Next Article in Journal
Nanocarrier Systems in Taste Masking
Previous Article in Journal
Purple Corn Silk Extract Attenuates UVB-Induced Inflammation in Human Keratinocyte Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Application of Quality by Design Approach to the Pharmaceutical Development of Anticancer Crude Extracts of Crocus sativus Perianth

by
Olha Mykhailenko
1,*,
Liudas Ivanauskas
2,
Ivan Bezruk
1,
Vilma Petrikaitė
3,4 and
Victoriya Georgiyants
1
1
Department of Pharmaceutical Chemistry, National University of Pharmacy, 61168 Kharkiv, Ukraine
2
Department of Analytical and Toxicological Chemistry, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
3
Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
4
Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
*
Author to whom correspondence should be addressed.
Sci. Pharm. 2022, 90(1), 19; https://doi.org/10.3390/scipharm90010019
Submission received: 21 January 2022 / Revised: 16 February 2022 / Accepted: 1 March 2022 / Published: 3 March 2022

Abstract

:
The application of the Quality by Design (QbD) concept to extracts obtained from Crocus sativus perianth with potential anticancer activity will ensure the safety, efficiency, and quality control of the entire technological process, as well as determine the critical factors affecting the quality of extracts. Potentially critical points of the production of the plant extracts, including the cultivation and processing of the plant materials, the extraction process, and the choice of solvents, were identified using the Ishikawa diagram and FMEA risk assessment methods as well as the corrective actions proposed. The Herbal Chemical Marker Ranking System (HerbMars) approach was used to justify the Q-markers choice of Crocus, which takes into account bioavailability, pharmacological activity, and the presence of the selected standard. An experimental design (DoE) was used to assess the influence of potentially critical factors on the efficiency of the compound extraction from raw materials with water or ethanol. The presence of 16 compounds in Crocus perianth was determined by HPLC and their quantitative assessment was established. Selected compounds (ferulic acid, mangiferin, crocin, rutin, isoquercitrin) can be used for the quality control of Crocus perianth. In addition, the stigmas from the Volyn region met the requirements of ISO 3632 for saffron as a spice (category I). The cytotoxic activity against melanoma (IGR39) and triple-negative breast cancer (MDA-MB-231) cell lines of the hydroethanolic extract of C. sativus perianth was significantly more pronounced than the water extract, probably due to the chemical composition of the constituent components. The results show that the QbD approach is a powerful tool for process development for the production of quality herbal drugs.

1. Introduction

Despite the increasing popularity of herbal drugs, there are a large number of reviews [1,2,3] describing the adverse effects on the patient’s health due to changes in the quality, effectiveness, and content of biologically active compounds in them. Herbal drugs, like any drug, must meet quality, safety, and efficacy requirements. The implementation of the ICH Q8, Q9, and Q10 guidelines has combined the stages of pharmaceutical product development, where quality assurance of the medicinal product is laid down at each stage of production by the requirements of Good Manufacturing Practice (GMP). In this regard, the World Health Organization (WHO) has developed several guidelines for the standardization steps of herbal products [4]. The Quality by Design (QbD) concept is a part of the ICH Q8 (R2) “Pharmaceutical Development” guideline and ensures product quality by modeling and controlling the manufacturing process itself, and not just relying on the final product at the end of the process to be the quality test [5,6]. This approach allows the development of reliable, controlled processes and operations, which results in a quality product. The methodological approach based on the QbD was applied in the current work to plan the process of obtaining plant extracts from Crocus sativus perianth with anticancer effect.
Saffron is one of the most valuable spices in the world, which is cultivated in several countries for the food industry. Saffron, in filaments, is the dried dark-red stigmas of the C. sativus (Iridaceae family plant) flower. The stigmas are separated from the perianth, and all other parts of the flower are a by-product [7]. It takes approximately 150,000 flowers to produce 1 kg of Crocus stigma. However, Crocus perianth contains various biologically active compounds and can be considered as a potential medicinal raw material. A review of scientific articles showed that C. sativus flower extracts have different pharmacological activities: antibacterial (125 mg/mL) [8,9], antioxidant, and free radical scavenging properties [10,11]; is antiproliferative (ED50 0.42 mg/mL) against Caco-2 cells [12]; antityrosinase activity (50 µg/mL); individual compounds, such as crocin 1 and safranal have inhibitory activity against human monoamine oxidases [13]; etc. Saffron, as a spice, is also grown in the Ukraine [14], where there is a large amount of raw perianth after Crocus stigma processing.
Since Crocus raw material from the Ukraine is a new source for the production of herbal drugs, it is necessary to evaluate its chemical composition. The chemical composition of herbal raw materials is usually variable and depends on environmental and technological factors, so it is important to ensure control during the production of the product. Harvesting, drying, storage, transportation, and processing methods (e.g., the extraction method and the polarity of the extracting solvent, component instability, etc.) have a particular impact on the quality of the starting herbal raw material [15]. However, for C. sativus raw material standardization, parameters have not been developed. Assessment of the quality control of herbal raw materials, extracts, and drugs based on active markers is mandatory. Quality markers are “chemically defined constituents or groups of constituents of an herbal substance, an herbal preparation or herbal medicinal product that serve for quality control purposes, independent of whether they have any therapeutic activity”. The EMA describes two different categories of chemical/quality markers. The constituents of an herbal medicine responsible for its therapeutic activity or active markers and the constituents that are characteristic for its taxon or analytical markers [16].
The Herbal Chemical Marker Ranking System [17] was proposed for the selection of quality markers (Q-markers) for herbal raw materials and preparations. This approach takes into account the bioavailability, the reported bioactivity, the quantitative content of the metabolite, and its physiological effects associated with the intended use of the raw material, as well as the commercial availability of the standard. It is important for the Q-marker to be able to trace it throughout the entire production process, from raw materials, then obtained extracts, to finished products.
Thus, the current work aimed was to apply the QbD concept to experimental design; analysis of current processes and risk management to ensure the quality of obtained plant extracts; determination of the most appropriate chemical markers for quality control of C. sativus perianth and extracts; development of a quality assessment method for multicomponent analysis; fingerprinting of C. sativus; assessment of the antioxidant and cytotoxic potential of the obtained extracts.

