Send Orders for Reprints to
[email protected]
Anti-Cancer Agents in Medicinal Chemistry
1288
Anti-Cancer Agents in Medicinal Chemistry, 2016, 16, 1288-1300
RESEARCH ARTICLE ISSN: 1871-5206 eISSN: 1875-5992
Impact Factor: 2.722
Macromolecular Drug Targets in Cancer Treatment and Thiosemicarbazides as Anticancer Agents The journal for in-depth reviews and high quality research papers on Anti-Cancer Agents
Ş. Güniz Küçükgüzel* and Göknil P. Coşkun Marmara University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Haydarpaşa, 34668, İstanbul-Turkey
ARTICLEHISTORY Received: May 28, 2015 Revised: August 13, 2015 Accepted: September 03, 2015 DOI: 10.2174/18715206166661602191602 56
Abstract: Cancer is known as abnormal cell division and consisting of a group of diseases on various organ tissues. Many therapies are available in cancer treatment such as chemotherapy, radiotherapy etc. Without damaging normal tissue, there is a huge need for specified anticancer drugs which have effect only on abnormal cancer cells. Therefore, advances in anticancer drug discovery in treating cancer in the recent years, directed towards to the macromolecular targets. Heterocyclic molecules, such as fluconazole, acetazolamide, etc., have a significant role in health care and pharmaceutical drug design. Thiosemicarbazides (NH 2-NH-CSNH2) are the simplest hydrazine derivatives of thiocarbamic acid and are not only transition compounds, but they are also very effective organic compounds. Thiosemicarbazides possess an amide and amine protons, carbonyl and thione carbons. These structures have attracted the attention of the researchers in the development of novel compounds with anticonvulsant, antiviral, anti-inflammatory, antibacterial, antimycobacterial, antifungal, antioxidant and anticancer activities. Recently, a number of thiosemicarbazides are available commercially as anticancer drugs for novel anticancer drug discovery. Antineoplastic or anticancer drugs prevent or inhibit the maturation and proliferation of neoplasms. These observations have been guiding the researchers for the development of new thiosemicarbazides that possess anticancer activity.
Keywords: Thiosemicarbazide, hydrazinocarbothioamide, anticancer, apoptosis, caspase-3, isothiocyanate. 1. INTRODUCTION Cancer is known to be the most life threatening disease because of its hard recovery and relapse. Even though it is determined as ‘uncontrolled cell division’, its pathogenesis is still being discovered. Furthermore, understanding the underlying mechanisms and searching for factors that control the metastatic spread of cancer cells are critical in the area of cancer research. As a result, anticancer drugs have different mechanisms of action of their own in terms of healing the disease. Current anticancer drugs are killing the normal cells, as well as they have the ability of killing cancer cells. Therefore, there is a big gap of a ‘targeted’ anticancer drugs. However, as the technology is advancing, new drug targets are identified and considered as new anti-cancer drug targets in order to discriminate the death of normal cells. The role of anticancer drugs is to slow down and hopefully stop the growth and spreading of cancer. There are three possible ways which can be helpful in meeting the goals associated with the use of the most commonlyused anticancer agents. Many studies today have focused on cancer treatment because cancer is the secondary death cause after cardiovascular diseases. Cancer cells have also the ability of developing resistance against current anticancer drugs and therefore, multi-drug therapy or specific targeted therapy is on the line. In addition, the viral and bacterial infections are the most important threats for humanity and these factors can play a significant role in the tumor growth. For example, Hepatitis A Virus (HAV) is a type of picornavirus. It can cause acute self limited hepatitis. Hepatitis C virus (HCV) has been identified in 1989 as the ethiological agent of parenteral non-A non-B hepatitis. HCV often causes liver cirrhosis and hepatocellular carcinoma. Quorum sensing (QS) plays a vital role in both symbiotic bacteriahost and pathogenic interactions. As an example of this; QS is related to the formation of biofilms in Pseudomonas aeruginosa. In
*Address correspondence to this author at the Marmara University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Haydarpaşa 34668 İstanbul, Turkey; Tel: +9 0216 414 29 62; Fax: +90216 345 29 52; E-mail:
[email protected] 1875-5992/16 $58.00+.00
addition, the bacterium is a prevalent pathogen in nosocomial infections and causes mortality in cystic fibrosis (CF). CF is a risk factor in malignant tumor, such as leukemia, colorectal cancer. Helicobacter pylori is a gram-negative pathogen bacteria and it can survive in gastric conditions. H. pylori has the ability to colonize in the highly acidic environment. The bacterium metabolizes urea to ammonia; therefore, it can exist in the stomach. The ammonia surrounds the bacteria and protects it from low pH. Gastric cancer is a highly lethal disease. Several studies have revealed that this bacteria is a vital factor in the development of gastric cancer. Macromolecular drug targets in cancer treatment are known as enzymes, receptors, proteins, nucleic acids such as deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) and also the storage area of the body. 2. ENZYME DRUG TARGETS IN CANCER TREATMENT 2.1. Metalloproteinases (MMPs) Matrix metalloproteinases (MMPs) are type of enzymes that belongs to a calcium-dependent zinc-containing endopeptidase family. MMPs are secreted in normal physiological conditions (i.e. homeostasis). However, MMPs are controlled by hormones, growth factors, and cytokines. MMPs are secreted by a variety of coordinated tissue and pro-inflammatory cells (i.e. endothelial cells, fibroblasts, neutrophils, osteoblasts, macrophages, and lymphocytes). To date, at least 26 human MMPs, which are matrilysins, gelatinases, collagenases, stromelysins, are known. Recent studies show that, MMPs is a target macromolecule in cancer chemotherapy. Many synthetic and natural MMP inhibitors (MMPIs) have been defined as cytostatic and anti-angiogenic agents [1]. 2.2. Methionine Aminopeptidase 2 (MetAP2) Methionine aminopeptidase; cobalt-containing metalloproteases are divided into two families, type 1 (MetAP1) and type 2 (MetAP2). Methionine aminopeptidase 2 (MetAP2) is a metalloenzyme. MetAP2 is found in all organisms and lymphatic tissues. It plays important role in tissue repair and protein degradation which is hydrolytic removal of N-terminal methionine residues from nascent © 2016 Bentham Science Publishers
Macromolecular Drug Targets and Thiosemicarbazides
proteins, in the proliferation of endothelial cells [2]. They also have an important role in certain tumor cells. Angiogenesis can be seen in rheumatoid arthritis, diabetic retinopathy, and cancer. Due to the critical role of these enzymes for angiogenesis, MetAP2 has been one of the major targets in the anticancer drug development area. Fumagillin is an anti-angiogenic agent. It is targeted to MetAP-2. The fumagillin derivatives inhibit MetAP-2 and they can bind irreversibly to the active site of the MetAP-2 enzyme, but they have no relation with MetAP-1 enzyme in terms of binding or inhibition. JNJ-4929821 is an MetAP-2 inhibitor containing triazole structure [3]. 3-Anilino-5-benzylthio-1,2,4-triazole derivatives were identified by Marino et al., [4] as the inhibitors of the human metalloprotease, (MetAP2). In mesothelioma cells the inhibition of MetAP2 expression leads to the cell death. Apoptosis is a programmed cell death. B-cell lymphoma (Bcl-2) is an important protein, which regulates apoptosis in the cell. The regulation of Bcl-2 in colorectal cancer is well studied by various researchers [5]. Inhibition of angiogenesis is also important in cancer therapy and therefore, matrix metallo proteinases gained importance. Other drug targets such as cell cycle regulators, protein kinases, apoptosis modulators, protein farnesyltransferase, aromatase, carbonic anhydrase, histone deacetylase and telomerase are also considered as promising drug targets for the cancer therapy [6]. 2.3. Aromatase Aromatase belongs to cytochrome P450 (CYP) enzyme family and responsible for the synthesis of estrogens. Aromatase does not only exist in ovarians but also exists in periferic tissues and widespread in breast cancer cells. Recently, in breast cancer, aromatase inhibitors (i.e. letrozole, anastrozole, aminoglutethimide, testolactone and formestane) are being used for its treatment. Letrozole and anastrozole contains 1,2,4-triazole structure. Therefore, recently, the researchers studied on 1,2,4-triazoles as aromatase inhibitors. The 1,2,4-triazole is synthesized through thiosemicarbazide in an alkaline medium. Some researchers studied the aromatase enzyme inhibition effect of some azole derived antifungal agents [7]. Recently, researchers are focusing on whether aromatase inhibitors are used only in breast cancer or in another cancer diseases. 2.4. Carbonic Anhydrase The identified carbonic anhydrase isoforms (CAs) in mammals are fourteen. These enyzmes catalyze the chemical interconversion (hydration of CO2 to bicarbonate) at the physiological pH. This chemical interconversion occurs in lipogenesis, biosynthesis of several amino acids, gluconeogenesis, ureagenesis and pyrimidine synthesis. These isozymes are cytosolic (CA I, CA II, CA III, CA VII) enzymes. CA V exist only in mitochondria, and CA VI is excreted in saliva. The other CAs, (CA IV, CA IX, CA XII and CA XIV) are membrane-bound. CA IX and CA XII isoforms of the enzyme are correlated with tumors. Scozzafava et al. reported sulfonamides and these derivatives as carbonic anhydrase inhibitor [8]. 2.5. Farnesyltransferase (FTase) A heterodimeric metalloenzyme, farnesyltransferase (FTase) activity depends on zinc and magnesium, which is necessary for the chelation of the target protein’s farnesyl-accepting cysteine and the enzyme’s active site. The duty of FTase is to add 15-carbon isoprenyl farnesyl moiety. Ftase has an important duty in the transfer of farnesyl group to cysteinthiol of ‘CAAX’ c-terminal part in Gprotein which leads to G-protein activation [9-10]. Farnesylation leads Ras protein’s membrane localization. Later, this protein defines the exchange from an inactive to an active Ras-GTP-bound form. The three isoforms of Ras are known as K-ras, N-ras and
Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10 1289
