Abstract
Objective:
Breast cancer prevalence is continuing to rise worldwide. Despite the diversity of the current approaches and protocols to treat this heterogeneous disease, most of these face the challenges of side effects and resistance. Hence, novel and innovative approaches to the treatment of breast cancer are almost constantly needed. This study aimed to investigate the antiproliferative and death modalities induced by three thiosemicarbazide derivatives of captopril in two subtypes of breast cancer cell lines, the Estrogen- receptor positive MCF-7, and the Estrogen/progesterone- receptor-negative AMJ13.
Methods:
MTT assay was used to determine the cytotoxicity of the derivatives and their parent compound Captopril, Hematoxylin and Eosin (H&E) staining, Acridine Orange/ Ethidium Bromide (AO/EtBr) staining, Caspase immunocytochemistry analysis and ROS generation by Human ROMO1 ELISA assays were conducted to explore the type of cellular death induced by these derivatives.
Results:
One of the derivatives denoted as (8) demonstrated the best antiproliferative profile recording the highest cytotoxic effect with IC50 of (88.06 µM) and (66.82 µM) compared to that of captopril (849.8 µM),(1075µM) in MCF-7 and AMJ13 breast cancer cells respectively. In MCF-7 cells, derivative (8) imposed an apoptotic cellular death with the involvement of caspase-3, and caspase-9 and displayed a time-dependent ROS generation. In AMJ13 breast cancer cells, results revealed an extensive vacuole forming, non-apoptotic cellular death, without ROS generation, but with a significant implication of caspase-9.
Conclusion:
This study demonstrated the thiosemicarbazide derivative of captopril (8) as a promising antiproliferative agent against breast cancer cells displaying different cellular death modalities, signifying the versatility of the derivative and suggesting multitarget pathways. This study strongly recommends derivative (8) as a future leading molecule.
Key Words: Thiosemicarbazide Derivative of Captopril, Breast Cancer Cell Lines Antiproliferative, Death modalities
Introduction
In 2020, female breast cancer took the lead over lung cancer in the number of newly diagnosed cases worldwide [1]. The disease is characterized as highly heterogeneous with diverse subtypes classified on different bases. Molecularly and depending on gene expression of estrogen ER, progesterone PR and epidermal growth factor 2 into: luminal A (ER and /or PR)+, HER2 - ), luminal B ((ER and /or PR)+, HER2 ± ), HER2 overexpression (ER/PR)- HER2 + ), and basal-like (triple negative) TNBC (ER/PR)- HER2- ) [2, 3].
Over the years, many therapies have been developed for the treatment of all types of breast cancer, but almost all of them share the same challenges of side effects and resistance. In addition to that, the most aggressive with the worst prognosis TNBC still lacks standardized effective treatment [4].
As such, the pursuit of novel approaches seems in constant demand, and one of these new approaches is the Renin-Angiotensin System (RAS), which is implicated in almost all cancer hallmarks [5], including breast [6, 7]. Moreover, recent attention has been driven towards Angiotensin Converting Enzyme inhibitors (ACEi) and Angiotensin II type 1 Receptor Blockers (ARBs) role in oncology as potential chemopreventative, adjuvant or even anticancer agents [5, 8, 9].
Captopril, the first member of the ACEi family with a characteristic sulfhydryl group [10] was among those that were investigated for its potential role in the treatment of breast cancer with positive results [11-13].
Recently, Al-Saad and colleagues (2019) synthesized novel derivatives of captopril that demonstrated higher antiplatelet [14] and ACE inhibition activity than their parent compound captopril [15]. The authors justified the improvement to the incorporation of the thiosemicarbazide moiety that replaced the carboxylic acid group of captopril with the perseverance of the characteristic thiol group [14].
Worth mentioning, thiosemicarbazide/ thiosemicarbazone derivatives have biological activity against various types of cancer [16, 17]. These compounds are believed to induce their anticancer effects by inhibiting many enzymes and vital cell cycle targets, including ribonucleotide diphosphate reductase RNR) [18, 19], tubulin-colchicine and epidermal growth factor receptor (EGFR) tyrosine kinase (17), and topoisomerase II [20].
Remarkable antitumor activity of these derivatives has been documented by many studies in both the positive and negative hormonal receptor breast cancer cell lines. In the positive hormonal receptor breast cancer cell line (MCF-7), Malki and his fellow researchers (2015) demonstrated the anticancer effect of one of their novel thiosemicarbazide derivatives (4c) through targeting JNK signaling [21]. Other studies [22-27] investigated the antitumor activity of these derivatives in the negative hormonal receptors of breast cancer cells through targeting various cell targets.
Having in mind the implication of RAS in breast cancer [28] and the anticancer activities of both captopril and thiosemicarbazide, this work sought to investigate three thiosemicarbazide derivatives of captopril designated as (5, 7, and 8) (Figure 1) in two types of breast cancer cell lines; the estrogen receptor-positive MCF-7 and the aggressive estrogen/progesterone receptor-negative AMJ13 [29]. The derivatives were also investigated on the transformed non-tumorigenic HBL-100 breast cell line.
Figure 1.
Chemical Structure of Thiosemicarbazide Derivatives of Captopril (derivatives 5, 7 and 8)[14].