2. Results

2.1. Used of QbD Approach

The “Quality by Design” (QbD) concept originated in the field of quality management and has recently been applied to the process planning and manufacturing of pharmaceuticals [18]. It is defined in ICH Q8 Pharmaceutical Development as a systematic approach to development that starts with predetermined aims and focuses on product and process understanding and process management based on sound science and quality risk management. The European Medicines Agency (EMA) [6,16] adapted the QbD approach to improving understanding of the herbal drugs manufacturing process. However, despite the advantages of this modeling, the QbD approach has not yet been fully implemented at the planning and manufacturing stage of drugs. The philosophy and approach of QbD were applied to optimize the targeted search for anticancer compounds on the example of obtaining extracts from Crocus perianth.
The general workflow for the development of an herbal extract based on the application of the QbD approach, adapted for Crocus perianth, is shown in Figure 1. This approach guarantees a reliable process from the selection of herbal raw materials, extraction for herbal drug production, even if the starting material of herbal origin has a wide variety, which is typical for plant materials.
According to the QbD approach, pharmaceutical development should include at least the following elements and phases:
determination of the desired characteristics or the Quality Target Product Profile (QTPP);
identification of potentially Critical Quality Attributes of a drug (CQAs);
determination of possible Critical Process Parameters (CPPs) and the pharmaceutical substance characteristics and excipients (Critical Material Attributes (CMAs));
development and implementation of Design of Experiment (DoE), its optimization, the definition of control strategies, and improvements.
To develop the desired herbal drug substance by the ICH Q8 guidelines, the “Quality Target Product Profile” was compiled. It is based on quality indicators for anticancer herbal preparations, namely, high efficiency, low toxicity, and the possibility of long-term use in therapy [4,20,21,22]. Herbal compounds have a positive effect on the survival, immunomodulation, and quality of life of cancer patients, in combination with traditional therapeutic agents. The main compounds of Crocus flowers are phenolic compounds [7,10,11], which exhibit pronounced antioxidant properties. Therefore, the ultimate aim of the process is to obtain a high-quality plant extract from Crocus perianth with pronounced anticancer activity. To ensure the properties specified in the QTPP, a method was chosen to obtain extracts with different solvents to determine the most suitable extractant based on the results of chromatographic analysis.
Further, the Critical Quality Attributes (CQA) of the selected product [19] should be determined, which provide its desired quality with proper control during the production process. CQAs include physical, chemical, biological, or microbiological properties or characteristics of the starting plant material as well as the obtained extract. Following the QbD approach, CQAs are dynamic characteristics of the process development that need to be updated and improved during the product life cycle [5]. A risk analysis is used to identify points of process control to determine the CQAs. These CQAs are designed to obtain standard plant raw materials by complying with the GACP recommendations for plant cultivation, which will provide the traceability of herbal raw materials and guarantee a stable result of pharmacological activity. In addition, the critical points of the process are the quality control of raw materials, auxiliary materials, and the extraction process to ensure the plant extract is of consistent quality and composition.

2.2. Risk Assessment of Crocus Perianth Extracts Production

Risk assessment is part of risk management and should be conducted early in process development. Once the QTPP has been defined and the CQAs have been identified, the risk assessment and design space identification can be obtained either through experimentation or modeling. The guideline ICH Q9 “Quality Risk Management” provides a list of nine common risk management tools, namely, basic risk management facilitation methods (Ishikawa fishbone diagram, flowcharts, check sheets, etc.); fault tree analysis; risk ranking and filtering; preliminary hazard analysis; hazard analysis and critical control points; failure mode and effects analysis (FMEA); failure mode, effects, and criticality analysis (FMECA); hazard operability analysis; supporting statistical tools. According to the QbD conception, risk assessment has priority over DoE. Among the most commonly used tools are Ishikawa’s fishbone diagram and FMEA [23].
An Ishikawa diagram identifies and groups different kinds of effects, such as material properties, equipment design, and process parameters, that may pose a risk to certain CQAs, such as yield, purity, or overall processability [4,21,22]. The main branches of the fishbone are subdivided into sub-branches that reveal a more detailed causal relationship between potential cause and risk. This diagram may already identify critical process parameters (CPPs) that must be maintained within a certain range during the process and, therefore, must be part of a process control strategy and may require further study. An approximate early risk assessment for the procedure for obtaining plant extracts with a stable composition of components from Crocus perianth is shown in Figure 2.
The main risks of plant materials obtained with a stable chemical composition are the biological and physiological aspects of plant cultivation [24]. The environment during the cultivation and harvesting can lead to very different compositions of active compounds. In this regard, WHO proposed to adhere to the GACP recommendations on the possibility of traceability and obtaining the quality of starting raw material of plant origin [25]. In a previous study, it was shown that the C. sativus plant in the Kherson region, Ukraine, is grown in compliance with the developed and implemented Standard Operating Procedures for this crop, taking into account the climatic characteristics of the country [14]. Primary processing also has norms and requirements. A similar approach to cultivating saffron was used in the farm of the Volyn region, which guarantees the receipt of high-quality raw materials at the first stage of development. As a result of processing Crocus flowers and extracting its stigmas, a large mass of perianth is formed, which is production waste [12].
It can be seen that six main reasons have been identified in the current experiment, including the starting raw material of plant origin, primary processing of raw materials, extraction conditions, equipment, and the availability of trained personnel and associated sub-reasons. The first step is cultivating the plant in compliance with the GACP principles and primary processing of raw materials, then preparing the raw materials for extraction and conducting analysis to establish its chemical composition by HPLC. The impact of human error, equipment, and the environment on quality can be reduced through effective management and by adhering to standard operating procedures or maintenance schedules. The risks associated with the process and materials, in this case, obtaining quality feedstock and the process of selecting an extractant, are the most important for the characterization of the process [26].
The FMEA was used to quantify specific risks. FMEA, for the development of the Crocus perianth plant extract production process, is derived by evaluating the range of CPP defined by the Ishikawa diagram for the potential impact of risks that could affect quality indicators and the possibility of risks during the extraction of plant materials with solvents [27,28]. The preparation of the extract consists of several stages of primary processing, drying, grinding, extraction, and the actual obtainment of dry extracts from Crocus (Table 1). Analysis of failure mode and effect in Crocus extract production helps to prioritize risks and determine corrective actions to avoid identified problems.
The results of calculating the final risk priority number (RPN) made it possible to classify possible inconsistencies in the technological process at the extraction stage as unacceptable risks. The risks arising at the stage of raw material preparation, purification, and extraction of extracts have a significant impact. In the process of risk management, the risk level was assessed and methods of risk control and prevention were proposed.
The Hazard Analysis and Critical Control Points System (HACCP) helps to identify key processing steps of plant materials to minimize the risk of microbial contamination and should be performed by default. HACCP implementation risk analysis, integrated with FMEA, helped in the current study to predict the presence of probable hazards, including biological (microorganism, enzymatic activity), chemical (pesticides residues, mycotoxins), and physical (heavy metals, foreign matter, critical moisture, browning, excessive water activity, dust) that can occur at different stages of the process. In the production of Crocus perianth extracts, the primary processing stage of herbal raw materials, including harvesting, sorting, and drying, no matter what type of drying process is used, is considered CCP. In addition, the extraction process is also a defining stage of production. Thus, the production of herbal drugs should be carried out in compliance with the GMP, GACP, and HACCP guidelines with strict control of all types of risks.