H-ras. The mutations of the K-ras isoform are the most related ones in human cancers (i.e. colon, pancreatic, and lung cancers, which is related with K-ras mutations in the incidence of 40, 90, and 25%, respectively) [11]. FTases plays a key role in developing new anticancer agents because it can also attach the prenyl-moiety to Ras protein’s C-terminal cycteine. Ras protein is also found to be mutated in many human cancer types. On the light of foregoing, FTase inhibitors are promising drug candidates in cancer treatment and this enzyme is a possible anticancer drug target in the research area. Tipifarnib, Lonafarnib, BMS-214662 and L778123 was the first FTI tested in a clinical trial. These compounds have different toxicity profiles. 2.6. Glycogen Synthase Kinase (GSK-3b) The addition of phosphate molecules onto serine and threonine amino acid residues is mediated by serine/threonine protein kinase. A serine/threonine protein kinase, GSK-3, is responsible for regulating the glycogen metabolism. GSK-3 plays significant roles in various main intracellular stimulating pathways. Some main intracellular stimulating pathways are glucose regulation, cellular proliferation, immune responses, migration, apoptosis and inflammation. By the discovery of the GSK-3 duty in various cellular processes, GSK-3 becomes a target of interest for cancer treatment. Two known genes which have two isomers, encode GSK-3 and these isomers are GSK-3 alpha (GSK3a) and GSK-3 beta (GSK3b) which downregulates post-translational phosphorylation of Ser-9 of N-terminal domain. In a way, GSK-3b suppress the tumor by down-regulating numerous proto-onco-proteins [9]. GSK-3 was shown to induce apoptosis in the cell, including DNA damage and hypoxia. GSK-3 was found to expose a response to these stimulators through the regulation of transcription factors, some of which are p53 and heat shock factor-1 [12]. 2.7. Histone Deacetylases Histone tails contain amine groups from lysine and arginine amino acids and as a result of that, they are positively charged. The positive charges of those functional groups, provide the binding of histone tails and the DNA backbone. Normal cells have the ability of acetylation. This acetylation, changes amines into amides and the positive charges on the histone is being neutralized. This situation works against the binding affinity of histones and DNA. Chromatin growth occurs with the low binding affinity and this situation leads to generic transcription. The removal of the acetyl groups from an ε-N-acetyl lysine amino acid on a histone is being done by histone deacetylases. Acetyl groups are eliminated by histone deacetylase and the positive charge of the histone tails are increased. The elimination provides a perfect connection between the histones and DNA backbone. In a series of pathways within the living system; signal transduction; notch signaling pathway, cell cycle and importantly cancer, histone deacetylase is involved. In tumor pathogenesis, usual situation in human cancers are the decreased levels of acetylated Lys16 (K16-H4) and trimethylated Lys20 (K20-H4) of histone H4. The decrease in histone acetylation can be observed in tumorogenesis, tumor invasion and metastasis. Histone deacetylases therefore, were informed to be over expressed in many cancer types (i.e. prostate, gastric, breast, colon and cervical) [13]. Today, histone deacetylase inhibitors are popular in cancer treatment and considered to be a promising macromolecule to aim. 2.8. Tyrosine Kinase The duty of tyrosine kinase is to transfer a phosphate group, which exist in ATP, to a protein. These enzymes control the target protein function via transferring the phosphate group in ATP to the tyrosine’s hydroxyl group. This transfer, up-regulates or down-
1290 Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10
regulates diverse cellular functions. Tyrosine kinases are a family of protein kinase and they transfer phosphate groups to serine and threonine. Phosphorylation of a protein is significant in cell mechanisms, importantly in cell division. Tyrosine kinases and tyrosine phosphatases regulate the activity degree of cellular tyrosine kinase phosphorylation by their antagonizing activity. Today, tyrosine kinases have gained reputation as a macromolecular target in cancer treatment because of the tremendous effect in molecular cancer pathogenesis. Thus, some anticancer drugs (such as imatinib) were licensed on the market. While cancer pathogenesis is being enlightened, many of the tyrosine kinases are reported to have correlation with oncogenes or tumor suppressor which are epidemiologically relevant. Therefore, tyrosine kinase receptors are also great drug targets in cancer drug development [14] and tyrosine kinases were found to have an important role in different cancer types like osteosarcoma [15], non-small cell lung cancer [16] and prostate cancers [17]. 2.9. Telomerase Telomerase also called telomere terminal transferase and telomeres are located at the ends of chromosomes. It is a ribonucleoprotein and a cellular reverse transcriptase that adds new DNA on the telomeres [18]. The damage response in the cell which is regulated by p53 is triggered by few critical short telomeres. After that, cells enter M1 phase. In this M1 phase, cells have no ability to divide more and they are metabolically active. If the decrease in p53 function in the control points are damaged, cells can divide till their ‘crisis’. This is a period where cell growth and death are in balance. In other words, the cell can go to M2 phase. In this phase, telomeres become extremely short and illimitable chromosomal end joining and different situations produce an effectively unstable genome, at the end, the cells become apoptotic [19]. Telomerases gained importance in different cancer types such as non-small cell lung cancer, breast cancer, multiple myeloma and chronic lymphocytic leukemia. Over the passed years, numerous approaches have been published in terms of telomerase-based anticancer drugs such as telomerase inhibitors targeting hTERT (an important protein in telomerase biology) or hTR (telomerase RNA component), telomere-disrupting compounds, hTERT promoterdriven gene therapy, hTERT immunotherapy as well as inhibiting telomerase secretion or biogenesis. Its current inhibitor imetelstat was recently studied against esophageal cancer [20]. Imetelstat administratin in esophageal cancer cell resulted the inhibiton of telomerase activity and therefore decreased the growth of the cancer cells. 2.10. Thymidylate Synthase (TS) Thymidylate synthase (TS) is a significant and essential enzyme for DNA replication, transcription and the renewal of the DNA. TS uses 5,10-methylenetetrahydrofolate to convert deoxythymidine monophosphate (dTMP) from deoxyuridine monophosphate (dUMP) and forms thymine which is a nucleic acid in DNA [21]. Thymidylate synthase (TS) has essential duty in early steps of DNA biosynthesis. For this reason, this enzyme is a critical target in cancer chemotherapy. Although a known TS inhibitor, 5-fluorouracil (5-FU), which is on the market as anticancer drug, new targets are on the study field in order to over-come drug resistance in cancer therapy. A reverse correlation with the level of TS activity in tumor cells and 5-FU sensitivity have been shown in several preclinical in vitro and in vivo studies [22]. 2.11. Topoisomerase DNA topoisomerases are one of the established molecular drug targets for the evolution of anticancer drugs. They have a
Küçükgüzel and Coşkun
significant role in solving the DNA topological obstacles in DNA replication. They can do this by their activity of abruptly breaking and rejoining of one or two strands of DNA. The human DNA topoisomerases have two types known as topoisomerase I (topo I) and topoisomerase II (topo II). To date of its discovery, topoisomerase inhibitors such as camptothecin, etoposide, and teniposide are being used as anticancer agents in cancer treatment [23]. In cancer pathology, topoisomerase II was found to be responsible in cancer disease and became a macromolecule to target. The enzyme topoisomerase II regulates chromatin topology by operating a breakage-reunion cycle. This reunion cycle involves in formation of an intermediate, which is called cleavable complex, in which each subunit of the topo II homodimer is covalently linked to the newly created 50 phosphate ends [24]. Therefore, topoisopmerase II has an essential role in cancer treatment with different cancer diseases. 2.12. Proteasome In the cytosol and nucleus of all eukaryotic cells, the proteasome pathway is the main proteolytic system. The 26S proteasome, which is known as the proteasome, is an enzyme complex that exists in the nucleus and cytoplasm of all eukaryotic cells. The duty of proteasome is to break peptide bonds by proteolysis and degrade the proteins. The substrates of proteasome are transcription factors, tumor suppressors, signaling molecules, cell-cycle regulators, antiapoptotic proteins (e.g., Bcl-2) and inhibitory molecules (whose degradation activate other proteins), among others. If these proteins’ degredation collapses, the result is eventually massive. Proteosome has a significant role in drug resistance development, inhibition of chemotherapy-induced apoptosis and cancer cell proliferation. Therefore, it is considered to be a potential anticancer drug target to accomplish the anticancer drug resistance. In fact, the programmed cell death, which is known as apoptosis and leads to accumulation of p53 protein which promotes effective tumor suppression, is a result of the inhibition of proteasome function [25]. Current registered anticancer drug bortezomib is known to be a proteasome inhibitor in the cancer treatment. Bortezomib still has toxic effects to patients and there is a need of less toxic proteasome inhibitors even if there are some natural compounds proved to be proteasome inhibitors such as green tea polyphenols, curcumin, genistein, and resveratrol. Although the natural compounds are found to be less toxic than bortezomib, the cancer therapy still need a combination of lower dose bortezomib and these diatery polyphenols together. Therefore, a synthetic protesome inhibitor, may have the potential of being new anticancer drug [26]. 2.13. Urease Ureases are a class of enzyme family and they exist in bacteria, algae, fungi, plants and invertebrates. Even though the ureases have diverse protein structures, they perform a basic catalytic reaction. The hydrolysis of urea is the catalytic function. After the hydrolysis, carbonic acid and ammonia occur [27]. Ureases play different roles in multiple metabolic pathways, i.e. purine metabolism, arginine and proline metabolism, urea degradation, atrazine degradation and metabolism of carbamide intermediates. Ureases are also a group of enzymes that require nickel for the activity. It is also very hard for the enzyme to get nickel ions from the active side as it has a tight binding system. Recent studies were focused on the inactivation of urease because of the following reasons; 1) Urease is a unique enzyme which presents in H. pylori and is a need for the bacteria for its survival in acidic conditions. 2) H. pylori infections in the gastric flora causes gastric ulceration and lead gastric carcinoma. For those reasons, urease is considered to be a promising target in gastric carcinoma. In cancer treatment, it is very important to prevent the disease, even before its forming. Therefore, ulcerations
Macromolecular Drug Targets and Thiosemicarbazides
causing by H. pylori infections are important as these infections can cause cancer if it prolongs. 1,2,4-triazole compounds are found to be promising compounds for urease inhibition and studies recently focused on the inhibition effects of triazole derivatives [28-30]. 3. THE ROLE OF PROTEINS AND RECEPTORS IN CANCER TREATMENT 3.1. Hepatocyte Growth Factor (HGF) Hepatocyte Growth Factor (HGF) has a significant duty in wound healing, organ regeneration and embryonic organ development. HGF modulates morphogenesis, cell motility and cell growth and activates tyrosine kinase cascades. This protein and its receptor are believed to be an important target for anticancer drug therapy by Martin et al. [31] HGF is a dual player in the complex biology of cancer development and progression. HGF acts directly on and stimulates cancer cells. HGF also acts as an angiogenic factor and lymphangiogenic factor that aid the growth and spread of cancer cells. Besides this protein, a group of growth factors, (i.e., vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor) have become an interest on anticancer drug targets. 3.2. Breast Cancer 1 Protein (BRCA1) Breast cancer 1 protein (BRCA1) is reported to be a tumor suppressor in breast cancer development. Inactivation or hypermethylation of this protein damages the replication of DNA synthesis and increasing the risk of breast cancer. Romagnolo et al. [32] suggested this protein as a target macromolecule for the prevention and treatment of breast cancer. 3.3. Human Pregnane X Receptor (hPXR) The main transcription agent for CYP 3A4 and multidrug resistance protein 1 (MDR1) are known as human pregnane X receptor (hPXR). Some different ligands can activate the hPXR. Human pregnane X receptor can go under the name of (SXR) steroid X receptor, which is a member of nuclear receptor subfamily 1, group 1, member 2, (NR1I2). The receptor (hPXR) is exceedingly present in the intestine and the liver, and considerable amount exist in lungs and kidney. The hPXR is related with different types of cancers such as Barrett’s oesophagus, prostate, colorectal, colon, endometrial, breast, ovarian, oesophageal squamous carcinomas and osteosarcoma. The uncontrolled secretion of hPXR is extremely exist in neoplastic cells, even though the receptor hPXR is present in all cell of the body. hPXR is a hopeful anticancer drug target because of its importance and also it is believed, it can help to avoid the multidrug resistance. Developing new hPXR inhibitors is a way of finding new cancer treatment [33]. 3.4. Hypoxia-Inducible Factor (HIF-1a) Hypoxia-inducible factor (HIF-1a) is a heterodimer of bHLHPAS (basic HLH (helix-loop-helix)-PER-ARNT-SIM) proteins. HIF-1a mediates transcriptional activation of erythropoietin (EPO), angiogenic vascular endothelial growth factor (VEGF) and other genes to induce enhancement of oxygen delivery to cell. But overexpression or dysregulation of HIF-1a is highly connected to cancer biology and leads to proliferation of tumor cells and mediates drug resistance. Hence, HIF-1a inhibition can be vital for cancer therapy and HIF-1a is also a target compound for cancer treatment [9]. 3.5. Vascular Endothelial Growth Factor (VEGF) Vascular endothelial growth factor (VEGF) is a class of growth factor that belongs to the platelet derived growth factor superfamily. VEGF consists of several glycoproteins defined as VEGF-A, VEGF-B, VEGF-C, VEGF-D, and VEGF-E and the placental growth factor. Vascular endothelial growth factor–A, normally referred to simply as VEGF, is a dimeric glycoprotein of 34 to 45
Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10 1291
kDa that promotes angiogenesis by stimulating differentiation, migration of vascular endothelial cells and proliferation. VEGF is secreted by cells and it is a signal protein. These cells can trigger angiogenesis and vasculogenesis. VEGF's normal function is to create new blood vessels during embryonic development. Besides, it creates new vessels for muscles after exercise, new blood vessels after injury, and new vessels to bypass blocked vessels. VEGF expression is high during embryonic stages and is believed to play crucial role in vascular development (vasculogenesis). It is also reported to play significant role in generating new blood vessels in case of injury and heart block. In some solid tumors, over-expression of VEGF leads to enhanced tumor growth and metastasis. The tumor growth and metastasis can occur due to increased availability of nutrient renewal in the metabolizing cell [34]. Therefore, VEGF helps to expand the metastatic ability of the cancer cell. A pro-angiogenic cytocine, VEGF, simulates angiogenesis, a cellular modification which is essential for metastasis and tumor growth. The VEGF family protein’s angiogenic activity is regulated by three VEGFR receptors (VEGFR-1, VEGFR-2, and VEGFR-3) which are present on endothelial cells. The VEGFR-2 receptor, which is an essential kinase that takes role in various processes of angiogenesis by signaling in endothelial cells; including survival, endothelial cell proliferation, differentiation, vascular permeability, and migration. Thus, interruption of VEGFR-2 downstream signaling pathways become an important cancer target. Recently, preclinical and clinical studies show promising results of many small molecule inhibitors [21]. 3.6 Epidermal Growth Factor and Epidermal Growth Factor Receptor (EGF and EGFR) Epidermal growth factor (EGF) is a growth factor that regulates proliferation, cell growth and differentiation by binding to its receptor, epidermal growth factor receptor (EGFR). The 6045-Da protein, human EGF, consist three intramolecular disulfide bonds and 53 amino acid residues. Many different growth factors and growth factor receptors have been proven to take part in the independent evolution of cancer cells. The EGF-family of peptide growth factor and the EGFR have an essential duty in the progression of different cancer types and pathogenesis [35]. The ErbB or EGF receptors are members of protein tyrosine kinase (PTK) family. There are four ErbB receptor family members represents; ErbB1 (EGFR, HER1), ErbB2 (HER2/neu), ErbB3 (HER3), ErbB4 (HER4). These trans-membrane proteins can be triggered by binding with peptide growth factors of the EGF family of proteins [21]. EGFR function abnormalities are related with all crucial properties of cancer development, including invasion, autonomous cell proliferation, metastatic and angiogenic potential [36]. The number of EGFR is generally over-expressed in cancer cells rather than normal healthy cells. EGFR, is over secreted in human neoplastic cells i.e., glioblastoma, head and neck squamous cell carcinoma (HNSCC), breast, bladder, colorectal (CRC), ovarian and prostate carcinomas [36]. Therefore, inhibition of EGFR signaling has antiproliferative and therapeutic effects in cancer treatment as we described in this review, a thiosemicarbazide compound has the ability to inhibit EGFR [37]. 3.7. Tubulin Polymerization The large, hollow chambers of tubulin dimmers are called the microtubules. Microtubules are cytoskeleton component and polymeric proteins formed by reversible polymerization of a and b heterodimer tubulin. Tubulin is globular protein and indicates five definite families, the alpha (α), beta (β), gamma (γ), delta (δ), epsilon (ε) and zeta (ζ) tubulin. The α-tubulin and β-tubulin are the most important members of the tubulin family. They build the microtubule. This dynamic assembly/disassembly of microtubules mediates many eukaryotic cell functions like cell division,
1292 Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10
migration, shape alteration etc. and involves in a variety of fundamental cell routines like intracellular transports, cell signaling and apoptosis [38]. Tubulin is an essential eukaryotic protein that has a significant duty in cell division. Today, microtubule-targeted drugs are gaining importance in the therapy of different cancer types. The formation of microtubules is a vigorous process that involves the polymerization and depolymerization of tubulin heterodimers. Compounds that interfere with the microtubulin equilibrium in cells are useful in the treatment of human cancer. They interfere with this dynamic equilibrium by binding to tubulins and induce cell cycle arrest, resulting in cell death [39]. This is one of the reasons why microtubules are an attractive molecular target for anticancer agents. Numerous alkaloids, taxoids and natural products have been identified that binds to tubulin dimmers to seize microtubule assembly-disassembly process as target of interest for curing cancer [9]. 4. ROLE OF THIOSEMICARBAZIDES IN ANTI-CANCER ACTIVITY Thiosemicarbazides have been demonstrated to possess anticonvulsant [40-43], antiviral [44,45], hepatitis-C NS5B polymerase inhibitors [46], anti-inflammatory [47,48], antimalarial [49], antibacterial [50-59], antimycobacterial [45, 60-62], antioxidant [63], antifungal [64-68] and anticancer activities. Thiosemicarbazides have also taken the attention of the researchers because of their various biological activities [69,70] and considered to be promising compounds for different diseases. Isothiocyanates are inartificial chemical compounds found in usable cruciferous plants. In addition, Isothiocyanates which are used for the synthesis of thiosemicarbazides, possess vital importance. Phenyl isothiocyanate is used to determine amino acids and known as Edman reagent. Anticancer activities were reported for phenyl isothiocyanate and some other isothiocyanates and their efficacy of preventing cancer was studied. To date, studies have shown that, isothiocyanates have antineoplastic activity. They can inhibit the growth of different types of human cancer cells. Wu et al. [71] investigated in vitro anticancer activity of benzyl isothiocyanate (Fig. 1) and phenethyl isothiocyanate (Fig. 2) on highly metastatic human lung cancer L9981 cells, these compounds possessed activity with the IC50 value of 5.0 and 9.7 μM, respectively. Therefore, thiosemicarbazides, which are synthesized from isothiocyanates, have taken the attention of the researchers and their antitumor activity was reported. Thiosemicarbazides, containing an amide and amine protons, carbonyl and thione carbons are synthesized by heating the substituted hydrazines/hydrazides with isothiocyanates (Fig. 3) in the presence of solvents like ethanol [72,73], tetrahydrofuran or pyridine [74], dioxane [75], isopropanol [76], dimethylformamide [77] and toluene [78]. Thiosemicarbazides can be nomenclatured as 1-alkyl/aryl-4-alkyl/aryl-thiosemicarbazide or hydrazinocarbothioamide, [(substitutedamino)thioxomethyl]acid hydrazide. Published and known preparations of thiosemicarbazides include reactions of isothiocyanates with hydrazines, but isothiocyanates
N
C
S
Fig. (1). Benzyl isothiocyanate.
N C S
Fig. (2). Phenethyl isothiocyanate.
Küçükgüzel and Coşkun R O
NH
NH2
1
+
R (Ar)
N S
O R
NH
NH
NH S
1
R (Ar)
Fig. (3). General Synthesis of thiosemicarbazides.
are difficult to handle and store. Reduction of thiosemicarbazones by sodium borohydride is used for the preparation of thiosemicarbazide and only applicable if thiosemicarbazone is mono-, di-, and trisubstituted (but not tetra- or pentasubstituted). Another method for the synthesis of thiosemicarbazide is the reactions of hydrazines with reactive thiocarbamic acid derivatives; however, they are affected by side reactions. Reactions of cyanohydrazines with hydrogen sulfide can yield mono or disubstituted thiosemicarbazides. Last of all, condensation of 1,2,4-triazolyl or bis(imidazolyl) methanethiones with amines then with hydrazines gives di- and trisubstituted thiosemicarbazides [79]. The thiosemicarbazides are not only transition compounds, but they are also very effective organic compounds. Thiosemicarbazides are reactive agents and they are used for the synthesis of 1,3,4oxadiazoles with the presence of potassium iodide [73]. 4-thiazolidones are synthesized with the presence of thiosemicarbazides and absolute ethanol-anhydrous sodium acetate alkyl α-halogenoacetate [45, 72]. Thiosemicarbazides can be transferred to 1,3,4-thiadiazoles with the presence of sulphuric acid [80] and they can also be cyclisized to 1,2,4-triazoles in alkaline conditions [73] (Fig. 4). Recent studies have focused on metal complexes of thiosemicarbazides as they have an effect on the cancer research area. The researchers synthesized their metal complexes and evaluated these complexes for their anticancer effect. The anticancer effect mechanism of metal complexes is still not yet fully discovered, but they are considered to be promising and safer anticancer drugs for the future [81]. A new ligand N-salicyloyl-N´´-(p-hydroxybenzthioyl) hydrazine and its copper (II) complex were synthesized and in vivo anticancer activity of copper (II) complex of thiosemicarbazide has been studied against breast tumor in C3H/J strain mice and in vitro on murine mastocytoma (P-815) and human erythroleukemia (K-562) cells by Singh et al. [82]. The group later on reported anticancer mechanism and activity of the copper (II) 1,4-dibenzoyl-3thiosemicarbazide complexes. The higher inhibition of thymidine joining in Jurket and Daltons lymphoma tumor cells than metal complexes of the ligand was showed. In addition, anticancer activity of the copper (II) 1,4-dibenzoyl-3-thiosemicarbazide depends on apoptosis on the assumption of the ligand, whereas the anticancer activity of the copper (II) 1,4-dibenzoyl-3-thiosemicarbazide complexes, is not related with apoptosis. It is clear that the metal complexes stop cellular growth by binding with DNA [83]. N-salicyloyl-N´-o-hydroxythiobenzhydrazide and its Mn(II), Cu(II) ,Ni(II) Fe(III), Co(II), Zn(II) complexes were prepared by Shrivastav et al. [84] Mn(II), Cu(II) and Ni(II) complexes were detected to inhibit the growth of tumor in vitro, whereas, Fe(III), Co(II), Zn(II) complexes did not. Tumor bearing mice was implemented with Mn(II), Cu(II) and Ni(II) complexes exhibited reversion of tumor growth correlated with the trigger of apoptosis in lymphocytes. This study suggests that Mn and Cu complexes helps to extend the survival in tumor bearing animals by straightly inhibiting tumor cells and changing tumor correlated immunosuppression.