Materials and Methods
Chemicals and cell lines
The derivatives were supplied by Dr. Hiba. Najeh. AL-Saad, Department of Pharmaceutical Chemistry, College of Pharmacy, Basrah University, Basrah, Iraq. Derivatives (5, 7 and 8) molecular weights are 357.79, 384.88 and 380.46 respectively, all derivatives are highly pure compounds with 1HNMR analysis validation[14]. Captopril molecular weight (217.29) (≥ 98% HPLC) was provided from Sigma –Aldrich/ USA.
Cell Bank Unit of Experimental Therapy Department, ICCMGR, AL- Mustansiriyah University, Baghdad, Iraq supplied the cells (MCF-7, AMJ13 and HBL-100). AMJ13 and HBL-100 cells were maintained in RPMI-1640 medium (Gibco/USA), whereas MCF-7 was in MEM (U.S Biological, USA). All were supplied with (10%) fetal bovine serum (Bio west/ USA) and 1% of (Penicillin–Streptomycin) (Capricorn-Scientific, Germany) and Incubated at 37 C0 and (5%) CO2. Exponential growing cells were a prerequisite in every experiment.
Cytotoxicity
Cytotoxicity of the three-thiosemicarbazide derivatives and captopril was assessed by MTT assay. Simply, the procedure included seeding the 96 micro-plate with 1x 104 cells and incubating at 37oC until adherence and confluence were achieved. Followed by 72 hours of treatment with different concentration ranges (0– 2000 µM) for captopril and derivative (7), and (0-200 µM) for both derivatives (5) and (8) (the concentration ranges were selected based on a pilot study). Cells were then incubated for 4 hours with 50μl of MTT solution (2 mg/ml), which was then removed and solubilized with100 µl of DMSO for 20 minutes [30]. Optical densities were measured by an ELISA plate reader [31, 32]. Cytotoxicity was determined by the following equations [33].
Cytotoxicity (%) = (OD Control – OD sample) / OD Control × 100
Where OD Control = mean optical density of drug unexposed wells, OD sample mean optical density of drug-exposed wells, IC50 was then calculated using GraphPad Prism®, version 9.
Examination of morphological changes by Hematoxylin and Eosin staining (H&E)
Morphological changes were documented by H&E staining. The procedure was performed according to [34] with slight modifications. IC50 (88.08µM) and (66.82 µM) of derivative (8) were used to treat MCF-7 and AMJ13 respectively. Fixation was done with 2 drops of formalin for two minutes, followed by the addition of Hematoxylin for another two minutes. The slides were then washed with tap water for two minutes. Afterwards, two drops of Eosin were added and left for two minutes. The slides were then washed with distilled water and dehydrated by a series of ethanol dilutions (70%, 90%, and 100%). Xylene and Canada balsam were used for clearing and mounting respectively. The examination was done with Scopetek/USA Light microscope.
Assessment of apoptosis by the Acridine Orange/ Ethidium Bromide (AO/EtBr) staining
The assay was performed to assess the potential implication of apoptosis as a cellular death induced by derivative (8). Seeding of the cells were done in eight well chamber slides, followed by 72 hours of incubation with (88.06µM) and (66.82) of derivative (8) in MCF-7 and AMJ13 respectively. Double washing with sterile phosphate buffer saline preceded staining with 100 µl of AO/EtBr [35]. A fluorescence microscope (Olympus/ Japan) was used for examination, and Fiji (Image J version 1.53) was used for analysis.
Immunocytochemistry
In charged slides, cells were seeded and incubated for 10 minutes with -20o methanol. The endogenous peroxidase activity was blocked with 3% hydrogen peroxide, which was followed by thirty-minute incubation with normal goat serum. The slides were then incubated for one hour with primary antibodies dilution (anti-caspase-3, anti-caspase -9, and anti-PECAM-1 monoclonal IgG1) (Santa Cruz/ USA). Gentle washing with PBS was performed and repeated three times followed by incubation with a polymer helper that was once again followed by another PBS washing. 20- 30 minutes of incubation with secondary antibody dilution (polyperoxidase –anti-mouse/rabbit IgG/ Elabscience /USA/ E-IR-R213) ensued afterwards, which was followed by a third cycle of PBS washing. Counter-staining with pre-prepared DAB solution was done, followed by mounting, sealing and examination [36]. A digital camera (Micros, Austria) was used to take the images, and Fiji (image J version 1.53c) was used for analysis.
Assessment of ROS generation
Generation of ROS was assessed by Human ROMO1 ELISA Kit (Elabscience®, USA), which is supplied with a micro ELISA plate pre-coated with human ROMO1 antibody. All procedures were performed according to the manufacturer’s instructions. Binding with antibody occurs after incubation with the cell supernatant for 90 minutes at 2-8 Co. This was followed by one-hour incubation with Biotinylated Detection Ab working solution (Human ROMO1 specific) at 37 Co. Triplicate washing with a washing buffer was performed before and after incubation with (Avidin-Horseradish Peroxidase) HRP Conjugate working solution. In a dark room, the plate was supplied with substrate reagent and was incubated for 15 minutes. A stop solution was then added and the optical densities were measured by micro-plate reader at 450 nm.