2.3. Design of Experiment

The aim of the DoE was to gain a deeper understanding of the processes and planning of the current experiment (Figure 3). The plan of the experiment included: defining the problem (the current problem of cancer diseases in the world), choosing the direction of solving the problem (searching for herbal preparations with anticancer activity), then justifying the choice of the object of research (Crocus perianth–selection of herbal materials with a wide raw material base and a promising composition of compounds according to the literature search). Further, the definition and selection of quality markers for the possible quality control of the starting plant material, obtained plant extracts, and, subsequently, the herbal drugs were carried out. Therefore, we applied the Herb MaRS approach to select potential quality markers specifically for Crocus perianth extracts with anticancer properties. After that, the DoE provides for the chemical analysis of raw materials and obtaining extracts from the selected type of raw materials and their chemical analysis. As the final stage of the experiment, pharmacological tests of the obtained extracts for the presence of antioxidant and cytotoxic activity were carried out.
The study also included the determination of the quality of Crocus stigmas in accordance with ISO 3632. The quality of saffron depends on the concentration of the main metabolites, crocin, picrocrocin, and saffron, which are responsible for the color, flavor, and aroma of the spice. The availability of high-quality raw materials of Crocus stigma motivates farmers to expand the areas of cultivation and production of the spice itself. As a by-product of the technological process, a large biomass of perianth is formed, which is the main object of study in the current work. In this regard, farming in the Volyn region is very promising. The area of the plantation is almost 4 hectares. Results of crocins ( E 1 c m 1 % 440 nm was 253), safranal ( E 1 c m 1 % 330 nm was 30), and picrocrocin ( E 1 c m 1 % 257 was 95) amounts and moisture content (9.5%) of saffron showed that the tested sample fulfilled the ISO specifications for category I regarding moisture and the main spectrophotometric characteristics.

2.4. Selection of Crocus Perianth Q-Markers with the Herb MaRS-Approach

The choice of chemical markers is critical for the quality control of herbal raw material and drugs, as well as for identification and authentication. The most correct are therapeutic components (active compounds), that is, those components of the plant that determine its pharmacological activity. For plant materials, as a rule, there are several quality markers since the substances act synergistically. The choice of the chemical markers for quality assurance of the herbal raw material and crude extracts of Crocus perianth was made by using the Herbal Chemical Marker Ranking System (Herb MaRS) based on anticancer bioactivity against melanoma (IGR39) and triple-negative breast cancer (MDA-MB-231) cell lines. The innovative Herb MaRS procedure, for the identification of relevant chemical markers in complex medicinal plants, was developed by the National Institute of Complementary Medicine [17] and takes into account the bioavailability of the compound, the declared bioactivity and physiological effect associated with its intended use (in this experiment—anticancer), as well as the availability of a reference compound for analysis (Figure 4). In this work, the Herb MaRS approach was applied not to the collection from different herbs, but to one type of raw material (Crocus perianth) and to its various compounds. This approach was used to select quality markers based precisely on the approach of the system. Based on the Herb MaRS criteria, 19 compounds were selected, which were found in the raw material of C. sativus according to the various literature data [32,33,34].
The main components of Crocus stigma are crocins, safranal, and picrocrocin. They were also found by various authors in Crocus flowers [10,35]. In addition, cinnamic acid derivatives [35] and different flavonoids and their derivatives [10,12,34,36,37] have been found in various parts of Crocus (flowers, leaves, stigma). The qualitative and quantitative composition of phenolic compounds differ depending on the cultivation place and harvesting of Crocus raw materials. In addition, Table 2 shows isoflavonoids (nigricin, iristectorigenin B, tectorigenin), which are characteristic of plants of the Iridaceae family [38] and have previously been found in the leaves and stigmas of Crocus stigmas [39,40]. The compounds presented in Table 2 can be considered as potential Q-markers of C. sativus perianth in terms of relative importance in the treatment of cancer, provided they are found in the studied samples. In addition, all discussed compounds are available standards.

2.4.1. Traditional Saffron Used

Saffron is used in folk medicine in different countries for the treatment of cancer, eye diseases, disorders of the uneven system, and for normalizing metabolism [107]. Saffron has been used as a remedy for various diseases, including cancer, in ancient Arabic, Indian, and Chinese cultures. Saffron has traditionally been used for convulsions, asthma and bronchospasm, menstrual irregularities, liver disease, and pain. However, the main application was as a stimulant, aphrodisiac, and antidepressant [33]. As a homeopathic remedy, saffron is also presented in various pharmacopoeias of the world [108]. During the production of saffron, large masses of perianths are formed, which are a by-product of the production of spice. As a result of the analysis of the chemical composition of the perianth, as well as extracts from it, the presence of various compounds was established. The relevant compounds identified in the Crocus raw material are shown in Table 2 along with their activity and the corresponding Herb MaRS score.