Macromolecular Drug Targets and Thiosemicarbazides
Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10 1293 R
N
NH
NaOH
O
N O
I2 / KI NH
R
NH
NH
R
1. OH
1
H N
-
+
S
N
1
CH 3COONa X
R
R
1
H 2SO 4
CH 2COOR
O
1
N
S
2. H
N
N
N
R
1
NH S
S
Fig. (4). Synthesis of thiosemicarbazides derivatives.
El-Metwally et al. [85] studied the biological activities of some new metal complexes of thiosemicarbazide. The group synthesized Vo(II), Ni(II), Pd(II) and Cu(II) complexes of 1-(4-nitrophenyl)4-methly thiosemicarbazide. Pd(II) and Ni(II) complex of thiosemicarbazide exhibited DNA degradation and it does not depend on the length of contact time with the complex. These complexes can block the division of DNA in cancer cells and they can be considered as new anticancer agents in the treatment (Figs. 5, 6). O N
+
O
synthesized Pd (II) compounds are more cytotoxic for the LM3 cell line than cisplatin, exhibiting IC50 values between 2.79–8.84 µM whereas cisplatin was changing between 30.3 µM for LM3 and 4.3 µM for LP07. MTT studies proved that all complexes show better cytotoxic activity, exceeding that of cisplatin against the mammary adenocarcinoma LM3.
-
NH +
S
HN N
HN
+
H
Ni
S
N
H
NH
S
X
Ph3P
X: Br, I, SCN
Fig. (7).
NH NH
N
An interesting study comes to the light about the DNA cleavage of metal complexes of thiosemicarbazides. Mlahi et al. [87] studied the possible biological activity and of course, the synthesis of binary and ternary complexes derived from 4-allyl-1-(2-hydroxybenzoyl) thiosemicarbazide. Among the synthesized derivatives, two complexes and the ligand (thiosemicarbazide) were tested against DNA using agarose gel electrophoresis. Cu (II) complex (Fig. 8) showed significant effect on DNA damage.
Fig. (5). O N
+
O
-
O
N
HN
Pd
NH
NH
N NH
Pd
S
S
S
NH N
Cu
O
NH
OH 1/2
Fig. (6).
Cisplatin is now on the market with the indication of cancer treatment in many countries. Since its foundation by Rosenberg, platinum based drugs or possible other complexes gained importance. However, platinum based drugs have severe neurotoxicity and nephrotoxicity. Therefore, triphenylphosphane and Palladium (II) (which has structural and thermodynamic similarities to platinum) complexes of 4-phenyl-3-thiosemicarbazide were synthesized (Fig. 7), characterized and cytotoxic activities were investigated by Rocha et al. [86] Substitutions were prepared as chloro, bromo, iodo and -SC≡N. The in vitro growth inhibitory effect (IC50) of complexes chloroand -S-C≡N after 24 h by using the MTT assay were compared with that of cisplatin for two murine cancer cell lines, LM3 (mammary adenocarcinoma) and LP07 (lung adenocarcinoma). All the
Fig. (8).
Thiosemicarbazides possess diverse biological activities and they are active reagents for the synthesis of new heterocyclic compounds [69]. Some new thiosemicarbazide derivatives synthesized recently and their anticancer activities have been studied. Compound R-253 (Fig. 9), N-cyclopropyl-2-[6-(3,5-dimethylphenyl) thieno[3,2-d]pyrimidin-4-yl]hydrazine carbothioamide, was synthesized by Gururaja et al. [88] and evaluated for its inhibition of various tumor cells correlating microtubule networks. Microtubule networks have a significant duty in cancer development and as a target, this pathway is responsible for over expressed cell division. Inhibition of this pathway allows to discriminate only the cancer cells rather than the normal cells in the human body. R-253, specifically disrupts the microtubule network in cancer cells and
1294 Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10
Küçükgüzel and Coşkun
has less toxicity on normal dividing cells. Its anticancer effect is 20 nmol/l with EC50 value. Therefore, R-253 can be considered as an important new tubulin binding drug candidate for cancer treatment (Fig. 9).
N
N
HS O
S
NH
NH2
NH
Fig. (12). N
N
HS O
S
S
S
NH
NH NH N
NH
NH
NH2
Br
Fig. (13).
N
Fig. (9).
Al-Saadi et al. [89], investigated anticancer activities of some 2,3,5-trisubstituted thiazole derivatives and found anticancer activity of intermediate thiosemicarbazide derivative compound, 4fluorophenyl-1-[2-(N-4-fluorophenylthioureido)-4-methyl thiazole5-carbonyl]thiosemicarbazide (Fig. 10). In this study, the researchers have found that 4-fluorophenyl-1-[2-(N-4-fluorophenylthioureido)4-methyl thiazole-5-carbonyl] thiosemicarbazide is most effective against non-small lung cancer (HOP-92) and melanoma (SK-MEL-2) cell lines in vitro.
Zhang et al. [37] investigated the anticancer activity of new chalcone thiosemicarbazide molecules as the inhibitors of EGFR kinase. Among the tested compounds, compound bearing para methyl substitution on B ring (Fig. 14) have a strong inhibition activity to both EGFR and HepG2 cells with IC50 values of 0.35 and 0.78 µM respectively. The docking studies was also studied to place the active compound into the EGFR active site to discover the possible binding model. The mechanism of the anticancer activity of the tested compound was studied with Annexin-V assay. The results indicated that, chalcone thiosemicarbazide induced apoptosis in HepG2 cells. NH2
S
F S
O
N
F NH
NH
N
S
NH
NH HN
A
NH
B
S
Fig. (10).
Fig. (14).
The formation of N1-[2-(5,5-Dimethyl-3-oxocyclohex-1-enylamino) acetyl]-N4-phenyl thiosemicarbazide was refluxed in ethanol with substituted hydrazide and phenyl isothiocyanate. The synthesized compounds were evaluated in vitro for their anticancer activity. Using MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay, the anticancer effects of the new compounds were studied on hepatoma cell line (HepG2) at 10 mmol/ml. N1 -[2-(5,5Dimethyl-3-oxocyclohex-1-enylamino)acetyl]-N4-phenylthiosemicarbazide (Fig. 11) exhibited a better in vitro anticancer activity against hepatoma cell line (HepG2) [90].
Nofal et al. [92], synthesized new compounds with benzimidazole and hydroxylamine hydrochloride, ethyl cyanoacetate, guanidinium sulfate, thiourea, phenylhydrazine, methylhydrazine and/or hydrogen peroxide; giving different derivatives of benzimidazole. Their activity were investigated against HepG2 and neuron PC12 cancer cell lines. (2-(3-Cyano-4-(3,4,5-trimethoxyphenyl)-6-(1-methyl-1Hbenzo[d]imidazole-2-yl) pyridin-2-yloxy)acetyl)-4-methylthiosemicarbazide and (2-(3-cyano-4-(3,4,5-trimethoxyphenyl)-6-(1-methyl1H-benzo[d]imidazole-2-yl)pyridin-2-yloxy)acetyl)-4-phenylthiosemicarbazide (Figs. 15, 16) were proven to be the most active compounds against PC12 cancer cell line with IC50 value of 0.251 and 1.5 µM respectively. The result obtained from the study of the PC12 activities revealed that methyl derivative showed the most prompt activity in PC12 cell line due to the presence of sulfur atoms in the attached side chains that enhances the anticancer spectrum. The variation of the anticancer spectrum of activity between the two closely related isothiocyanate derivatives indicates that the cytotoxicity can be obtained in this class of compounds by attaching small alkyl group into thiosemicarbazide side chain (methyl derivative), rather than the larger aryl group (phenyl derivative).
O S NH NH
NH
NH O
Fig. (11).
Seven Stemazole derivatives were synthesized and characterized by Sun et al. [91]. The preliminary SAR studies proved that Stemazole (Fig. 12) (4-(4-(5-mercapto-1,3,4-oxadiazole-2-yl)phenyl) thiosemicarbazide) and Br-Stemazole (Fig. 13) (4-(2-bromo-4-(5mercapto-1,3,4-oxadiazole-2-yl)phenyl)thiosemicarbazide) possessed significant stem/progenitor cell proliferation-promoting activity. In this study, Sun et al. reported that Stemazole increased human HSCs expansion in a time-dependent and dose-dependent manner and was the strongest stem/progenitor cell proliferation activator among tested compounds. In addition, the studies propose that Stemazole is a new and effective main activator of stem/progenitor cell expansion and has an essential importance in the survival and development of various types of stem/progenitor cells in vitro.
O O
O
N S NH
N
N N
Fig. (15).
O
NH O
NH
Macromolecular Drug Targets and Thiosemicarbazides
Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10 1295
Siwek et al. [95] investigated the anticancer activity of 4ethoxycarbonylmethyl-1-(piperidin-4-yl-carbonyl)thiosemicarbazide (Fig. 20), a potent topoisomerase II inhibitor. From the docking studies, the inhibitory action is associated with the ATP binding pocket, however competitive assays are still needed to confirm this. This compound, then tested for anticancer activity using MTT assay and 4-ethoxycarbonylmethyl-1-(piperidin-4-yl-carbonyl)thiosemicarbazide decreased in both estrogen-receptor positive breast cancer (MCF-7) and estrogen-receptor negative breast cancer (MDA-MB-23). The IC50 values of the compound on the cell lines were 146 ±2 µM and 132 ±2 µM respectively.