Statistical analysis
Data was analysed using GraphPad Prism®, version 9. An unpaired t-test was used to compare between two groups with a significance of < 0.05, one– way ANOVA with Tukey’s multiple comparison test was used to compare the means of more than two groups. All results in this work were presented as mean ± SD.
Results
Cytotoxicity
Thiosemicarbazide derivative (8) has the best antiproliferative profile compared to the other derivatives (5 and 7) and their parent compound captopril. Where the derivative (8) has strongly inhibited the growth with an IC50 of (88.06µM) and (66.82µM) in MCF-7 and AMJ13 breast cancer cells respectively with lesser effect (153.3µM) on the transformed non-tumorigenic HBL -100 breast cells (Figure 2A). Compared to that of captopril which displayed an IC50 of (849.8 µM) and (1075µM) in MCF7 and AMJ13 respectively with no effect on non-tumorigenic breast cells (HBL-100) (Figure 2C).On the other hand, derivative (5) demonstrated improved cytotoxicity only on AMJ13 breast cancer cells with IC50 of (96.28 µM) with less effect on both MCF-7 (256.6 µM) and HBL-100 (204 µM) (Figure 2B). While Derivative (7) showed no inhibitory effect on any of the three cell lines (data not shown).
Figure 2.
Illustration of IC50 of A) thiosemicarbazide derivative (8). B) thiosemicarbazide derivative (5) and C) captopril in the estrogen receptor-positive MCF-7, ER/PR receptor negative AMJ13 breast cancer cell lines and in the non-tumorigenic breast cell line HBL-100. All results shown here were done as (n= 4, 3 and 2) with quadruplicate per experiment.
Identification of Cellular Death
Morphological changes by Hematoxylin and Eosin (H&E) staining
MCF-7 cells demonstrated mainly apoptotic features characterized by shrinkage of the cells and positional loss of organelles causing multiple opaque foci with slight vacuole degeneration (Figure 3A). AMJ13 displayed an atrophied pattern with extensive vacuole degeneration and some cytoplasm-free cells (Figure 3B).
Figure 3.
The Cell line Morphological Changes Following 72 hrs Exposure to Compound (8) A. MCF-7 (IC50 88.06). a) Control (untreated MCF-7 cell line) arrows indicate normal cells, whereas the arrowhead refers to a normal nucleus. b) The arrows refer to the vacuoles, whereas the asterisk refers to cell-free spaces. B) AMJ13 (IC50 66.82). a) Control (untreated AMJ-13 cell line). b) The black arrows refer to the vacuoles, the white ones refer to atrophied cells and the asterisk refers to cell-free space. All cells were stained with Hematoxylin and eosin staining and were imaged by light microscope 40 X.
Assessment of apoptosis by the Acridine Orange/ Ethidium Bromide (AO/EtBr) staining
The Acridine Orange/ Ethidium Bromide (AO/EtBr) staining was used for both quantitative and qualitative detection of apoptotic bodies induced by the derivative (8). In the MCF7 cell line, four stages of apoptosis were recorded, the viable cells having round nuclei and emitting green fluorescence. The early apoptotic cells with a greenish appearance demonstrate croissant-like chromatin condensation on the edges of irregularly shaped nuclei. The dead cells have the yellowish–orange appearance of rounded nuclei, and late apoptotic cells express condensed or fragmented chromatin of irregularly shaped nuclei with a yellow-orange stain (Figure 4a/ A2).
Figure 4.
AO/EtBr Double Staining Assay for Assessment of Apoptosis. a) A1 control MCF-7 cells, A2 MCF-7 treated cells with derivative (8) (IC50 = 88.06µM). The blue arrows refer to early apoptotic cells, while the white ones refer to the dead cells. B1) control AMJ-13 cells, B2) AMJ-13 treated cells with derivative (8) (IC50 =66.82µM). All images were visualized under a fluorescent microscope 40x. b) Statistical analysis comparing mean (percentage) apoptotic cells between control and derivative (8) treated cells calculated from AO/EtBr staining assay. An unpaired t-test was performed and the results represented are mean ± SD.
The analysis demonstrated a significant increase in the mean apoptotic cells (62.99%) compared to live cells (37.1%) in the MCF-7 cell line, and when compared to control cells there was a significant difference between the mean apoptotic cells with a p-value of (0.0001) (Figure 4b). These results indicate apoptosis as a type of cellular death produced in MCF-7 cells treated with a derivative (8).
On the other hand, in the AMJ13 cell line, the analysis revealed an insignificant number of apoptotic cells with a percentage of (23.6), and no significant difference was recorded between the mean apoptotic cells treated with derivative (8) to that of the control (untreated cells) (Figure 4b). Majority of the (23.6%) cells were either dead or late apoptotic (Figure 4a/B2).
Immunocytochemistry analysis
Monoclonal IgG1 were directed against caspase -3, caspase-9, and cell surface marker PECAM-1 to document their expression in the cells.
The results revealed a significant increase in caspase (3) (P-value 0.0082) and caspase -9 (P-value 0.0284) in MCF-7 cells treated with derivative (8) compared to the normal ones (Figure 5a/A2, A1/b) and (Figure 5a/B2, B1/b) respectively. In addition to that, derivative (8) imposed non-significant increase in PECAM-1 (CD-31) compared to non-treated cells (Figure 5a/C2, C1/b).