2.4.2. Current Pharmacology Application of Compounds

The anticancer activity of flavonoids in vitro, as well as their chemopreventive potential in vivo, have been long known. For example, antitumor activity against MDA-MB-231 cells [70], as well as melanoma, leukemia, and erythroleukemia for rutin, kaempferol, and quercetin has been previously documented [74,93,94]. Rutin (20 μM) significantly (p < 0.05) increased the cytotoxic activity in MDA-MB-231 cells of the chemotherapeutic agent’s cyclophosphamide and methotrexate. In another study, rutin at a dose of 30 mg/kg significantly reduced the growth of TNBC MDA-MB-231/GFP cells [75]. Administration of rutin at a dose of 10 mg inhibited the formation of B16 melanotic melanoma in C57BL/6 mice by more than 40% [71].
Crocins are the main chemical constituents of Crocus stigma. However, crocins are also found in Crocus flowers. Several studies have shown that crocins exhibit antitumor effects on various cancer cell lines [45,46]. Crocetin, trans-crocin-4, and safranal were reported to significantly inhibit the proliferation effect against breast cancer MDA-MB-231 (200 µg/mL) [47]. Crocin-induced apoptosis and cell cycle arrest in the G2/M phase in MDA-MB-231 cells in a dose-dependent manner (IC50 5.97 mg/mL) [48]. For the Crocus stigma water extract, an antitumor effect was revealed in vivo on a highly metastatic murine B16-F10 melanoma cell line [49]. There are studies [50] on breast cancer cells MCF-7 and MDA-MB-231 showing the concentration-dependent inhibition of proliferation by crocetin, the main metabolite of crocins. Crocin is suggested to be one of the most effective components of saffron for cancer treatment.
Apigenin at concentrations of 25 μM, 50 μM, 75 μM, and 100 μM inhibited the viability of MDA-MB-231 cell by 12%, 27%, 42%, and 49%, respectively [81]. A potent antiproliferative effect of apigenin was shown against the human melanoma A375 cell line (EC50 33.02 μM) [82]. In our previous experiment, apigenin showed the highest activity against melanoma IGR39 and breast cancer MDA-MB-231 cell lines (EC50 values were 131.8 ± 7.2 µM and 123.4 ± 19.0 µM, respectively) [39]. In other studies, apigenin-7-O-glucoside showed a more cytotoxic effect on colon cancer HCT116 cells compared to apigenin [83]. These data demonstrate that the sugar moiety of apigenin-7-O-glucoside has an important effect on the biological activity of apigenin.
Ferulic acid showed antitumor activity in various types of cancer such as colon and lung cancers, as well as tumors of the central nervous system. The authors showed a pronounced cytotoxic activity of ferulic acid in the MDA-MB-231 breast cancer cell line. The use of ferulic acid (3, 10, 30, and 100 µM) led to a decrease in viability, an increase in apoptosis, and suppression of the metastatic potential [54]. In another assay, ferulic acid (at a concentration of 120 μM) showed a pronounced cytotoxic activity against human skin melanoma cells (SK-MEL-3), significantly reducing cell viability compared to the control [56].
The pronounced antitumor potential of mangiferin was established in models of the breast cancer MDA-MB-231 cell line in vitro and in vivo [62]. Thus, mangiferin has immunomodulatory, antioxidant, anti-inflammatory, antiviral, antidiabetic, and anticancer properties [59,61]. Additionally, the anticancer effect of mangiferin has been confirmed in mouse models at doses between 5 and 10 mg/kg against ascitic fibrosarcoma [63], 50 and 100 mg/kg (oral) against benzo(a)pyrene-induced lung carcinogenesis [64,65], and 100 mg/kg (oral) against ER-negative breast cancer.
These and other compounds determine the anticancer potential of C. sativus [12,31]. Therefore, each of these compounds can be considered as a potential Q-marker of activity. The next step of the experiment was to determine the presence of the selected compounds in raw materials and extracts of Crocus perianth from the Ukraine. It is necessary to take into account the quantitative content of each component, its therapeutic activity, and the availability of a reference standard so that it can be proposed as a quality marker according to the chosen approach.

2.5. Assessment of C. sativus Perianth and Its Crude Extract Chemical Composition

2.5.1. Perianth Raw Material

A combined method using HPLC fingerprints and quantitative analysis to assess the stability of the quality of plant compounds was applied to analyze the chemical composition of Crocus perianth and its extracts. Sample pretreatment conditions and HPLC chromatography conditions were first optimized by investigating the effect of extraction solvents, extraction times and methods on extraction efficiency, as well as the influence of the mobile phase and detection wavelength on the efficiency of chromatographic separation of marker compounds. The optimal extraction and chromatographic conditions for Crocus used in this study can be found in the literature [39]. A typical HPLC chromatogram with a satisfactory resolution of 16 chemical markers is shown in Figure 5. The purity of the peaks was determined by comparing the retention times and UV spectra. The combination of a fingerprint with the quantification of analytes is more informative and can be obtained simultaneously. Of the 16 standards used, only 12 were found in the studied samples, and 4 compounds were additionally detected by the recalculation method. In Crocus raw material, flavonoids rutin (160.45 mg/g), apigenin-7-O-glucoside (8.11 mg/g), and isoorientin (7.39 mg/g) were found in the largest amount. Furthermore, mangiferin, isoquercitrin, rutin ferulic acid, as well as isoflavones (tectoridin, nigricin, iristectorignin B) were firstly identified in C. sativus perianth from the Ukraine.
The HPLC method was validated for parameters such as linearity range, LoD, LoQ, accuracy, precision, repeatability (Table S1), and specificity for each analyte. The calibration curve equations of standards along with the significance, LoD, and LoQ values, are presented in Table 3. The regression equation and correlation coefficient ranging from 0.9994 to 0.9999 revealed a good linearity response within the tested ranges. The series of calibration solutions were prepared and separated under the optimal conditions as described above. The LoD and LoQ values indicate that the proposed method demonstrates good sensitivity for the quantitative determination of 16 phenolic compounds in Crocus perianth. The repeatability was observed in the range from 0.23–1.06%, which was satisfactory and indicated the good repeatability of the proposed method. The determination of the main compounds in the test solutions was carried out by comparing the retention times of peaks and the UV spectrum obtained from the chromatogram of the standard solution (Table S2). All results showed repeatability, accuracy, high sensitivity, and good linearity of the method.

2.5.2. Perianth Extracts

Considering that the composition of Crocus active metabolites differs depending on the extractant used, ethanol and hydroethanolic extracts from perianth were obtained (Table 4). All peaks in the chromatograms were identified by comparison with the chromatogram of the mixed reference solution at three wavelengths of 270, 310, and 440 nm. At 440 nm the main saffron component, crocins, were determined. Crocins, as hydrophilic carotenoids, were better extracted in the water extract with an amount of 3.79 mg/g, whereas in the hydroethanolic extract it was only 0.2 mg/g. According to Montoro et al. [10], crocins were also detected in C. sativus perianths by the LC-ESI-MS method in positive ion mode.
Mangiferin was found in both extracts of Crocus perianths. The mangiferin content was higher in the water extract (1.09 mg/g) than in the hydroethanolic extract (0.89 mg/g), due to the presence of a glycoside residue. It is known that plants of the Iris genus (Iridaceae) accumulate various xanthones, mainly C-glycosylxanthones [38]. In the Crocus genus, mangiferin was identified (color reactions) only in C. aureus and C. stellaris leaves [33]. Therefore, mangiferin can be considered as a marker of the family. Taking into account the large mass of perianth waste, this type of raw material can be considered as an alternative to obtaining an individual mangiferin compound with potential antivirus activity.
Previous reports showed the presence of kaempferol and some quercetin glycosides, isoorientin, isoqueritrin, and astragalin in C. sativus tepals extract [7,50]. Flavonoid glycosides are better extracted by water, which we can observe for apigenin-7-O-glucoside and isoorientin (flavone C-glycoside of luteolin), which were identified only in the water extract of the perianth, 2.59 mg/g, 4.76 mg/g, and 0.67 mg/g, respectively. In addition, a similar pattern was observed for other identified flavonoid glycosides. For rutin (quercetin-3-O-rutinoside), a significantly higher content was found for the water extract of Crocus perianth (81.16 mg/g) than for the hydroethanolic extract (65.79 mg/g). It should be noted that rutin was first identified in C. sativus [7,10]. Isoqurcitrin (quercetin 3-O-glucoside) and quercetin were found in both Crocus perianth extracts. The content of isoquercetrin in the hydroethanolic extract was less (1.32 mg/g) than in the water extract (1.78 mg/g), while the content of quercetin in the extracts was approximately the same (0.2 mg/g).
Isoflavones have been identified in C. sativus for the first time. In the perianth water extract, the content of tectoridin (7-glucoside tectorigenin) was higher (1.42 mg/g) compared to the hydroethanolic extract (0.92 mg/g). Iristectorigenin B was found in the perianth in an approximately equal amount as the extracts (0.14 mg/g). The content of nigricin in the hydroethanolic extract was higher (0.10 mg/g) than in water (0.05 mg/g), the solubility and extractivity of the compound are due to the presence of the 2-OCH3 group in the molecule, which makes it difficult to dissolve in water. Thus, the influence of substituents on the solubility of compounds is once again confirmed by the example of the dependence of the content on the structure. Previously, tectoridin and iristectorigenin B were isolated or identified only in the Iris plants genus [38]. This is the first report of the identification of those compounds in C. sativus perianth. In addition, new compounds identified in Crocus perianth extracts are nigricin, mangiferin, and rutin. Only a small amount of ferulic acid, which is a biosynthetic precursor of O-phenylpropanoids in plants, were found in the perianth extracts.
By comparing the content of all compounds in the perianth extracts, it can be seen that the yield of the components in the water extract is higher since glycoside derivatives are more identified. Thus, the obtained results of the chemical composition and content of biologically active compounds of C. sativus has led to pharmacological studies of its extracts.
Among the constituents, the amount of rutin in the raw material of C. sativus perianth is high (>160 mg/g), therefore, it was selected as a Q-marker (ranking score 5). After structure–activity analysis and evaluation of the Herb MaRS criteria, five main compounds, including crocins, rutin, isoquercitrin, ferulic acid, and mangiferin, were selected based on their use for the treatment of different cancer cell lines (Figure 6). This ranking scale takes into account the clinical and pharmacological uses of the compounds and their claimed indications.

