034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Promising reports exist regarding the use of arsenic trioxide (ATO) as first-line treatment in acute promyelocytic leukemia (APL). Although the in vitro effect of ATO is extensively studied, the in vivo mechanism(s) of ATO action is mostly unknown. PATIENTS AND METHODS Newly diagnosed APL patients were involved and received ATO (0.15 mg.kg/day) for 28 days as induction followed by consolidation therapy. Bone marrow (BM) aspirates were obtained on days 0, 14 and 28 of treatment for further molecular studies. Clinical findings and white blood cell counts were recorded as well. RESULTS Complete remission was observed in 17 (85%) patients with the median duration of 28 days (18-38) and cumulative dosage of median 280 mg (180-350). Hyperleukocytosis and APL differentiation syndrome (63%), gastrointestinal disorders (30%), liver enzyme elevation and night sweating (50%) were the most prevalent side-effects. The expression of Bax, ERK1 and p38 proteins and caspase-3 activity increased significantly in promyelocytes of BM aspirates at days 14 and 28 of induction therapy. CONCLUSION(S) These findings point toward the role of p38 and Bax in the induction of apoptosis, which was confirmed by increase in caspase-3 activity. However, the increase in ERK1 expression with regard to leukocytosis could translate to a proliferative/differentiation effect.
Collapse
Affiliation(s)
- A Mandegary
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman
| | - R Hosseini
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran
| | - S H Ghaffari
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran
| | - K Alimoghaddam
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran; Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - S Rostami
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran
| | - A Ghavamzadeh
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran
| | - M H Ghahremani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran; Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Momeny M, Zakidizaji M, Ghasemi R, Dehpour AR, Rahimi-Balaei M, Abdolazimi Y, Ghavamzadeh A, Alimoghaddam K, Ghaffari SH. Arsenic trioxide induces apoptosis in NB-4, an acute promyelocytic leukemia cell line, through up-regulation of p73 via suppression of nuclear factor kappa B-mediated inhibition of p73 transcription and prevention of NF-kappaB-mediated induction of XIAP, cIAP2, BCL-XL and survivin. Med Oncol 2009; 27:833-42. [PMID: 19763917 DOI: 10.1007/s12032-009-9294-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2009] [Accepted: 08/16/2009] [Indexed: 01/24/2023]
Abstract
The purpose of the present study is to evaluate the effects of arsenic trioxide (ATO) on human acute promyelocytic leukemia NB-4 cells. Microculture tetrazolium test, bromodeoxyuridine (BrdU) cell proliferation assay, caspase 3 activity assay, cell-based nuclear factor kappa B (NF-kappaB) phosphorylation measurement by ELISA and real-time RT-PCR were employed to appraise the effects of ATO on metabolic activity, DNA synthesis, induction of programmed cell death and NF-kappaB activation. The suppressive effects of ATO on metabolic potential, cell proliferation and NF-kappaB activation were associated with induction of apoptosis in NB-4 cells. In addition, an expressive enhancement in mRNA levels of p73, cyclin-dependent kinase inhibitor 1A (p21), tumor protein 53-induced nuclear protein 1 (TP53INP1), WNK lysine deficient protein kinase 2 (WNK2) and lipocalin 2 coupled with a significant reduction in transcriptional levels of NF-kappaB inhibitor beta (IKK2), Nemo, BCL2-like 1 (BCL-X(L)), inhibitor of apoptosis protein 1 (cIAP2), X-linked inhibitor of apoptosis protein (XIAP), survivin, Bcl-2, TIP60, ataxia telangiectasia (ATM), SHP-2 and sirtuin (SIRT1) were observed. Altogether, these issues show for the first time that ATO treatment could trammel cell growth and proliferation as well as induces apoptosis in NB-4 cells through induction of transcriptional levels of p73, TP53INP1, WNK2, lipocalin 2 as well as suppression of NF-kappaB-mediated induction of BCL-X(L), cIAP2, XIAP and survivin. Furthermore, the inductionary effects of ATO on transcriptional stimulation of p73 might be through cramping the NF-kappaB module (through suppression of p65 phosphorylation as well as transcriptional hindering of IKK2, ATM and Nemo) along with diminishing the mRNA expression of TIP60, SHP-2 and SIRT1.