O O
O
N S NH
N
N
O
NH
N
NH
O
Fig. (16). HN
A new series of thiosemicarbazide derivatives as anticancer drugs were prepared via 4-(2-pyridyl)-3-thiosemicarbazide with phenyl isothiocyanate, benzoyl isothiocyanate, phenyl isocyanate and 4-pyridyl isothiocyanate by Yousef et al. [93]. All compounds were tested for their anticancer activity against Erlich Ascides Carcinoma (EAC) (in vitro) bearing rat were found to increase the life span. 1-(amino-N-(pyridin-2-yl)methanethiol-4-(pyridin-2-yl) thiosemicarbazide (Fig. 17) has remarkably decreased the viable ascitic cell count as indicated by trypan blue dye exclusion assay. The obtained data of the tested compound exhibited minimal adverse effects on the treated animals as compared to those of the untreated control groups. The two pyridine moieties and two SH groups in this compound significantly enhance the anticancer activity. Therefore, a new way of chemotherapy can be Fe chelation therapy, particilary in consequence of the increase of resistance to well-known anticancer drugs and due to the presence of two SH groups which show a high chelation power towards Fe rather than oxygen. S NH N
NH
NH
S
HCl NH
O NH
NH
O
O
Fig. (20).
Koca et al. [96] studied the cell viability of different cancer cell lines of new thiosemicarbazide derivatives, carrying pyrazole moiety. Containing pyrazole ring thiosemicarbazide derivatives were synthesized through one pot reaction of 4-benzoyl-1,5-diphenyl1H-pyrazole-3-carbonyl chloride with ammonium thiocyanate and various amines. Anticancer activities of containing pyrazole ring thiosemicarbazide derivatives were studied on human colon, liver and leukemia cancer cell lines. Compounds bearing 4-clorophenyl, 4-fluorophenyl and 4-bromophenly substitution increased the cell viability up to 100 % in 24 hours. Compound bearing only phenyl ring with no halogen substitution, showed higher activity against leukemia cancer cells than the halogen substituted compounds. However, halogen substituted compounds (Figs. 21, 22) have similar anticancer activity against human colon, liver and leukemia cancer cell lines.
NH
O
O
NH
S
N
NH
Fig. (17).
S
N N
He et al. [94], synthesized new quinazoline thiosemicarbazide derivatives and tested their anticancer activities in vitro against oral, nasopharyngeal, gastric, breast and lung carcinoma human cancer cell lines. Among the tested compounds, all of them (Figs. 18, 19) showed remarkable activity and from the structure-activity relationship results, the group enlightened that nonsubstituted quinazoline and benzene rings or chloro or fluoro substituted benzene ring in these derivatives would be the most favorable for their anticancer activity.
F
Fig. (21). O
O NH NH N
O
S
S
N Br
NH NH
NH
R1 N
N
Fig. (22).
R1: 2-Cl, 3-Cl, 4-Cl, 2-F, 4-F
Fig. (18). H3C
H3C
O
O
S NH N
NH
O NH
N
R2: 2-Cl, 3-Cl, 4-Cl, 2-F, 4-F
Fig. (19).
R2
Indolamine-2,3-dioxygenase (IDO), a promising macromolecular drug target for anticancer immunotherapy, was investigated for its inhibitors by Serra et al. [76] The new synthesized thiosemicarbazides showed higher inhibition than the known inhibitor, benzo[d]thiazole-2(3H)thione (Fig. 23) derivatives. Structure activity relationship studies showed that, substitution in the m- and p-position relative to the phenyl thiosemicarbazide are very promising, however, o-substitution always decreases the activity. A cyano substitution (Fig. 24) on the phenyl ring showed the highest inhibition with the IC50 value of 1.2 µM.
1296 Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10
Küçükgüzel and Coşkun
showed no activity. However, when alkyl substitution was replaced to ethyl ester, compounds showed activity between 1.6×10-3 and 9.3 µM IC50 value.
S S N H
O
R
Fig. (23). NH
NH NH2
R
NH
1
S
S
O NH
N
NH
NH
R
2
S
N
MDA-MB-231 and HeLa; R:CH3, R1: COOC2H5, R2: C2H5
Fig. (24). Fig. (27).
Malki et al. synthesized novel thiosemicarbazides and 1,3,4oxadiazoles derived from thiosemicarbazides to study their anticancer activity on human MCF-7 breast cancer cell lines [97]. Compound 2-(3-(4-chlorophenyl)-3-hydroxybutanoyl)-N-phenylhydrazinecarbothioamide (Fig. 25) exihibited the best anticancer activity against MCF-7 breast cancer cells. This compound stimulated the pro-survival proteins (Bax, Bad and ERK1/2), while it does not stimulate anti-apoptotic proteins (Bcl-2, Akt and STAT-3) on western blotting analysis. 2-(3-(4-chlorophenyl)-3-hydroxybutanoyl)-Nphenylhydrazinecarbothioamide might activate apoptosis in human MCF-7 cells by targeting c-Jun N-terminal kinase (JNK) signaling it has also been reported in this study. NH O
NH
OH
NH
S
Cl
Fig. (25).
Khaled et al. [98] synthesized containing new pyrazolo[3,4d]pyrimidine-4-one thiosemicarbazides for the activity against MCF-7 breast cancer cells. Different derivatives with hydroxylamine hydrochloride, urea, thiosemicarbazide, phenyl hydrazine and aromatic amines were synthesized and all of them showed anticancer activity. 3,6-Dimethyl-4-oxo-1-phenyl-1,4-dihydro-5Hpyrazolo[3,4-d]pyrimidine-5-carbothiohydrazide (Fig. 26) showed anticancer activity with IC50 values of 52 µM. O
S
N
NH
NH2
N N
N
Fig. (26).
Mavrova et al. [99] synthesized new thiosemicarbazides from thieno[2,3-d]pyrimidin-4(3H)-one and investigated their anticancer activity against HeLa, HT-29, HepG2, MDA-MB-231, and Lep3 cell lines. The activity of the compounds were changing according to the substitutions and this study indicated that two thiosemicarbazide compounds N-ethyl-2-{[5-methyl-4-oxo-6-(2-oxobutyl)-3,4-dihydrothieno [2,3-d]pyrimidin-2-yl]acetyl}hydrazinecarbothioamide (Fig. 27) and N-phenyl-2-{[5-methyl-4-oxo-6-(2-oxobutyl)-3,4-dihydrothieno [2,3-d]pyrimidin-2-yl]acetyl}hydrazinecarbothioamide (Fig. 28) showed activity against HeLa and MDA-MB-231, compound N-phenyl-2{[5-methyl-4-oxo-6-(2-oxobutyl)-3,4-dihydrothieno [2,3-d] pyrimidin2-yl]acetyl}hydrazinecarbothioamide (Fig. 28) showed activity against HeLa, HT-29, HepG2, MDA-MB-231 and Lep3 cell lines. When an alkyl substitution remains in R1 position, compounds
R R
O NH
1
S
O
N
NH
NH
NH
R
2
S
HT-29, MDA-MB-231, HepG2, HeLa, Lep3 R: CH3, R1: COOC2H5, R3: C6H5
Fig. (28).
N-(4-aryl/cyclohexyl)-2-(pyridine-4-ylcarbonyl)hydrazinecarbothioamide derivatives (Fig. 29) were synthesized and tested for their anticancer activity against HT1080 (skin), HepG2 (liver) and A549 (lung) cancer cell lines by Bhat et al. [100]. Compounds bearing chloro, nitro and phenyl substitution were chosen for the cytotoxicity test in this study. These compounds could be considered as possible anticancer agents in drug development because of their cytotoxic effect on the tested cancer cell lines. N
S NH
R NH
NH
O R: Phenyl, 4-chlorophenyl, 2-nitrophenyl
Fig. (29).
Yadagiri et al. [101] synthesized some new 1,3,4-oxadiazole, 1,3,4-thiadiazole and 1,2,4-triazole derivatives starting from the active compounds thiosemicarbazides. They investigated their in vitro anticancer activity against cervical, breast, pancreatic and alveolar human cancer cell lines. Among the tested compounds, 2-[(9-chloro-2,3-dimethyl-6,7-dihydro-5H-benzo[7]annulen-8-yl) carbonyl]-N-phenylhydrazinecarbothioamide (Fig. 30) inhibited cell growth significantly in three human cancer cell lines with GI50 values of 0.471 µM against HeLa, 0.928 µM against PANC1 and 0.759 µM against A549. Cl H3C
O
NH NH
NH S
H3C
Fig. (30).
Refat et al. [102] studied the anticancer activity of new thiosemicarbazide ligand and its copper complex against hepatocellular carcinoma cells (HepG-2 cell line), breast carcinoma cells (MCF-7 cell line) and colon carcinoma cells (HCT cell line) as well as their structure elucidation. Binuclear Cu(II) and Mn(II) newly transition metal complexes of the 5-benzylidene-3-(4chlorophenyl)-6-oxo-5,6-dihydro-1H-[1,2,4]triazine-2-carbothioic
Macromolecular Drug Targets and Thiosemicarbazides
Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10 1297
acid amide (HL1) and 5-(3-bromo-4-methoxybenzylidene)-3-(4chloro-phenyl)-6-oxo-5,6-dihydro-1H-[1,2,4]triazine-2-carbothioic acid amide (HL2) derived from the condensation of oxazolinone with thiosemicarbazide have been prepared. When compared with standard anticancer drug doxorubicin, 5-benzylidene-3-(4-chlorophenyl)-6-oxo-5,6-dihydro-1H- [1,2,4]triazine-2-carbothioic acid amide (HL1) ligand was reported to have activity against HEPG-2, MCF-7 and HCT cell lines. Furthermore, the ligand, showed activity against MCF-7, HEPG-2 and HCT cell lives with IC50 values of 0.800, 1.60, 1.00 µg respectively (Fig. 31).
AUTHOR’S PROFILE Ş. Güniz Küçükgüzel, Ph.D from the Marmara University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry. She has been working there for 22 years. Since many years she has been engaged in the synthesis of compounds with biological activity. She is the author of 42 publications (31 SCI and 11 nonSCI) five (EP and WO) and six (TPE) patent applications. CONFLICT OF INTEREST The authors confirm that this article content has no conflict of interest.
O
NH N
N
ACKNOWLEDGEMENTS
NH2
Declared none. S
REFERENCES [1] Cl
Fig. (31).
[2]
N-Alkyl/Arylsubstituted-2-{[1-methyl-5-(4-methylbenzoyl)1H-pyrrol-2-yl]acetyl}hydrazinecarbothioamides have been synthesized with the reaction of 2-[1-methyl-5-(4-methylbenzoyl)1H-pyrrol-2-yl]acetohydrazide [103] and substituted alkyl/aryl isothiocyanates by Dadaş et al. [104]. N-(4-fluorophenyl)-2-{[1methyl-5-(4-methylbenzoyl)-1H- pyrrol-2-yl]acetyl}hydrazinecarbothioamide (Fig. 32) was tested for anticancer activity against androgen-independed human prostate cancer PC-3 (ATCC, CRL1435), human colon cancer HCT-116 (ATCC, CCL-247) and HT-29 (ATCC, HTB-38) cancer cell lines using MTT assay in comparising with Tolmetin. 4-Fluorophenyl substituted tolmetin thiosemicarbazide exhibited anticancer activity against PC-3 prostate cancer cell line with IC50 value of 185.4 µM.