2.6. Assessment of Crocus Perianth Crude Extract Bioactivity

2.6.1. Antioxidant Activity

Nine compounds with antioxidant properties were identified in Crocus perianth extracts including mangiferin, isoorientin, ferulic acids, rutin, apigenin-7-glucoside, iristectorigenin B, and nigricin using the ABTS post-column assay. The antioxidant activity of the identified compounds was assessed by comparing their activity to the Trolox standard and expressed as TEAC values (mmol/g) and presented in Table 5. The perianth hydroethanolic extract showed higher activity (400.86 mmol/g) than Trolox (385.5 mmol/g). In the case of ABTS, stronger antioxidant activity was observed in Crocus perianth hydroethanolic extracts for mangiferin (128.13 mmol/g) and quercetin (121.11 mmol/g). Crocus raw material has shown potential bioactivity and antioxidant activity related to its radical scavenging capacity.

2.6.2. Cytotoxic Activity of Extracts

C. sativus perianth water and hydroethanolic extracts reduced the viability of melanoma (IGR39) (EC50 0.50 and 0.58 mg/mL, respectively) and triple-negative breast cancer (MDA-MB-231) (EC50 1.25 and 1.20 mg/mL, respectively) cell lines (Figure 7) via in vitro assay. Both extracts were approximately twice more active against the melanoma IGR39 than the breast cancer MDA-MB-231 cell line. The ethanolic extracts from the perianth were slightly more active than the water extract against the melanoma cells line. The Crocus perianth water extract was more active against the melanoma than the breast cancer cell line.
The activity of the water extract of Crocus perianth is higher for the melanoma IGR39, probably due to the high content of glycosides, which is also similar to some of the authors’ studies [85,109]. Some published data showed that glycosylated flavonoids exhibit more pronounced effects (anticancer, antidiabetic, antistress, antiallergic, antidegranulating, anti-inflammatory) than their aglycones [110]. However, there is a lack in vivo studies to confirm or disprove these findings.

3. Materials and Methods

3.1. Plant Material

C. sativus L. perianth was collected from the farm “Crocus.pro” (Ukraine) in November 2021. The saffron field was located in Borochyche village, Horokhiv district, Volyn region, Ukraine (50°25′46″ N; 24°49′25″ E) at an altitude of 222 m a.s.l. Stigmas were separated from flowers and dried for 2–3 h at 36 °C in a forced-air oven. Dried stigmas and flowers were stored in dark glass jars at 4 °C until analysis was performed. The specimen was deposited at the Herbarium of V.M. Karazin Kharkiv National University, Ukraine (CWN, voucher specimen No. CWN0056541). The plant cultivation and primary process were in accordance with the WHO Guidelines on Good Agricultural and Collection Practices (GACP) [25]. Following procedures established by ISO 3632 1, 2:2010–2011, moisture content and the amount of picrocrocin, crocins, and safranal for Crocus stigma were determined to identify the sample quality category.

3.2. Quality Characterization of Crocus Perianth

The powdered materials of Crocus perianth (0.1 g, 60 mesh) or crude extracts were weighed into a volumetric flask, and methanol (10 mL) was used for extraction. The flask was placed in an ultrasonic bath at room temperature (20 ± 2 °C) for 30 min. The solutions were filtered through a membrane filter (0.45 μm) into glass vials. An aliquot of 10 μL was injected twice into the HPLC system for analysis. The reference compounds were used to prepare the standard solutions at a concentration of 1.0 mg/mL in methanol and used for calibration. The samples were stored at 4 °C before use.

3.3. Extraction Procedure of Crocus Perianth for Bioassay

Crocus perianth was dried, ground, and the powder was extracted with distilled water in a water bath at 100 °C (100 g, 1 L, 60 min × 3) or 70% ethanol at room temperature (100 g, 1 L, 60 min × 3). The extracts were concentrated to dryness.

3.4. Condition of HPLC and HPLC-ABTS Analysis

Detailed conditions of the component analysis of the plant samples by HPLC are described in previous works [39]. A brief description is given as follows: compound separations were performed in an ACE C18 column (250 mm × 4.6 mm, 5.0 μm; Zorbax Eclipse Plus, Agilent, Santa Clara, CA, USA). The flow rate was 1 mL/min. The solvent system included solvent A (0.1% acetic acid in water) and solvent B (acetonitrile). An ultrasonic bath was used for degassing, then all solvents were filtered using a filter with a 0.22 μm membrane. A linear gradient program was applied: 0 min–95% A and 5% B, 7 min–95% A and 5% B, 67 min–0% A and 100% B, 69 min–95% A and 5% B, 75 min–95% A and 5% B. The column temperature was kept constant at 25 °C. The injection volume of the sample solution was adjusted to 10 μL. Chromatographic peak identification was carried out according to the analyte and reference compound retention time by comparing the UV absorption spectra of the reference compounds and analytes obtained with a diode array detector. A quantitative method for the compound determination has been reported in previous work [39]. The detector wavelength was set to 270 nm, 310 nm, and 440 nm. HPLC-ABTS analysis was performed using a Waters Alliance 2695 separation module system with some modifications. The standard Trolox antioxidant (0.3995 µmol/g) was used for the preparation of the calibration curve. Trolox equivalent antioxidant capacity (TEAC) was used to express antioxidant activity [111]. Validation of the HPLC method was performed according to the guidelines ICH Q2 (R1) “Validation of analytical procedures” [112] by the following parameters: LoD, LoQ, specificity, linearity, and precision (Tables S1 and S2). Details are described in the Supplementary Materials section.