Figure 5.
a/ Immunocytochemistry Images of MCF-7 Cells Treated with (88.06µM) of Compound (8). Expression of caspase-3 in A1 control cells, A2 treated cells. Expression of caspase -9 in B1 control cells, B2 in treated cells expressing significant increase represented by the dark brown staining around the cells. Expression of PECAM-1 in C1 control and C2 treated cells. b/ Statistical analysis of quantitative expression of caspase -3, caspase-9 and PECAM-1 demonstrating a significant increase in caspase- 3 (p-value 0.0082) and caspase -9 (p-value 0.0284). Analysis was performed using 3-5 immunocytochemistry images analyzed by Fiji (Image J version 1.53). An unpaired t test was done and all results shown here are mean ± SD.
While in the AMJ13 cell line, the Derivative of captopril (8) imposed a statistically significant increase in caspase -9 (p-value 0.0003) (Figure 6b) illustrated as dark brown pigmentation around the cells (Figure 6a/B2) compared to very light scattered brown pigmentation control cells (Figure 6a/B1).
Figure 6.
a/ Immunocytochemistry Images of AMJ13 Cells Treated with (66.92µM) Derivative (8). Expression of caspase-3 in A1 control cells, A2 treated cells. Expression of caspse -9 in B1 control cells, B2 in treated cells expressing significant increase represented by the dark brown staining around the cells. Expression of PECAM-1 in C1 control and C2 treated cells. b/ Statistical analysis of quantitative expression of caspase -3, caspase-9 and PECAM-1 from immunocytochemistry test. An unpaired t-test was done and the results represented here are mean ± SD.
Contrary to caspase -9, the derivative had an insignificant increase in caspase -3 (Figure 6a/ A2, b) and PECAM-1. (Figure 6a/C2.b) compared to controls (Figure 6a/A1) and (Figure 6a/C1) respectively.
Assessment of ROS-inducing activity of the derivative
Two experiments were conducted, one included 72 hours of incubation of the cells with different concentrations of derivative (8) (IC50, up and down the IC50), and the second was performed by treatment of the cells with fixed concentration (IC50 of the derivative) at different time intervals (2, 6, and 12 hours).
In the MCF7 cell line, the first experiment’s highest level of ROS was achieved at the lowest concentration (25 µM), whereas MCF-7 basal ROS level was documented at both the IC50 (88.06 µM) and the highest concentration (250 µM) (Figure 7A).
Figure 7.
Generation of ROS in MCF-7 Cell Line after Treatment with Derivative (8). A) Cells were treated with three different concentrations (IC50, below, and above the IC50) of compound (8) for 72 hours. B) Cells were exposed to only 88.06 (µM) of derivative (8) for 2, 6, and 12 hours. Results are from two experiments with two replicates. Data presented here are mean + SD.
On the other hand, the second experiment, documented a time-dependent increase in ROS level to reach its maximum following 12 hours of incubation (Figure 7B), indicating that the ROS level decreased gradually to reach the basal level after 72 hours. In the AMJ13 cell line, results demonstrated no change in basal ROS level (data not shown).
Discussion
Globally, Breast cancer prevalence is increasing and its heterogeneity renders many therapies almost useless in combating certain sub-types of this disease.
Over the years, many therapies and personalized treatments have been developed, however many challenges still arise. As such, novel and innovative approaches for the treatment of breast cancer seem like an endless pursuit (3). Among the newly pursued approaches are the Renin-Angiotensin System inhibitors [5] and thiosemicarbazaide/ thiosemicarbazone derivatives [16, 17]. In this research, the two approaches have been joined by employing novel derivatives of the ACEi (Captopril) that include thiosemicarbazide moiety and were previously found to have an improved ACE inhibition activity over their parent compound captopril [15]
This work investigated three derivatives namely (5, 7, and 8) in the estrogen receptor-positive (MCF-7) and the highly aggressive estrogen/ progesterone receptor-negative (AMJ13) breast cancer cell lines. Results demonstrated derivative (8) having the best antiproliferative profile affecting both cell lines with much less effect on the non-tumorigenic breast cell line HBL-100. The distinct profile of the cytotoxicity imposed by the three derivatives can be explained based on their chemical structure. The substitution at the phenyl moiety is necessary for the anti-breast cancer function, where the 4 methoxy moiety of derivative (8) documented the highest inhibition of proliferation on all cell lines, whereas, the chlorine of derivative (7) abrogated this function. It is worth mentioning that this structure-activity relationship (SAR) was not uncommon for thiosemicarbazide derivatives and was documented by others [37].
Moreover, the no substitution of derivative (5) might be cell-specific where it exerted a strong effect on the ER/PR receptor negative AMJ13 with little effect and even can be referred to as not affecting MCF-7 when compared to transformed cells. To further elaborate, the presence of an electron-withdrawing group at the para-phenyl position will lead to the loss of activity as displayed with the MTT assay. Thus, for hydrophobic head unsubstituted, or substituted with electron donated group was essential for activity against breast cancer cell lines.