[3]
[4]
[5]
CH3
[6] [7] O
CH3 N
O
[8]
NH
NH NH S
F
[9]
Fig. (32). [10]
5. CONCLUSION Macromolecular targets that are involved in the development of cancer research is rapidly increasing and researchers are focusing on developing new anti-cancer drug targets that are only be leaded to certain molecules. Thiosemicarbazides are reactive agents and they are used for the synthesis of 1,3,4-thiadiazoles, 1,3,4-oxadiazoles, 1,3-thiazolidine-4ones and 1,2,4-triazoles and therefore, the thiosemicarbazide synthesis approach has been observed in a lot of useful applications in drug research and development. The number of publications on thiosemicarbazide synthesis and their anticancer activity has been increasing in the recent years. In this paper, macromolecular targets and the related articles and reviews of thiosemicarbazides, their metal complexes and their anticancer activity have been reported. Future researches may be focused on molecular docking and molecular binding studies of thiosemicarbazides.
[11]
[12] [13] [14] [15]
[16]
Rajeshwar, P.; Verma, H.; Corwin, H. Matrix metalloproteinases (MMPs): Chemical-biological functions and (Q)SARs. Bioorg. Med. Chem., 2007, 15, 2223-2268. Philip, S.; Inturi, B.; Bhavya, K.; Pujar, G.V.; Purohit, M. 3dQSAR and molecular docking studies on 1, 2, 4 triazoles as MetAP2 inhibitors. Int. J. Pharm. Sci., 2014, 6, 205-212. Garrabrant, T.; Tuman, R.W.; Ludovici, D.; Tominovich, R.; Simoneaux, R.L.; Galemmo, R.A.Jr.; Johnson, D.L. Small molecule inhibitors of methionine aminopeptidase type 2 (MetAP-2) fail to inhibit endothelial cell proliferation or formation of microvessels from rat aortic rings in vitro. Angiogenesis, 2004, 7, 91-96. Marino, J.P.Jr.; Fisher, P.W.; Hofmann, G.A.; Kirkpatrick, R.B.; Janson, C.A.; Johnson, R.K.; Ma, C.; Mattern, M.; Meek, T.D.; Ryan, M.D.; Schulz, C.; Smith, W.W.; Tew, D.G.; Tomazek, T.A. Jr.; Veber, D.F.; Xiong, W.C.; Yamamoto, Y.; Yamashita, K.; Yang, G.; Thompson, S.K. Highly potent inhibitors of methionine aminopeptidase-2 Based on a 1,2,4-Triazole pharmacophore. J. Med. Chem., 2007, 50, 3777-3785. Selvakumar, P.; Lakshmikuttyamma, A.; Das, U.; Pati, H.N.; Dimmock, J.R.; Sharma, R.K. NC2213: A novel methionine aminopeptidase 2 inhibitor in human colon cancer HT29 cells. Mol. Cancer, 2009, 8(65). Nam, N.H.; Parang, K. Current targets for anticancer drug discovery. Curr. Drug Target., 2003, 4(2), 159. Ayub, M.; Stitch, S.R. Effect of ketoconazole on placental aromatase, 3β-hydroxysteroid dehydrogenase-isomerase and 17β-hydroxysteroid dehydrogenase. J. Steroid Biochem., 1986, 25, 981-984. Scozzafava, A.; Owa, T.; Mastrolorenzo, A.; Supuran, C.T. Anticancer and antiviral sulfonamides. Curr. Med. Chem., 2003, 10, 925-953. Radadiya, A.; Shah, A. Bioactive benzofuran derivatives: An insight on lead developments, radioligands and advances of the last decade. Eur. J. Med. Chem., 2015, 97, 356-376. Haluska, P.; Dya, G.K.; Adjeib, A.A. Farnesyl transferase inhibitors as anticancer agents. Eur. J. Cancer, 2002, 38, 1685-1700. Saha, A.K.; Liu, L.; Simoneaux, R.; DeCorte, B.; Meyer, C.; Skrzat, S.; Breslin, H.J.; Kukla, M.J.; End, D.W. Novel triazole based inhibitors of Ras farnesyl transferase. Bioorg. Med. Chem. Lett., 2005, 15, 5407-5411. Forde, J.E.; Dale, T.C. Glycogen synthase kinase 3: A key regulator of cellular fate. Cell. Mol. Life Sci. 2007, 64, 1930-1944. Ropero, S.; Esteller, M. The role of histone deacetylases (HDACs) in human cancer. Mol. Oncol., 2007, 1, 19- 25. Paul, M.K.; Mukhopadhyay, A.K. Tyrosine kinase - Role and significance in Cancer. Int. J. Med. Sci. 2004, 1(2), 101-115. Ségaliny, A.I.; Tellez-Gabriel, M.; Heymann, M.F.; Heymann, D. Receptor tyrosine kinases: Characterisation, mechanism of action and therapeutic interests for bone cancers. J. Bone Oncol., 2015, 4, 1-12. Arrieta, O.; Cardona, A.F.; Corrales, L.; Campos-Parraa, A.D.; Sánchez-Reyes, R.; Amieva-Riveraa, E.; Rodríguez, J.; Vargas, C.; Carranza, H.; Otero, J.; Karachalioue, N.; Astudillof, H.; Rafael, Rosell, R.; on behalf of CLICaP1. The impact of common and rare EGFR mutations in response to EGFR tyrosine kinase inhibitors
1298 Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10
[17]
[18] [19] [20]
[21] [22]
[23]
[24]
[25] [26]
[27] [28]
[29]
[30]
[31]
[32] [33]
[34] [35]
[36] [37]
and platinum-based chemotherapy in patients with non-small cell lung cancer. Lung Cancer, 2015, 87, 169-175. Wiese, H.; Gelis, L.; Wiese, S.; Reichenbach, C.; Jovancevic, N.; Osterloh, M.; Meyer, H.E.; Neuhaus, E.M.; Hatt, H.H.; Radziwill, G.; Warscheid, B. Quantitative phosphoproteomics reveals the protein tyrosine kinase Pyk2 as a central effector of olfactory receptor signaling in prostate cancer cells. Biochim. Biophys. Acta, 2015, 1854, 632-640. Shay, J.W.; Wright, W.E. Role of telomeres and telomerase in cancer. Sem. Cancer Biol., 2011, 21, 349-353. Goldman, M.A. The role of telomeres and telomerase in cancer. Drug Discov, Today, 2003, 8(7), 294-296. Wu, X.; Smavadati, S.; Nordfjäll, K.; Karlsson, K.; Qvarnström, F.; Simonsson, M.; Bergqvist, M.; Gryaznov, S.; Ekman, S.; PaulssonKarlsson, Y. Telomerase antagonist imetelstat inhibits esophageal cancer cell growth and increases radiation-induced DNA breaks. Biochim. Biophys. Acta, 2012, 1823, 2130-2135. Bajaj, S.; Asati, V.; Singh, J.; Roy, P.P. 1,3,4-Oxadiazoles: An emerging scaffold to target growth factors, enzymes and kinases as anticancer agents. Eur. J. Med. Chem., 2015, 97, 124-141. Rose, M.G.; Farrell, M.P.; Schmitz, J.C. Thymidylate Synthase: A critical target for cancer chemotherapy. Clin. Colorectal Cancer, 2002, 1(4), 220-229. Kadayat, T.M.; Song, C.; Shin, S.; Magar, T.B.T.; Bist, G.; Shrestha, A.; Thapa, P.; Na, Y.; Kwon, Y.; Lee, E.S. Synthesis, topoisomerase I and II inhibitory activity, cytotoxicity, and structureactivity relationship study of 2-phenyl- or hydroxylated 2-phenyl-4aryl-5H-indeno[1,2-b]pyridines. Bioorg. Med. Chem., 2015, 23, 3499-3512. Demoulin, B.; Hermant, M.; Castrogiovanni, C.; Staudt, C.; Dumont, P.Resveratrol induces DNA damage in colon cancer cells by poisoning topoisomerase II and activates the ATM kinase to trigger p53-dependent apoptosis. Toxicol. In Vitro, 2015, 29, 11561165. Adams, J. The proteasome: Structure, function, and role in the cell. Cancer Treat. Rev., 2003, 29(1), 3-9. Landis-Piwowar, K.R.; Milacic, V.; Chen, D.; Yang, H.; Zhao, Y.; Chan, T.H.; Yan, B.; Dou, Q.P. The proteasome as a potential target for novel anticancer drugs and chemosensitizers. Drug Resist. Updates, 2006, 9, 263-273. Krajewska, B. Ureases, I. Functional, catalytic and kinetic properties: A review. J. Mol. Catal. B: Enzym., 2009, 59, 9-21. Serwar, M.; Akhtar, T.; Hameed, S.; Khan, K.M. Synthesis, urease inhibition and antimicrobial activities of some chiral 5-aryl-4-(1phenylpropyl)-2H-1,2,4-triazole-3(4H)-thiones. ARKIVOC. 2009, vii, 210-221. Xu, Y.P.; Qin, J.; Sun, S.M.; Liu, T.T.; Zhang, X.L.; Qian, S.S.; Zhu, H.L. Synthesis, crystal structures, molecular docking and urease inhibitory activity of nickel(II) complexes with 3-pyridinyl4-amino-5-mercapto- 1,2,4-triazole. Inorg. Chim. Acta, 2014, 423, 469-476. Amtul, Z.; Rasheed, M.; Choudhary, M.I.; Rosanna, S.; Khan, K.M.; Atta-ur-Rahman. Kinetics of novel competitive inhibitors of urease enzymes by a focused library of oxadiazoles/thiadiazoles and triazoles. Biochem. Biophys. Res. Commun. 2004, 319, 10531063. Martin, T.A.; Jiang, W.G. Hepatocyte growth factor and its receptor signalling complex as targets in cancer therapy. Anticancer Agents in Med. Chem., 2010, 10, 2-6. Romagnolo, A.P.G.; Romagnolo, D.F.; Selmin, O.I. BRCA1 as target for breast cancer prevention and therapy. Anticancer Agents Med. Chem., 2015, 15, 4-14. Rathod, V.; Jain, S.; Nandekar, P.; Sangamwar, A.T. Human pregnane X receptor: A novel target for anticancer drug development. Drug Discov. Today, 2014, 19(1), 63-70. Mahapatra, D.K.; Bharti, S.K.; Asati, V. Anti-cancer chalcones: Structural and molecular target perspectives. Eur. J. Med. Chem., 2015, 98, 69-114. Salomon, D.S.; Brandt, R.; Ciardiello, F.; Normanno, N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit. Rev. Oncol. Hematol., 1995, 19, 183-232. Bianco, R.; Gelardi, T.; Damiano, V.; Ciardiello, F.; Tortora, G. Rational bases for the development of EGFR inhibitors for cancer treatment. Int. J. Biochem. Cell Biol., 2007, 39, 1416-1431. Zhang, J.H.; Qian, Y.; Zhu, D.D.; Yang, X.G.; Zhu, H.L. Synthesis, molecular docking and biological evaluation of chalcone
Küçükgüzel and Coşkun
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48] [49]
[50] [51]
[52]
[53]
[54] [55]
[56] [57]