3.5. In Vitro Assessment of Cytotoxic Activity

The potential cytotoxic effect of Crocus perianth extracts against melanoma (IGR39) and triple-negative breast cancer (MDA-MB-231) cell lines was determined by an MTT viability assay as described before [39]. Details can be found in the Supplementary Materials section.

3.6. Experimental Design

According to the guideline ICH Q8 “Pharmaceutical Development” [5], the QbD conception is based on a clear definition of the aim of the experiment, planning, and control at each stage of the entire process of obtaining plant extracts from Crocus perianth. The Design of Experiments (DoE) was used to design the design space. The experimental design included the selection of research objects, reagents, and extractants, parameters of extraction processes, chromatography conditions, and primary pharmacological screening. To reduce the number of experiments, a final screening design was used to investigate the effect of these parameters on the manufacturing process. Many process parameters can also be studied in very few experiments.

3.7. Risk Analysis

Risk assessment consists of identifying and describing potential hazards, assessing exposure, and characterizing the risk. An Ishikawa diagram analysis was performed by the guideline ICH Q9 “Quality Risk Management” [27], to identify factors that could affect the extraction of compounds from Crocus raw material. Failure Mode and Effect Analysis (FMEA) is proposed as an indispensable tool for classifying risks based on severity (S), probability of occurrence (O), and probability of detection (D) of raw materials at risk. In the FMEA, risk in the final product is expressed in terms of RPN (risk priority number), which is defined as follows: RPN = S × O × D. If RPN > 130, corrective action should be taken. FMEA considers any element that is part of the entire system [29]. The analysis of all possible reasons why each component or subsystem may not perform its intended function is carried out based on both the best opinion of experts and historical information on similar elements. The FMEA analysis is built based on the evaluation of hazardous treatments and the calculation of RPN, as shown in Table 1 and Table S3, and corrective actions are proposed for each identified hazard.

3.8. Herb MaRS Approach

To determine the most appropriate chemical markers for the quality control of C. sativus perianth and extracts, the Herbal Chemical Marker Ranking System (Herb MaRS) was used (developed by the National Institute of Complementary Medicine (NICM) at the University of Western Sydney, 2014 [17]). The Herb MaRS method takes into account various factors associated with herbal ingredients, such as the availability of biological activity studies and purely chemical reference standards; the relationship of the traditional or current use of the herb to its therapeutic use or pharmacological effects; the concentration of the chemical marker in the herbal product and the toxicity or maximum recommended dose. The Herb MaRS criteria contain a priority list of chemical markers, rationally ranked on a scale from 0 to 5, with 5 indicating the most appropriate chemical marker and rank 0 denoting least suitable. In addition, the “X” category indicates that there were no studies on the biological activity of the compound available at the time of selection. These compounds cannot be completely ruled out as potential chemical markers due to unknown activity.

3.9. Data Analysis

All data processing was carried out using the LabSolutions Analysis Data System (Shimadzu Corporation). Phenolic compound content and Trolox equivalent antioxidant activity of phenolic compounds was expressed as mean ± standard error (SE), n = 3. Statistical analysis was performed using one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison test with the software package Prism v.5.04 (GraphPad Software Inc., La Jolla, CA, USA). The value of p < 0.05 was taken as the significance level. The data were processed using the Microsoft Office Excel 2010 (Microsoft, JAV) software package.

4. Conclusions

It is now especially important to develop and apply standards that guarantee the quality, safety, and efficacy of herbal drugs. The design of experimentation and the selection of adequate chemical markers for quality control purposes require a good knowledge of the chemical composition of medicinal plants and their associated biological properties. We applied the Herb MaRS criteria to prioritize the selection of chemical markers for quality control for C. sativus perianth, whilst also taking into account cancer bioactivity and its concentration in the extract. Crocins, flavonoid glycosides (rutin, isoquercitrin), xanthone mangiferin, and ferulic acid have been proposed as potential chemical markers for the quality control of Crocus perianth and its crude extracts. C. sativus perianth has shown potent antioxidant and anticancer bioactivities that might be related to the radical scavenging capacity of its major components. Thus, further pharmacological tests of Crocus raw materials as anticancer raw materials have scientific justification and prospects. In addition, the presence of high-quality stigmas (I category according to ISO 3632) will determine the availability of the raw material base of Crocus perianth. Implementing the Quality by Design approach will assist in managing the risks for quality control and processes in herbal drugs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/scipharm90010019/s1, Tables S1–S3; Figures S1–S5. Materials and Methods.

Author Contributions

Methodology, Data curation, Writing—original draft, O.M.; Formal analysis, Investigation, I.B.; Conceptualization, Resources, Visualization, L.I.; Methodology, Investigation, V.P.; Conceptualization, Writing—review and editing, Supervision, V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

This research contains only Supplementary Material information.

Acknowledgments

The authors would like to thank the Lithuanian University of Health Sciences for providing instrumentation support and also express their gratitude to the farm “Crocus.pro” of the Volyn region for consultation and assistance in the fieldwork.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviation

ABTS2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt
CPPcritical process parameters
CQAscritical quality attributes
DoEDesign of experiment
EMAEuropean Medicines Agency
FDAUnited States Food and Drug Administration
FMEAFailure Modes and Effects Analysis
GACPGood Agricultural and Collection Practices
GAPGood Agricultural Practice
GHPGood Handling Practice
GMPGood Manufacturing Practice
HACCPHazard Analysis and Critical Control Points System
Herb MaRSHerbal Chemical Marker Ranking System
HPLC-DADhigh-performance liquid chromatography coupled with diode array detector
ICHInternational Council for the Harmonization of Specifications for Pharmaceutical Products for Human Use
QbDQuality by Design
Q-markerquality marker
QTPPquality target product profile
WHOWorld Health Organization