Collapse
MESH Headings
- Acute-Phase Proteins/biosynthesis
- Acute-Phase Proteins/genetics
- Apoptosis/drug effects
- Arsenic Trioxide
- Arsenicals/pharmacology
- Ataxia Telangiectasia Mutated Proteins
- Baculoviral IAP Repeat-Containing 3 Protein
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Caspase 3/biosynthesis
- Caspase 3/genetics
- Cell Cycle Proteins/biosynthesis
- Cell Cycle Proteins/genetics
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Cell Line, Tumor/pathology
- Cyclin-Dependent Kinase Inhibitor p21/biosynthesis
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Heat-Shock Proteins/biosynthesis
- Heat-Shock Proteins/genetics
- Histone Acetyltransferases/biosynthesis
- Histone Acetyltransferases/genetics
- Humans
- I-kappa B Kinase/biosynthesis
- I-kappa B Kinase/genetics
- Inhibitor of Apoptosis Proteins/biosynthesis
- Inhibitor of Apoptosis Proteins/genetics
- Lipocalin-2
- Lipocalins/biosynthesis
- Lipocalins/genetics
- Lysine Acetyltransferase 5
- Microtubule-Associated Proteins/biosynthesis
- Microtubule-Associated Proteins/genetics
- NF-kappa B/antagonists & inhibitors
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/genetics
- Oxides/pharmacology
- Protein Serine-Threonine Kinases/biosynthesis
- Protein Serine-Threonine Kinases/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/biosynthesis
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- Sirtuin 1/biosynthesis
- Sirtuin 1/genetics
- Survivin
- Transcription, Genetic/drug effects
- Tumor Protein p73
- Tumor Suppressor Proteins/biosynthesis
- Tumor Suppressor Proteins/genetics
- Ubiquitin-Protein Ligases
- X-Linked Inhibitor of Apoptosis Protein/biosynthesis
- X-Linked Inhibitor of Apoptosis Protein/genetics
- bcl-X Protein/biosynthesis
- bcl-X Protein/genetics
Collapse
Affiliation(s)
- Majid Momeny
- Hematology, Oncology and Bone Marrow Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ravandi F. Therapy-related acute promyelocytic leukemia: further insights into the molecular basis of the disease and showing the way forward in therapy. Leuk Lymphoma 2009; 50:1073-4. [PMID: 19557628 DOI: 10.1080/10428190902995655] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
21
|
Ravandi F, Estey E, Jones D, Faderl S, O'Brien S, Fiorentino J, Pierce S, Blamble D, Estrov Z, Wierda W, Ferrajoli A, Verstovsek S, Garcia-Manero G, Cortes J, Kantarjian H. Effective treatment of acute promyelocytic leukemia with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab ozogamicin. J Clin Oncol 2009; 27:504-10. [PMID: 19075265 PMCID: PMC4881307 DOI: 10.1200/jco.2008.18.6130] [Citation(s) in RCA: 299] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Accepted: 08/20/2008] [Indexed: 02/03/2023] Open
Abstract
PURPOSE We examined the outcome of patients with newly diagnosed acute promyelocytic leukemia (APL) treated with all-trans-retinoic acid (ATRA) and arsenic trioxide (ATO) with or without gemtuzumab ozogamicin (GO) but without traditional cytotoxic chemotherapy. PATIENTS AND METHODS From February 2002 to March 2008, 82 patients with APL were treated with a combination of ATRA plus ATO. The first cohort of 65 patients received ATRA and ATO (beginning on day 10 of ATRA). High-risk patients (WBCs >or= 10 x 10(9)/L) received GO on the first day. From July 2007, the second cohort of 17 patients received ATRA and ATO concomitantly on day 1. They also received GO on day 1, if high risk, and if their WBC increased to more than 30 x 10(9)/L during induction. Monitoring for PML-RARA fusion gene was conducted after induction and throughout consolidation and follow-up. RESULTS Overall, 74 patients achieved complete remission (CR) and one achieved CR without full platelet recovery after the induction, for a response rate of 92%. Seven patients died at a median of 4 days (range, 1 to 24 days) after inclusion in the study from disease-related complications. The median follow-up is 99 weeks (range, 2 to 282 weeks). Among the responding patients, three experienced relapse at 39, 52, and 53 weeks. Three patients died after being in CR for 14, 21, and 71 weeks, all from a second malignancy. The estimated 3-year survival rate is 85%. CONCLUSION The combination of ATRA and ATO (with or without GO) as initial therapy for APL was effective and safe and can substitute chemotherapy-containing regimens.