Worth mentioning, that these derivatives demonstrated different antiproliferative profiles when tested in other cancer cell lines, including HCT-116, A549, HeLa and HepG2 (unpublished data). These investigations confirmed that the derivatives are indeed versatile and behave depending on the type of cell involved.
To elucidate the cytotoxicity of derivative (8), further investigations focused on the type of cellular death and the potential mode of action exerted by this derivative. Results revealed the cellular-dependent behaviour of the derivative, where in MCF-7 the derivative induced apoptotic type of death with the involvement of caspase-3, caspase-9 and ROS generation. On the other hand, apoptosis’s distinct morphological profile was absent in AMJ13 cell lines, which demonstrated an extensive vacuole-forming type of death, without ROS generation, but with the significant implication of caspase-9.
This can be explained by the fact that caspase-9 activity is not limited to apoptosis; caspase-9 is implicated in the induction of paraptosis (a type of programmed non-apoptotic vacuole-inducing cell death) [38].
Numerous studies have described thiosemicarbazide/ thiosemicarbazone derivatives exhibiting several types of cellular death, including apoptosis [22, 39, 40], mitosis-like death [41], autophagy [42, 43], and paraptosis [44, 45]. As well as, exhibiting multiple cell death types at the same time as apoptosis and autophagy [43, 46, 47].
Notably, molecular docking studies revealed the derivative as a potential paraptosis-inducing agent through targeting the most important signalling pathways involved (JNK1, p38 and MEK-2) (supplementary material).
Hence, derivative (8) might have induced ROS production in the MCF-7 cell line because the cell is vulnerable to ROS-induced apoptosis while exhibiting another form of cell death independent from ROS in AMJ13; what type of death remains to be elucidated with further investigations. It is valuable to say that this is not the first time that a thiosemicarbazide derivative induces cell death that is neither apoptotic nor necrotic without ROS production. Cavazzoni and his colleagues witnessed the same behaviour from their thiosemicarbazone derivative (referred to as copper(II) complex 5) [48]. Hager et al. [45] also documented a paraptotic cell death with their compound Me2NNMe2 that elicited its cytotoxicity independently from ROS generation, rather it was associated with an increased level of the oxidised form of glutathione.
It is noteworthy that this cellular-dependent activity of derivative (8) was also witnessed in previous investigations of ours as an antiangiogenic agent. The derivative proved to be a promising antiangiogenic agent targeting multiple factors in the angiogenesis of breast cancer cells in particular the MCF-7 cell line with a different effect on AMJ13 cell lines [49].
In conclusion, this study has demonstrated the thiosemicarbazide derivative of captopril (8) as a very promising molecule against breast cancer cell lines. The derivative displayed a highly improved antiproliferative activity than captopril against either cell, the ER receptor positive MCF-7 and the highly aggressive ER/PR receptor negative AMJ13 breast cancer cells. The derivative caused an apoptotic death with the potential implication of caspase- 3 and caspase -9 along with time-dependent ROS generation in MCF-7, and an extensive vacuole forming, non-apoptotic cellular death, without ROS generation, but with significant implication of caspase-9 in AMJ13. Whether this cellular-dependent behaviour of the derivative is related to the distinct characteristics of each cell line or is based on its versatility of targets that may or may not involve RAS components, the derivative proved to be a promising antiproliferative agent against breast cancer cells and is certainly recommended as a leading molecule in future investigations.
Limitations
The study could have included more detailed techniques regarding the two types of death, such as Western blot, TEM, flow cytometery. Lack of the availability of these techniques at the time of commencing the investigations, greatly limited that possibility. Currently, these techniques are available and the proceeding studies of these derivatives will include those techniques.
Author Contribution Statement
Conceptualization, Baan AL-Jasani and Hiba AL- Saad; Data curation, Baan AL-Jasani; Formal analysis, Baan AL-Jasani; Investigation, Baan AL-Jasani; Methodology, Baan AL-Jasani; Project administration, Baan AL-Jasani and Hayder Sahib; Resources, Baan AL-Jasani and Hiba AL- Saad; Supervision, Hayder Sahib; Validation, Hayder Sahib; Visualization, Baan AL-Jasani, Hayder Sahib and Hiba AL- Saad; Writing – original draft, Baan AL-Jasani; Writing – review & editing, Baan AL-Jasani
Acknowledgements
The authors acknowledge the support from the Department of Experimental Therapy, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriyah University for kindly providing the cell lines.