References

  1. Jimenez-Garcia, S.; Vazquez-Cruz, M.; Guevara-González, R.G.; Torres-Pacheco, I.; Cruz-Hernandez, A.; Feregrino-Perez, A. Current approaches for enhanced expression of secondary metabolites as bioactive compounds in plants for agronomic and human health purposes. Pol. J. Food Nutr. Sci. 2013, 63, 67–78. [Google Scholar] [CrossRef] [Green Version]
  2. Gomes-Araújo, R.; Martínez-Vázquez, D.G.; Charles-Rodríguez, A.V.; Rangel-Ortega, S.; Robledo-Olivo, A. Bioactive Compounds from agricultural residues, their obtaining techniques, and the antimicrobial effect as postharvest additives. Int. J. Food Sci. 2021, 2021, 9936722. [Google Scholar] [CrossRef] [PubMed]
  3. Goyal, M.R.; Rasul Suleria, H.A. Human health benefits of plant bioactive compounds. In Potentials and Prospects, 1st ed.; Apple Academic Press: Palm Bay, FL, USA, 2021; p. 396. [Google Scholar]
  4. European Medicines Agency. Guideline on Quality of Herbal Medicinal Products/Traditional Herbal Medicinal Products; EMA/HMPC/201116/2005 Rev. 2; European Medicines Agency: Amsterdam, The Netherlands, 2011. [Google Scholar]
  5. European Medicines Agency (EMA). ICH Guideline Q8 (R2) on Pharmaceutical Development. Step 5; EMA/CHMP/ICH/167068/2004; European Medicines Agency: Amsterdam, The Netherlands, 2017. [Google Scholar]
  6. EMA. EMA-FDA Pilot Program for Parallel Assessment of Quality-by-Design Applications: Lessons Learnt and Q&A Resulting from the First Parallel Assessment; EMA/430501/2013; Human Medicines Development and Evaluation; European Medicines Agency and U.S. Food and Drug Administration: Silver Spring, MD, USA, 2013. [Google Scholar]
  7. Moratalla-López, N.; Bagur, M.J.; Lorenzo, C.; Salinas, M.E.M.R.; Alonso, G.L. Bioactivity and bioavailability of the major metabolites of Crocus sativus L. flower. Molecules 2019, 24, 2827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Asgarpanah, J.; Darabi-Mahboub, E.; Mahboubi, A.; Mehrab, R.; Hakemivala, M. In-vitro evaluation of Crocus sativus L. petals and stamens as natural antibacterial agents against food-borne bacterial strains. Iran. J. Pharm. Sci. 2013, 9, 69–82. [Google Scholar]
  9. Agarwal, N.; Kolba, N.; Jung, Y.; Cheng, J.; Tako, E. Saffron (Crocus sativus L.) flower water extract disrupts the cecal microbiome, brush border membrane functionality, and morphology in vivo (gallus gallus). Nutrients 2022, 14, 220. [Google Scholar] [CrossRef]
  10. Montoro, P.; Maldini, M.; Luciani, L.; Tuberoso, C.I.; Congiu, F.; Pizza, C. Radical scavenging activity and LC-MS metabolic profiling of petals, stamens, and flowers of Crocus sativus L. J. Food Sci. 2012, 77, 893–900. [Google Scholar] [CrossRef]
  11. Stelluti, S.; Caser, M.; Demasi, S.; Scariot, V. Sustainable processing of floral bio-residues of saffron (Crocus sativus L.) for valuable biorefinery products. Plants 2021, 10, 523. [Google Scholar] [CrossRef]
  12. Sánchez-Vioque, R.; Santana-Méridas, O.; Polissiou, M.; Vioque, J.; Astraka, K.; Alaizd, M.; Herraiz-Peñalvera, D.; Tarantilis, P.A.; Girón-Calle, J. Polyphenol composition and in vitro antiproliferative effect of corm, tepal and leaf from Crocus sativus L. on human colon adenocarcinoma cells (Caco-2). J. Funct. Foods 2016, 24, 18–25. [Google Scholar] [CrossRef] [Green Version]
  13. De Monte, C.; Carradori, S.; Chimenti, P.; Secci, D.; Mannina, L.; Alcaro, F.; Petzer, A.; N’Da, C.I.; Gidaro, M.C.; Costa, G.; et al. New insights into the biological properties of Crocus sativus L.: Chemical modifications, human monoamine oxidases inhibition and molecular modeling studies. Eur. J. Med. Chem. 2014, 82, 164–171. [Google Scholar] [CrossRef]
  14. Mykhailenko, O.; Desenko, V.; Ivanauskas, L.; Georgiyants, V. Standard operating procedure of Ukrainian saffron cultivation according with Good Agricultural and Collection Practices to assure quality and traceability. Ind. Crops Prod. 2020, 151, 112376–112387. [Google Scholar] [CrossRef]
  15. Shanaida, M.; Golembiovska, O.; Hudz, N.; Wieczorek, P.P. Phenolic compounds of herbal infusions obtained from some species of the Lamiaceae family. Curr. Issues Pharm. Med. Sci. 2018, 31, 194–199. [Google Scholar] [CrossRef] [Green Version]
  16. European Medicines Agency. Overview of Comments Received on Draft ‘Reflection Paper on Markers Used for Quantitative and Qualitative Analysis of Herbal Medicinal Products and Traditional Herbal Medicinal Products’; EMEA/HMPC/253629/2007; European Medicines Agency: Amsterdam, The Netherlands, 2008. [Google Scholar]
  17. Bensoussan, A.; Lee, S.; Murray, C.; Bourchier, S.; van der Kooy, F.; Pearson, J.; Liu, J.; Chang, D.; Khoo, C. Choosing chemical markers for quality assurance of complex herbal medicines: Development and application of the Herb MaRS criteria. Clin. Pharmacol. Ther. 2015, 97, 628–640. [Google Scholar] [CrossRef] [PubMed]
  18. Yu, L.X. Pharmaceutical quality by design: Product and process development, understanding, and control. Pharm. Res. 2008, 25, 781–791. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, L.; Mao, S. Application of quality by design in the current drug development. Asian J. Pharm. Sci. 2017, 12, 1–8. [Google Scholar] [CrossRef] [Green Version]
  20. European Medicines Agency. Guideline on the Evaluation of Anticancer Medicinal Products in Man; EMA/CHMP/205/95 Rev.5; European Medicines Agency: Amsterdam, The Netherlands, 2017. [Google Scholar]
  21. European Medicines Agency. Questions and Answers on Quality of Herbal Medicinal Products/Traditional Herbal Medicinal Products—Revision 6; EMA/HMPC/41500/2010 Rev.6; European Medicines Agency: Amsterdam, The Netherlands, 2018. [Google Scholar]