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Unit 428, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cashin R, Burry L, Peckham K, Reynolds S, Seki JT. Acute renal failure, gastrointestinal bleeding, and cardiac arrhythmia after administration of arsenic trioxide for acute promyelocytic leukemia. Am J Health Syst Pharm 2008; 65:941-6. [DOI: 10.2146/ajhp060616] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Richard Cashin
- Pharmacy, David Thompson Health Region, Red Deer, Alberta, Canada
| | - Lisa Burry
- Department of Pharmacy, and Associate Scientist, Department of Medicine, Mount Sinai Hospital University, University of Toronto, Toronto, Ontario
| | - Kenneth Peckham
- Hematology Services, Princess Margaret Hospital, University Health Network (UHN), Toronto
| | - Stuart Reynolds
- UHN, Toronto, and Staff Intensivist, Mount Sinai Hospital, Toronto
| | - Jack T. Seki
- Princess Margaret Hospital, UHN, and Assistant Professor, Pharm.D. Program, University of Toronto, Toronto
| |
Collapse
|
23
|
Martínez-Poveda B, Quesada AR, Medina MÁ. The anti-angiogenic 8-epipuupehedione behaves as a potential anti-leukaemic compound against HL-60 cells. J Cell Mol Med 2008; 12:701-6. [PMID: 18419606 PMCID: PMC3822555 DOI: 10.1111/j.1582-4934.2007.00134.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 10/03/2007] [Indexed: 01/08/2023] Open
Abstract
We have previously reported that 8-epipuupehedione, a synthetic derivative of sesquiterpenes found in several kinds of sponges, is a potent inhibitor of angiogenesis. Here, we show that 8-epipuupehedione is also a potent anti- leukaemic compound, targeting three hallmarks of malignancy: proliferation, survival and extra-cellular matrix re-modelling. To fulfil this goal, we use the HL-60 promyeolocytic cells as our model system and the following experimental procedures: cell growth assay, Hoetsch staining, cell cycle analysis and DNA fragmentation, caspase 3 activity and zymographic assays. Our results show that this compound inhibits proliferation and has potent and specific pro-apoptotic effects on HL-60 promyelocytic cells, inducing their nuclei and DNA fragmentation, as well as caspase 3 activity activation. Furthermore, 8-epipuupehedione strongly inhibits matrix metalloproteinase-2 and urokinase production by HL-60 cells. These results suggest that 8-epipuupehedione could be an attractive drug for further evaluation in the treatment of leukemia.