Ethical declaration
Ethical clearance was granted from all the required institutions and laboratories
Data Availability Statement
Data is within the article and can be provided by the corresponding author upon reasonable request
Conflicts of Interest
The authors declare no conflict of interest
Supplementary materials
References
- 1.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- 2.Makki J. Diversity of breast carcinoma: Histological subtypes and clinical relevance. Clin Med Insights Pathol. 2015;8:23–31. doi: 10.4137/CPath.S31563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Burguin A, Diorio C, Durocher F. Breast cancer treatments: Updates and new challenges. J Pers Med. 2021;11(8):808. doi: 10.3390/jpm11080808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Waks AG, Winer E P. Genetic testing and counseling for hereditary breast cancer in the era of panel testing and multigene panels. JAMA Oncol. 2021;7:204–11. [Google Scholar]
- 5.Wegman-Ostrosky T, Soto-Reyes E, Vidal-Millan S, Sanchez-Corona J. The renin-angiotensin system meets the hallmarks of cancer. J Renin Angiotensin Aldosterone Syst. 2015;16(2):227–33. doi: 10.1177/1470320313496858. [DOI] [PubMed] [Google Scholar]
- 6.Vinson GP, Barker S, Puddefoot JR. The renin-angiotensin system in the breast and breast cancer. Endocr Relat Cancer. 2012;19(1):R1–19. doi: 10.1530/ERC-11-0335. [DOI] [PubMed] [Google Scholar]
- 7.Almutlaq M, Alamro AA, Alamri HS, Alghamdi AA, Barhoumi T. The effect of local renin angiotensin system in the common types of cancer. Front Endocrinol (Lausanne) 2021;12:736361. doi: 10.3389/fendo.2021.736361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pinter M, Kwanten WJ, Jain RK. Renin-angiotensin system inhibitors to mitigate cancer treatment-related adverse events. Clin Cancer Res. 2018;24(16):3803–12. doi: 10.1158/1078-0432.CCR-18-0236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Pinter M, Jain RK. Targeting the renin-angiotensin system to improve cancer treatment: Implications for immunotherapy. Sci Transl Med. 2017;9(410):eaan5616. doi: 10.1126/scitranslmed.aan5616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bhardwaj G. How the antihypertensive losartan was discovered. Expert Opin Drug Discov. 2006;1(6):609–18. doi: 10.1517/17460441.1.6.609. [DOI] [PubMed] [Google Scholar]
- 11.Small W Jr, Molteni A, Kim YT, Taylor JM, Ts’ao CH, Ward WF. Mechanism of captopril toxicity to a human mammary ductal carcinoma cell line in the presence of copper. Breast Cancer Res Treat. 1999;55(3):223–9. doi: 10.1023/a:1006233521325. [DOI] [PubMed] [Google Scholar]
- 12.Napoleone E, Cutrone A, Cugino D, Amore C, Di Santo A, Iacoviello L, et al. Inhibition of the renin-angiotensin system downregulates tissue factor and vascular endothelial growth factor in human breast carcinoma cells. Thromb Res. 2012;129(6):736–42. doi: 10.1016/j.thromres.2011.11.047. [DOI] [PubMed] [Google Scholar]
- 13.Cho WK, Shin SW, Kim SY, Hong CW, Choi C, Park W, et al. Immunomodulatory effect of captopril and local irradiation on myeloid-derived suppressor cells. Radiat Oncol J. 2016;34(3):223–9. doi: 10.3857/roj.2016.01816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Al-Saad HN, Mahmood AAR, Al-Bayati RIJOjoc. Design, synthesis, docking study and antiplatelet evaluation of new thiosemicarbazide derivatives derived from captopril. Orient J Chem. 2019;35(2):829. [Google Scholar]
- 15.Al-Saad HN, Kubba AAJRJoP. Evaluation of new thiosemcarbazides derived from captopril as angiotensin-converting enzyme inhibitors with docking study, and predicted-admet analysis. Res J Pharm Technol. 2020;13(6):2733–41. [Google Scholar]
- 16.Kucukguzel SG, Coskun GP. Macromolecular drug targets in cancer treatment and thiosemicarbazides as anticancer agents. Anticancer Agents Med Chem. 2016;16(10):1288–300. doi: 10.2174/1871520616666160219160256. [DOI] [PubMed] [Google Scholar]
- 17.Dincel ED, Guzeldemirci NUJM. Synthesis and computer-aided drug design studies of novel thiosemicarbazide derivatives as potent and target-oriented anti-cancer agents. Int J Med Sci. 2020;9(2):305–13. [Google Scholar]
- 18.Finch RA, Liu MC, Cory AH, Cory JG, Sartorelli AC. Triapine (3-aminopyridine-2-carboxaldehyde thiosemicarbazone; 3-ap): An inhibitor of ribonucleotide reductase with antineoplastic activity. Adv Enzyme Regul. 1999;39(1):3–12. doi: 10.1016/s0065-2571(98)00017-x. [DOI] [PubMed] [Google Scholar]