thiosemicarbazide derivatives as novel anticancer agents. Eur. J. Med. Chem., 2011, 46, 4702-4708. Kaur, R.; Kaur, G.; Gill, R.K.; Soni, R.; Bariwal, J. Recent developments in tubulin polymerization inhibitors: An overview. Eur. J. Med. Chem., 2014, 87, 89-124. Wang, Y.T.; Qin, Y.J.; Yang, N.; Zhang, Y.L.; Liu, C.H.; Zhu, H.L. Synthesis, biological evaluation, and molecular docking studies of novel 1-benzene acyl-2-(1-methylindol-3-yl)-benzimidazole derivatives as potential tubulin polymerization inhibitors. Eur. J. Med. Chem., 2015, 99, 125-137. Parmar, S.S.; Joshi, P.C.; Ali, B.; Cornatze, W.E. Anticanvulsant activity and inhibition of respiration in rat brain homogenates by substituted oxadiazoles. J. Pharm. Sci., 1974, 63(6), 872-875. Ali, B.; Kumar, R.; Parmar, S.S.; Dwivedi, C.; Harbison, R.D. Antihemolytic and anticonvulsant activities of 1-(2,4-dichloro/ 2,4,5-tricholorophenoxyacetyl)-4-alkyl/arylthiosemicarbazides and their inhibition of NAD-dependent oxidations and monoamine oxidase. J. Pharm. Sci., 1975, 64(8), 1329-1333. Parmar, S.S.; Chaudhary, M.; Chaudhary, S.K.; Kumar, S.; Spiro, H.R. Anticonvulsant activity and selective inhibition of NADdependent oxidations in rat brain homogenates by newer mercaptotriazoles. J. Pharm. Sci., 1977, 66(7), 971-975. Singh, S.P.; Pandey, B.R.; Kumar, S.; Parmar, S.S. Anticonvulsant activity and inhibition of respiration in rat brain homogenates by substituted trimethoxybenzamides. J. Pharm. Sci., 1978, 67(12), 1682-1685. Singh, S.P.; Bahadur, S. Synthesis of some new substituted thiosemicarbazides as potential antiviral agents. Arch. Pharm. (Weinheim), 1983, 316, 817-821. Küçükgüzel, Ş.G.; Kocatepe, A.; Clercq, E.D.; Şahin, F.; Güllüce, M. Synthesis and biological activity of 4-thiazolidinones, thiosemicarbazides derived from diflunisal hydrazide. Eur. J. Med. Chem., 2006, 41, 353-359. Çıkla, P.; Arora, P.; Basu, A.; Talele, T.T.; Kaushik-Basu, N.; Küçükgüzel, Ş.G. Etodolac thiosemicarbazides: A novel class of hepatitis C virus NS5B polymerase inhibitors. Marmara Pharm. J., 2013, 17, 138-146. Raman, K.; Singh, H.K.; Salzman, S.K.; Parmar, S.S. Substitued thiosemicarbazides and corresponding cyclized 1,3,4-oxadiazoles and their anti-inflammatory activity. J. Pharm. Sci., 1993, 82(2), 167-169. Nargund, L.V.G.; Reddy, G.R.N.; Hariprasad, V. Anti-inflamatory activity of substituted 1,3,4-oxadiazoles. J. Pharm. Sci., 1994, 83(2), 246-248. Dzitko, K.; Paneth, A.; Plech, T.; Pawelczyk, J.; Staczek, P.; Stefanska, J.; Paneth, P. 1,4-Disubstituted thiosemicarbazide derivatives are potent inhibitors of toxoplasma gondii proliferation. Molecules, 2014, 19, 9926-9943. Habib, N.S.; Khalil, M.A. Synthesis and antimicrobial activity of novel quinazolone derivates. J. Pharm. Sci., 1984, 73(7), 982-985. Goswami, B.N.; Kataky, J.C.S.; Baruah, J.N. Synthesis and antibacterial activityof 1-(2,4-dicholorobenzoyl)-4-substituted thiosemicarbazides, 1,2,4-triazoles and their methyl derivates. J. Heterocyclic Chem., 1984, 21, 1225-1229. Rida, S.M.; Labouta, I.M.; Salama, H.M.; Ghany, Y.S.; ElGhaz-Zaui, E.; Kader, O. Syntheses and in vitro antimicrobial evaluation of some benzimidazol-2-ylmethylthioureas, benzimidazol2-ylacetylthiosemicarbazides and products of their condensation with monochloroacetic acid. Pharmazie., 1986, 41(7), 475-478. Kumar, H.; Patel, V.; Fernandes, P.S. Synthesis and biological activities of some substituted pyrazolylmethylene-1,2,4-triazoles, 1,3,4-thiadiazoles and -1,3,4-oxadiazoles. J. Ind. Chem. Soc., 1990, 67, 401-403. Ergenç, N.; Ilhan, E.; Ötük, G. Synthese und biologische wirkung einiger 1,4-disubstituierter thiosemicarbazide und deren 1,2,4triazol-5-thion-derivate. Pharmazie., 1992, 47(1), 59-60. Doğan, H.N.; Rollas, S.; Erdeniz, H. Synthesis, structure elucidation and antimicrobial activity of some 3-hydroxy-2-naphthoic acid hydrazide derivatives. Il Farmaco., 1998, 53(7), 462-467. Doğan, H.N.; Duran, A.; Yemni, E. Synthesis and antibacterial activity of 1-(3-hydroxy-2-naphthoyl)-4-substituted thiosemicarbazides. Drug Metab. Drug Interact., 1999, 15(2-3), 187-195. Gökçe, M.; Çakır, B.; Özçelik, B.; Abbasoğlu, U.; Şahin, M.F. Synthesis and antimicrobial activity of some 4-alkyl and aryl substituted [(2-oxobenzothiazolin-3-yl)acetyl]-3-semicarbazides and thiosemicarbazides. J. Fac. Pharm. Gazi., 2000, 17(1), 61-70.
Macromolecular Drug Targets and Thiosemicarbazides [58]
[59]
[60]
[61] [62]
[63]
[64] [65]
[66]
[67]
[68]
[69]
[70] [71]
[72]
[73]
[74]
[75] [76]
[77] [78]
Pishawikar, S.A.; More, H.N. Synthesis of mannich bases of thiosemicarbazide as DNA polymerase inhibitors and novel antibacterial agents. Int. J. Pharm. Bio. Sci., 2013, 4(1), 549-556. Sambanthamoorthy, K.; Hickman, M.; Pattabiraman, N.; Palys, T.; Wagar, E.J. Modulating acinetobacter baumannii biofilm development with molecules containing 3,4,5-trimethoxy-N,N’,N’trimethylbenzohydrazide moiety. Bioorg. Med. Chem. Lett., 2015, 25, 2238-2242. Cesur, Z.; Güner, H.; Ötük, G. Synthesis and antimycobacterial activity new imidazo[2,1-b]thiazole derivates. Eur. J. Med. Chem., 1994, 29, 981-983. Cesur, N.; Cesur, Z. Some imidazo[1,2-a]pyridine derivates as possible antimycobacterials. J. Fac. Pharm. İstanbul., 1998, 32, 29-35. Cesur, N.; Birteksöz, S.; Ötük, G. Synthesis and biological evaluation of some new thiosemicarbazide, 4-thiazolidinone, 1,3,4oxadiazole and 1,2,4-triazole-3-thione derivates bearing imidazo [1,2-a]pyridine moiety. Acta Pharma. Turcica., 2002, 44, 23-41. Ünver, Y.; Sancak, K.; Çelik, F.; Birinci, E.; Küçük, M.; Soylu, S.; Arslan Burnaz, N. New thiophene-1,2,4-triazole-5(3)-ones: Highly bioactive thiosemicarbazides, structures of Schiff bases and triazoleethiols. Eur. J. Med. Chem., 2014, 84, 639-650. Ram, J.V.; Pandey, H.N. Synthesis of 5-membered heterocycles and related compounds. Chem. Pharm. Bull., 1974, 22(12), 2778-2783. Kalyoncuoğlu, N.; Rollas, S.; Sür-Altiner, D.; Yeğenoğlu, Y. 1-[p- (Benzoylamino)benzoyl]-4-substituted thiosemicarbazides: synthesis and antibacterial and antifungal activities. Pharmazie., 1992, 47(10), 796-797. Rollas, S.; Karakuş, S.; Durgun, B.B. Synthesis and antimicrobial activity of some 1,4-disubstituted thiosemicarbazide and 2,5disubstituted 1,3,4-thiadiazole derivates. Il Farmaco. 1996, 51(12), 811-814. Çapan, G.; Ulusoy, N.; Ergenç, N.; Kiraz, M. New 6phenylimidazo[2,1-b]thiazole derivates: Synthesis and antifungal activity. Monatsh. für Chemie., 1999, 130, 1399-1407. Varvaresou, A.; Tsantili-Kakoulidou, A.; Siatra-Papastaikoudi, T.; Tiligada, E. Synthesis and biological evaluation of indole containing derivates of thiosemicarbazide and their cyclic 1,2,4triazole and 1,3,4-thiadiazole analogs. Arzneim. Forsch. Drug Res., 2000, 50(1), 48-54. Singhal, S.; Arora, S.; Agarwal, S.; Sharma, R.; Singhal, N. A review on potential biological activities of thiosemicarbazides. World J. Pharm. Pharm. Sci., 2013, 2(6), 4661-4681. Arora, S.; Agarwal, S.; Singhal, S. Anticancer Activities of thiosemicarbazides/thiosemicarbazones: A Review. Int. J. Pharm. Pharm. Sci., 2014, 6(9), 34-41. Wu, X.; Zhu, Y.; Yan, H.; Liu, B.; Li, Y.; Zhou, Q.; Xu, K.; Isothiocyanates induce oxidative stress and suppress the metastasis potential of human non-small cell lung cancer cells. BMC Cancer, 2010, 10, 269-279. Küçükgüzel, Ş.G.; Oruç, E.E.; Rollas, S.; Şahin, F.; Özbek, A. Synthesis, Characterisation and biological activity of novel 4thiazolidinones, 1,3,4-oxadiazoles and some related compounds. Eur. J. Med. Chem., 2002, 37(3), 197-206. Küçükgüzel, Ş.G.; Küçükgüzel, İ.; Tatar, E.; Rollas, S.; Şahin, F.; Güllüce, M.; Clercq, E.D.; Kabasakal, L. Synthesis of some novel heterocyclic compounds derived from diflunisal hydrazide as potential anti-infective and anti-inflammatory agents. Eur. J. Med. Chem., 2007, 42, 893-901. Kane, J.M.; Staeger, M.A.; Dalton, C.R.; Miller, F.P.; Dudley, M.W.; Ogden, A.M.L.; Kehne, J.H.; Ketteler, H.J.; McCloskey, T.C.; Senyah, Y.; Chmielewski, P.A.; Miller, J.A. 5-Aryl-3(alkylthio)-4H-1,2,4-triazoles as selective antagonists of strychnineinduced convulsions and potential antispastic agents. J. Med. Chem., 1994, 37, 125-132. Rollas, S. Bazı l-aroil-4-alkil/ariltiyosemikarbazidler. Doğa Bilim Dergisi, Tıp., 1983, 7, 65-73. Serra, S.; Moineaux, L.; Vnacraeynest, C.; Masereel, B.; Wouters, J.; Pochet, L.; Frédérick, R. Thiosemicarbazide, a fragment with promising indolamine-2,3 dioxygenase (IDO) inhibition properties. Eur. J. Med. Chem., 2014, 82, 96-105. Creangă, A.A.; Bercean, V.N.; Badea ,V.; Ledeţi, I.; Csunderlik, C. Benzoylation of thiosemicarbazide. Chem. Bull., 2008, 53(67), 45-49. Omran, A. Synthesis and characterization of new thiacalix[4] arenes bearing semicarbazide and thiosemicarbazide fragments at the lower rim. Chin. J. Chem., 2009, 27, 1937-1941.
Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10 1299 [79]
[80] [81]
[82]
[83]
[84]
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94] [95]
[96] [97]
Katritzky, A.R.; Khashab, N.M.; Gromova, A.V. Preparations of diversely substituted thiosemicarbazides and N-hydroxythioureas. Arkivoc., 2006, 3, 226-236. Gülerman, N.N.; Rollas, S.; Ülgen, M. Synthesis of some substituted 1, 3,4-thiadiazole derivatives. J. Pharm. Univ.Mar., 1994, 10(1), 11-16. Sathisha, M.P.; Budagumpi, S.; Kulkarni, N.V.; Kurdekar, G.S.; Revankar, V.K.; Pai, K.S.R. Synthesis, structure, electrochemistry and spectral characterization of (D-glucopyranose)-4phenylthiosemicarbazide metal complexes and their antitumor activity against Ehrlich Ascites Carcinoma in Swiss albino mice. Eur. J. Med. Chem., 2010, 45, 106-113. Singh, N.K.; Nagendra, S.; Prasad, G.C.; Shrivastava, S.A. Antitumor activity studies of newly synthesized N-salicyloyl-N′-(phydroxybenzthioyl)hydrazine and its Copper(II) complex both in vivo and in vitro. Bioorg. Med. Chem., 1997, 5(2), 245-251. Singh, N.K.; Singh, S.B.; Shrivastav, A.; Singh, S.M. Spectral, magnetic and biological studies of 1,4-dibenzoyl-3-thiosemicarbazide complexes with some first row transition metal ions. Proc. Indian Acad. Sci., 2001, 113(4), 257-273. Shrivastava, A.; Singha, N.K.; Singhb, S.M. Synthesis, characterization and antitumor studies of Mn(II), Fe(III), Co(II), Ni(II), Cu(II) and Zn(II) complexes of N-Salicyloyl-N0 -o-hydroxythiobenzhydrazide. Bioorg. Med. Chem., 2002, 10, 887-895. El-Metwally, N.M.; Al-Hazmi, G.A.A. Spectroscopic evaluation for VO(II), Ni(II), Pd(II) and Cu(II) complexes derived from thiosemicarbazide: A special emphasis on EPR study and DNA cleavage. Spectrochim. Acta A, 2013, 107, 289-295. Rocha, F.V.; Barra, C.V.; Mauro, A.E.; Carlos, I.Z.; Nauton, L.; Ghozzi,, M.E.; Gautier, A.; Morel, L.; Netto, A.V.G. Synthesis, characterization, X-ray structure, DNA Cleavage, and Cytotoxic Activities of Palladium(II) complexes of 4-Phenyl-3thiosemicarbazide and triphenylphosphane. Eur. J. Inorg. Chem., 2013, 4499-4505. Mlahi, M.R.; Azhari, S.J.; El-Asmy, A.A.; Mostafa, M.M. Comparative spectroscopic and DFT calculations of binary and ternary complexes derived from 4-allyl-1-(2-hydroxybenzoyl) thiosemicarbazide (L1) and 2,20-dipyridyl. Spectrochim. Acta A, 2015, 134, 465-472. Gururaja, T.L.; Goff, D.; Kinoshita, T.; Goldstein, E.; Yung, S.; McLaughlin, J.; Pali, E.; Huang, J.; Singh, R.; Daniel-Issakani, S.; Hitoshi, Y.; Cooper, R.D.G.; Payan, D.G. R-253 Disrupts microtubule networks in multiple tumor cell lines. Clin. Cancer Res., 2006, 12(12), 3831-3842. Al-Saadi, M.S.; Faidallah, H.M.; Rostom, S.A.F. Synthesis and biological evaluation of some 2,4, 5-trisubstituted thiazole derivatives as potential antimicrobial and anticancer agents. Arch. Pharm. Chem. Life. Sci., 2008, 341, 424 - 434. El-Sabbagh, O.I.; Rady, H.M. Synthesis of new acridines and hydrazones derived from cyclic β -diketone for cytotoxic and antiviral evaluation. Eur. J. Med. Chem., 2009, 44, 3680-3686. Sun, Y.; Wang, W.; Sun, Y.; Han, M. Synthesis and biological evaluation of a novel stem/progenitor cells proliferation activator: 4-(4-(5-mercapto-1,3,4-oxadiazole-2-yl)phenyl)thiosemicarbazie (Stemazole). Eur. J. Med. Chem., 2011, 46, 2930-2936. Nofal, Z.M.; Soliman, E.A.; El-Karim, S.S.A.; Elzahar, M.I.; Srour, A.M.; Sethumadhavan, S.; Maher, T.J. Novel benzimidazole derivatives as expected anticancer agents. Acta. Pol. Pharm. Drug Res., 2011, 68(4), 519-534. Yousef, A.T.; Badria, A.F.; Ghazy, E.S.; El-Gammal, A.O.; ElReash, G.M.A. In vitro and in vivo antitumor activity of some synthesized 4-(2-pyridyl)-3-Thiosemicarbazides derivatives. Int. J. Med. Med Sci., 2011, 3(2), 37-46. He, J.; Wang, X.; Zhao, X.; Liang, Y.; He, H.; Fu, L. Synthesis and antitumor activity of novel quinazolinone derivatives containing thiosemikarbazide moiety. Eur. J. Med. Chem., 2012, 54, 925-930. Siwek, A.; Staczek, P.; Wujec, M.; Bielawski, K.; Bielawska, A.; Paneth, P. Cytotoxic effect and molecular docking of 4ethoxycarbonylmethyl-1-(piperidin-4-ylcarbonyl)thiosemicarbazide-a novel topoisomerase II inhibitör. J. Mol. Mod., 2013, 19, 1319-1324. Koca, I.; Aykut, O.; Coskun K.A.; Tutar, Y. Synthesis and anticancer activity of acyl thioureas bearing pyrazole moiety. Bioorg. Med. Chem., 2013, 21, 3859-3865. Malki, A.; Elbayaa, R.Y.; Ashour, H.M.; Loffredo, C.A.; Youssef , A.M. Novel thiosemicarbazides induced apoptosis in human MCF-
1300 Anti-Cancer Agents in Medicinal Chemistry, 2016, Vol. 16, No. 10
[98]
[99]
[100]
[101]
7 breast cancer cells via JNK signaling. J. Enzyme. Inhib. Med. Chem., 2014, 3, 1-10. Khaled, R.A.A.; Eman, K.A.A.; Mohamed, A.A.; Rasha, R.A.; Rania, B.B. Synthesis and anticancer activity of some new pyrazolo[3,4-d]pyrimidin-4-one derivatives. Molecules., 2014, 19, 3297-3309. Mavrova, A.T.; Wessilinova, D.; Tsenov, J.A.; Lubenov, L.A. Synthesis and antiproliferative activity of some new thieno[2,3d]pyrimidin-4(3H)-ones containing 1,2,4-triazole and 1,3,4thiadiazole moiety. Eur. J. Med. Chem., 2014, 86, 676-683. Bhat, M.A.; Khan, A.A.; Khan, S.; Al-Omar, M.A.; Parvez, M.K.; Al-Dosari, M.S.; Al-Dhfyan, A. Synthesis and anti-Candidal activity of N-(4-aryl/cyclohexyl)- 2-(pyridine-4-yl carbonyl) hydrazinecarbothioamide. Bioorg. Med. Chem. Lett., 2014, 24, 1299-1302. Yadagiri, B.; Gurrala, S.; Bantu, R.; Nagarapu, L.; Polepalli, S.; Srujana, G.; Jain, N. Synthesis and evaluation of benzosuberone
Küçükgüzel and Coşkun
[102]
[103]
[104]
embedded with 1,3,4-oxadiazole, 1,3,4-thiadiazole and 1,2,4triazole moieties as new potential anti proliferative agents. Bioorg. Med. Chem. Lett., 2015, 25, 2220-2224. Refat, M.S.; El-Deen, I.M.; El-Garib, M.S.; El-Fattah, W.A. Spectroscopic and anticancer studies on new synthesized copper(II) and manganese(II) complexes with 1,2,4-Triazines thiosemicarbazide. Russ. J. Gen. Chem., 2015, 85(3), 692-707. Küçükgüzel, Ş.G.; Koç, D.; Çıkla-Süzgün, P.; Özsavcı, D.; BingölÖzakpınar, Ö.; Mega-Tiber, P.; Orun, O.; Erzincan, P.; Sağ-Erdem, S.; Ş ahin, F. Synthesis of tolmetin hydrazide-hydrazones and discovery of a potent apoptosis inducer in colon cancer cells. Arch. Pharm. Chem. Life Sci., 2015, 348(10), 730-742. Dadaş, Y.; Coşkun, G.P.; Bingöl-Özakpınar, Ö.; Özsavcı, D.; Küçükgüzel, Ş.G. Synthesis and anticancer activity of some novel tolmetin thiosemicarbazides. Marmara Pharm. J., 2015, 19, 259-267.