Collapse
Affiliation(s)
- Beatriz Martínez-Poveda
- Department of Molecular Biology and Biochemistry, Faculty of Sciences,
University of Málaga, Málaga, Spain
| | - Ana R Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences,
University of Málaga, Málaga, Spain
- ClBERER, Málaga, Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences,
University of Málaga, Málaga, Spain
- ClBERER, Málaga, Spain
| |
Collapse
|
24
|
Almeida B, Silva A, Mesquita A, Sampaio-Marques B, Rodrigues F, Ludovico P. Drug-induced apoptosis in yeast. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1436-48. [PMID: 18252203 DOI: 10.1016/j.bbamcr.2008.01.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 12/21/2007] [Accepted: 01/07/2008] [Indexed: 01/04/2023]
Abstract
In order to alter the impact of diseases on human society, drug development has been one of the most invested research fields. Nowadays, cancer and infectious diseases are leading targets for the design of effective drugs, in which the primary mechanism of action relies on the modulation of programmed cell death (PCD). Due to the high degree of conservation of basic cellular processes between yeast and higher eukaryotes, and to the existence of an ancestral PCD machinery in yeast, yeasts are an attractive tool for the study of affected pathways that give insights into the mode of action of both antitumour and antifungal drugs. Therefore, we covered some of the leading reports on drug-induced apoptosis in yeast, revealing that in common with mammalian cells, antitumour drugs induce apoptosis through reactive oxygen species (ROS) generation and altered mitochondrial functions. The evidence presented suggests that yeasts may be a powerful model for the screening/development of PCD-directed drugs, overcoming the problem of cellular specificity in the design of antitumour drugs, but also enabling the design of efficient antifungal drugs, targeted to fungal-specific apoptotic regulators that do not have major consequences for human cells.
Collapse
Affiliation(s)
- B Almeida
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal
| | | | | | | | | | | |
Collapse
|
25
|
Jantunen E, Heinonen K, Mahlamäki E, Penttilä K, Kuittinen T, Lehtonen P, Pyörälä M, Hänninen A, Nousiainen T. Secondary acute promyelocytic leukemia: an increasingly common entity. Leuk Lymphoma 2007; 48:190-1. [PMID: 17325866 DOI: 10.1080/10428190600961736] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
26
|
Abstract
Current treatment for acute promyelocytic leukemia (APL) usually includes an induction phase with all-trans retinoic acid (ATRA) and anthracycline-based chemotherapy, followed by a consolidation phase of anthracycline-based chemotherapy and maintenance therapy with ATRA with or without low-dose chemotherapy for 1-2 years. This treatment strategy results in a high complete remission (CR) rate of about 90% and an overall survival rate of 80%. About 5%-30% of patients relapse, mainly patients with high-risk APL. Relapse at extramedullary sites, which occurs in approximately 3%-5% of patients, is emerging as a new issue. Treatment of relapsed/advanced APL includes the use of arsenic trioxide (ATO), gemtuzumab ozogamicin, and hematopoietic stem cell transplantation. ATO is currently the most effective therapeutic agent in relapsed APL. Hematopoietic stem cell transplantation is becoming a common strategy after achieving remission with ATO. Autologous transplant appears to have a more favorable outcome than allogeneic transplant in this setting, particularly when carried out during second remission, primarily because of significantly higher treatment-related mortality with allogeneic transplants. Allogeneic transplant, however, should be strongly considered for patients who remain molecularly positive. Future directions for APL therapy should include developing agents that can prevent relapse, particularly for high-risk patients. Other future treatment strategies may include use of ATO administered concomitantly or sequentially with chemotherapy, gemtuzumab or FLT-3 inhibitors that may obviate the need for autologous transplantation, and posttransplant maintenance perhaps with FLT-3 inhibitors.
Collapse
MESH Headings
- Aminoglycosides/therapeutic use
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/therapeutic use
- Arsenic Trioxide
- Arsenicals/therapeutic use
- Clinical Trials as Topic
- Gemtuzumab
- Hematopoietic Stem Cell Transplantation
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Multicenter Studies as Topic
- Neoplasm Recurrence, Local/drug therapy
- Oxides/therapeutic use
- Transplantation, Autologous
- Transplantation, Homologous
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/genetics
Collapse
Affiliation(s)
- Martin S Tallman
- Northwestern University Feinberg School of Medicine, Division of Hematology-Oncology, Robert H. Lurie Comprehensive Cancer Center, 676 N St. Clair Street, Suite 850, Chicago, IL 60611, USA.
| |
Collapse
|
27
|
Current Awareness in Hematological Oncology. Hematol Oncol 2006. [DOI: 10.1002/hon.753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|