- 19.Ertas M, Sahin Z, Bulbul EF, Bender C, Biltekin SN, Berk B, et al. Potent ribonucleotide reductase inhibitors: Thiazole-containing thiosemicarbazone derivatives. Arch Pharm (Weinheim) 2019;352(11):e1900033. doi: 10.1002/ardp.201900033. [DOI] [PubMed] [Google Scholar]
- 20.Wei L, Easmon J, Nagi RK, Muegge BD, Meyer LA, Lewis JS. 64cu-azabicyclo[322]nonane thiosemicarbazone complexes: Radiopharmaceuticals for pet of topoisomerase ii expression in tumors. J Nucl Med. 2006;47(12):2034–41. [PubMed] [Google Scholar]
- 21.Malki A, Elbayaa RY, Ashour HM, Loffredo CA, Youssef AM. Novel thiosemicarbazides induced apoptosis in human mcf-7 breast cancer cells via jnk signaling. J Enzyme Inhib Med Chem. 2015;30(5):786–95. doi: 10.3109/14756366.2014.971781. [DOI] [PubMed] [Google Scholar]
- 22.El Majzoub R, Fayyad-Kazan M, Nasr El Dine A, Makki R, Hamade E, Gree R, et al. A thiosemicarbazone derivative induces triple negative breast cancer cell apoptosis: Possible role of mirna-125a-5p and mirna-181a-5p. Genes Genomics. 2019;41(12):1431–43. doi: 10.1007/s13258-019-00866-y. [DOI] [PubMed] [Google Scholar]
- 23.Siwek A, Stączek P, Wujec M, Bielawski K, Bielawska A, Paneth PJJomm. Cytotoxic effect and molecular docking of 4-ethoxycarbonylmethyl-1-(piperidin-4-ylcarbonyl)-thiosemicarbazide—a novel topoisomerase ii inhibitor. J Mol Model. 2013;19(3):1319–24. doi: 10.1007/s00894-012-1679-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Yee EMH, Brandl MB, Black DS, Vittorio O, Kumar N. Synthesis of isoflavene-thiosemicarbazone hybrids and evaluation of their anti-tumor activity. Bioorg Med Chem Lett. 2017;27(11):2454–8. doi: 10.1016/j.bmcl.2017.04.002. [DOI] [PubMed] [Google Scholar]
- 25.Sólimo AM, Santacruz MS, Vanzulli S, Coggiola O, de Kier Joffé EB, Finkielsztein L, et al. Anti-metastatic action of an n4-aryl substituted thiosemicarbazone on advanced triple negative breast cancer. Heliyon. 2020;6(10):e05161. doi: 10.1016/j.heliyon.2020.e05161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Afrasiabi Z, Stovall P, Finley K, Choudhury A, Barnes C, Ahmad A, et al. Targeting triple negative breast cancer cells by n3-substituted 9,10-phenanthrenequinone thiosemicarbazones and their metal complexes. Spectrochim Acta A Mol Biomol Spectrosc. 2013;114:114–9. doi: 10.1016/j.saa.2013.04.122. [DOI] [PubMed] [Google Scholar]
- 27.Bai C, Wu S, Ren S, Zhu M, Luo G, Xiang H. Synthesis and evaluation of novel thiosemicarbazone and semicarbazone analogs with both anti-proliferative and anti-metastatic activities against triple negative breast cancer. Bioorg Med Chem. 2021;37:116107. doi: 10.1016/j.bmc.2021.116107. [DOI] [PubMed] [Google Scholar]
- 28.Arrieta O, Villarreal-Garza C, Vizcaino G, Pineda B, Hernandez-Pedro N, Guevara-Salazar P, et al. Association between at1 and at2 angiotensin ii receptor expression with cell proliferation and angiogenesis in operable breast cancer. Tumour Biol. 2015;36(7):5627–34. doi: 10.1007/s13277-015-3235-3. [DOI] [PubMed] [Google Scholar]
- 29.Al-Shammari AM, Alshami MA, Umran MA, Almukhtar AA, Yaseen NY, Raad K, et al. Establishment and characterization of a receptor-negative, hormone-nonresponsive breast cancer cell line from an iraqi patient. Breast Cancer (Dove Med Press) 2015;7:223–30. doi: 10.2147/BCTT.S74509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sylvester PW. Optimization of the tetrazolium dye (MTT) colorimetric assay for cellular growth and viability. Drug Design and Discovery: Methods and Protocols. 2011:157–68. doi: 10.1007/978-1-61779-012-6_9. [DOI] [PubMed] [Google Scholar]
- 31.Zhang L, Zheng Q, Yang Y, Zhou H, Gong X, Zhao S, et al. Synthesis and in vivo sar study of indolin-2-one-based multi-targeted inhibitors as potential anticancer agents. Eur J Med Chem. 2014;82:139–51. doi: 10.1016/j.ejmech.2014.05.051. [DOI] [PubMed] [Google Scholar]
- 32.Li RD, Wang HL, Li YB, Wang ZQ, Wang X, Wang YT, et al. Discovery and optimization of novel dual dithiocarbamates as potent anticancer agents. Eur J Med Chem. 2015;93:381–91. doi: 10.1016/j.ejmech.2015.02.030. [DOI] [PubMed] [Google Scholar]
- 33.Van Meerloo J, Kaspers GJ, Cloos J. Cell sensitivity assays: the MTT assay. Cancer cell culture: methods and protocols. Springer; 2011. pp. 237–45. [DOI] [PubMed] [Google Scholar]
- 34.Vorster C, Joubert A. In vitro effects of 2-methoxyestradiol-bis-sulphamate on cell growth, morphology and cell cycle dynamics in the mcf-7 breast adenocarcinoma cell line. Biocell. 2010;34(2):71–9. [PubMed] [Google Scholar]
- 35.Kareem SH, Naji AM, Taqi ZJ, Jabir MSJJoI. Polyvinylpyrrolidone loaded-mnznfe2o4 magnetic nanocomposites induce apoptosis in cancer cells through mitochondrial damage and p53 pathway. J Inorg Organomet Polym. 2020;30(12):5009–23. [Google Scholar]
- 36.Oberska P, Jedrzejczak-Silicka M, Michalek K, Grabowska M. Initial assessment of suitability of mcf-7 and hepg2 cancer cell lines for aqp3 research in cancer biology. Acta Histochem. 2021;123(4):151716. doi: 10.1016/j.acthis.2021.151716. [DOI] [PubMed] [Google Scholar]
- 37.Coskun GP, Djikic T, Hayal TB, Turkel N, Yelekci K, Sahin F, et al. Synthesis, molecular docking and anticancer activity of diflunisal derivatives as cyclooxygenase enzyme inhibitors. Molecules. 2018;23(8):1969. doi: 10.3390/molecules23081969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sperandio S, de Belle I, Bredesen DE. An alternative, nonapoptotic form of programmed cell death. Proc Natl Acad Sci U S A. 2000;97(26):14376–81. doi: 10.1073/pnas.97.26.14376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sadat Shandiz SA, Montazeri A, Abdolhosseini M, Hadad Shahrestani S, Hedayati M, Moradi-Shoeili Z, et al. Functionalization of ag nanoparticles by glutamic acid and conjugation of ag@ glu by thiosemicarbazide enhances the apoptosis of human breast cancer mcf-7 cells. J Clust Sci. 2018;29(6):1107–14. [Google Scholar]
- 40.Haribabu J, Balachandran C, Tamizh MM, Arun Y, Bhuvanesh NS, Aoki S, et al. Unprecedented formation of palladium (ii)-pyrazole based thiourea from chromone thiosemicarbazone and [pdcl2 (pph3) 2]: Interaction with biomolecules and apoptosis through mitochondrial signaling pathway. J Inorg Biochem. 2020;205:110988. doi: 10.1016/j.jinorgbio.2019.110988. [DOI] [PubMed] [Google Scholar]
- 41.De Bortoli M, Taverna E, Maffioli E, Casalini P, Crisafi F, Kumar V, et al. Lipid accumulation in human breast cancer cells injured by iron depletors. J Exp Clin Cancer Res. 2018;37(1):75. doi: 10.1186/s13046-018-0737-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hancock CN, Stockwin LH, Han B, Divelbiss RD, Jun JH, Malhotra SV, et al. A copper chelate of thiosemicarbazone nsc 689534 induces oxidative/er stress and inhibits tumor growth in vitro and in vivo. Free Radic Biol Med. 2011;50(1):110–21. doi: 10.1016/j.freeradbiomed.2010.10.696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bisceglie F, Alinovi R, Pinelli S, Galetti M, Pioli M, Tarasconi P, et al. Autophagy and apoptosis: Studies on the effects of bisthiosemicarbazone copper(ii) complexes on p53 and p53-null tumour cell lines. Metallomics. 2016;8(12):1255–65. doi: 10.1039/c6mt00170j. [DOI] [PubMed] [Google Scholar]
- 44.Hager S, Pape VFS, Posa V, Montsch B, Uhlik L, Szakacs G, et al. High copper complex stability and slow reduction kinetics as key parameters for improved activity, paraptosis induction, and impact on drug-resistant cells of anticancer thiosemicarbazones. Antioxid Redox Signal. 2020;33(6):395–414. doi: 10.1089/ars.2019.7854. [DOI] [PubMed] [Google Scholar]
- 45.Hager S, Korbula K, Bielec B, Grusch M, Pirker C, Schosserer M, et al. The thiosemicarbazone me(2)nnme(2) induces paraptosis by disrupting the er thiol redox homeostasis based on protein disulfide isomerase inhibition. Cell Death Dis. 2018;9(11):1052 . doi: 10.1038/s41419-018-1102-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhang J, Zhang Z, Jiang M, Li S, Yuan H, Sun H, et al. Developing a novel gold(iii) agent to treat glioma based on the unique properties of apoferritin nanoparticles: Inducing lethal autophagy and apoptosis. J Med Chem. 2020;63(22):13695–708. doi: 10.1021/acs.jmedchem.0c01257. [DOI] [PubMed] [Google Scholar]
- 47.Trejo-Solis C, Jimenez-Farfan D, Rodriguez-Enriquez S, Fernandez-Valverde F, Cruz-Salgado A, Ruiz-Azuara L, et al. Copper compound induces autophagy and apoptosis of glioma cells by reactive oxygen species and jnk activation. BMC Cancer. 2012;12(1):156 . doi: 10.1186/1471-2407-12-156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Rogolino D, Cavazzoni A, Gatti A, Tegoni M, Pelosi G, Verdolino V, et al. Anti-proliferative effects of copper(ii) complexes with hydroxyquinoline-thiosemicarbazone ligands. Eur J Med Chem. 2017;128:140–53. doi: 10.1016/j.ejmech.2017.01.031. [DOI] [PubMed] [Google Scholar]
- 49.Baan M, AL-Jasani HBS, Hiba N Al-Saad. The anti-angiogenic potential of thiosemicarbazide derivative of captopril (8) in breast cancer cell lines. Jordan J Biol Sci. 2024;17(3):423–33. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Data is within the article and can be provided by the corresponding author upon reasonable request