  22. European Medicines Agency. Guideline on Specifications Test Procedures and Acceptance Criteria for Herbal Substances Herbal Preparations and Herbal Medicinal Products Traditional Herbal Medicinal Products—Revision 2; EMA/HMPC/162241/2005 Rev. 2; European Medicines Agency: Amsterdam, The Netherlands, 2011. [Google Scholar]
  23. Schmidt, A.; Strube, J. Distinct and quantitative validation method for predictive process modeling with examples of liquid-liquid extraction processes of complex feed mixtures. Processes 2019, 7, 298. [Google Scholar] [CrossRef] [Green Version]
  24. Borges, C.V.; Minatel, I.O.; Gomez-Gomez, H.A.; Lima, G.P.P. Medicinal Plants: Influence of Environmental Factors on the Content of Secondary Metabolites. In Medicinal Plants and Environmental Challenges; Ghorbanpour, M., Varma, A., Eds.; Springer International Publishing: Cham, Switzerland, 2017; Volume 15, pp. 259–277. [Google Scholar]
  25. EMEA. Guideline on Good Agricultural and Collection Practice (GACP) for Starting Materials of Herbal Origin, London, European Medicines for Human Use; EMEA/HMPC/246816/2005; EMEA: London, UK, 2006. [Google Scholar]
  26. Uhlenbrock, L.; Sixt, M.; Strube, J. Quality-by-Design (QbD) process evaluation for phytopharmaceuticals on the example of 10-deacetylbaccatin III from yew. Resour. Effic. Technol. 2017, 3, 137–143. [Google Scholar] [CrossRef]
  27. European Medicines Agency. ICH Guideline Q9 on Quality Risk Management; EMA/CHMP/ICH/24235/2006; European Medicines Agency: Amsterdam, The Netherlands, 2015. [Google Scholar]
  28. Savchenko, L.; Pidpruzhnykov, Y.; Ivanauskas, L.; Lukošius, A.; Georgiyants, V. Risk assessment for compounding ointments quality by Ishikawa diagram construction. Farmacia 2021, 69, 688–696. [Google Scholar] [CrossRef]
  29. Fahmy, R.; Kona, R.; Dandu, R.; Xie, W.; Claycamp, G.; Hoag, S.W. Quality by design I: Application of failure mode effect analysis (FMEA) and Plackett-Burman design of experiments in the identification of “main factors” in the formulation and process design space for roller-compacted ciprofloxacin hydrochloride immediate-release tablets. AAPS Pharm. Sci. Technol. 2012, 13, 1243–1254. [Google Scholar]
  30. Desai, A.G.; Qazi, G.N.; Ganju, R.K.; El-Tamer, M.; Singh, J.; Saxena, A.K.; Bedi, Y.S.; Taneja, S.C.; Bhat, H.K. Medicinal plants and cancer chemoprevention. Curr. Drug Metab. 2008, 9, 581–591. [Google Scholar] [CrossRef] [Green Version]
  31. Sagbo, I.J.; Otang-Mbeng, W. Plants used for the traditional management of cancer in the eastern cape province of South Africa: A review of ethnobotanical surveys, ethnopharmacological studies and active phytochemicals. Molecules 2021, 26, 4639. [Google Scholar] [CrossRef]
  32. Bolhassani, A. Chapter 10—Bioactive components of saffron and their pharmacological properties. In Studies in Natural Products Chemistry; Elsevier: Amsterdam, The Netherlands, 2018; Volume 58, pp. 289–311. [Google Scholar]
  33. Mohtashami, L.; Amiri, M.S.; Ramezani, M.; Emami, S.A.; Simal-Gandara, J. The genus Crocus L.: A review of ethnobotanical uses, phytochemistry and pharmacology. Ind. Crops Prod. 2021, 171, 113923–113948. [Google Scholar] [CrossRef]
  34. Montoro, P.; Tuberoso, C.I.G.; Maldini, M.; Cabras, P.; Pizza, C. Qualitative profile and quantitative determination of flavonoids from Crocus sativus L. petals by LC-MS/M. Nat. Prod. Commun. 2008, 3, 2013–2016. [Google Scholar] [CrossRef] [Green Version]
  35. Li, C.Y.; Lee, E.J.; Wu, T.S. Antityrosinase principles and constituents of the petals of Crocus sativus. J. Nat. Prod. 2004, 67, 437–440. [Google Scholar] [CrossRef] [PubMed]
  36. Trapero, A.; Ahrazem, O.; Rubio-Moraga, A.; Jimeno, M.L.; Gómez, M.D.; Gómez-Gómez, L. Characterization of a glucosyltransferase enzyme involved in the formation of kaempferol and quercetin sophorosides in Crocus sativus. Plant Physiol. 2012, 159, 1335–1354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Vignolini, P.; Heimler, D.; Pinelli, P.; Ieri, F.; Sciullo, A.; Romani, A. Characterization of by-products of saffron (Crocus sativus L.) production. Nat. Prod. Commun. 2008, 3, 1959–1962. [Google Scholar] [CrossRef] [Green Version]
  38. Singab, A.N.B.; Ayoub, I.M.; El-Shazly, M.; Korinek, M.; Wu, T.-Y.; Cheng, Y.-B.; Wu, Y.-C. Shedding the light on Iridaceae: Ethnobotany, phytochemistry and biological activity. Ind. Crops Prod. 2016, 92, 308–335. [Google Scholar] [CrossRef]
  39. Mykhailenko, O.; Petrikaitė, V.; Korinek, M.; El-Shazly, M.; Chen, B.-H.; Yen, C.-H.; Hsieh, C.-F.; Bezruk, I.; Dabrišiūtė, A.; Ivanauskas, L.; et al. Bio-guided bioactive profiling and HPLC-DAD fingerprinting of Ukrainian saffron (Crocus sativus stigma): Moving from Correlation toward Causation. BMC Complement. Med. Ther. 2021, 21, 203. [Google Scholar] [CrossRef]
  40. Mykhailenko, O.; Bezruk, I.; Ivanauskas, L.; Lesyk, R.; Georgiyants, V. Characterization of phytochemical components of Crocus sativus leaves using HPLC-MS/MS and GC-MS: A new potential by-product. Sci. Pharm. 2021, 89, 28. [Google Scholar] [CrossRef]
  41. Khazdair, M.R.; Boskabady, M.H.; Hosseini, M.; Rezaee, R.M.; Tsatsakis, A. The effects of Crocus sativus (saffron) and its constituents on nervous system: A review. Avicenna J. Phytomed. 2015, 5, 376–391. [Google Scholar]
  42. Nassiri-Asl, M.; Hosseinzadeh, H. Neuropharmacology effects of saffron (Crocus sativus) and its active constituents. In Bioactive Nutraceuticals and Dietary Supplements in Neurological and Brain Disease; Academic Press: Cambridge, MA, USA, 2015; Volume 3, pp. 29–39. [Google Scholar]
  43. Mir, M.A.; Ganai, S.A.; Mansoor, S.; Jan, S.; Mani, P.; Masoodi, K.Z.; Amin, H.; Rehman, M.U.; Ahmad, P. Isolation, purification and characterization of naturally derived crocetin beta-D-glucosyl ester from Crocus sativus L. against breast cancer and its binding chemistry with ER-alpha/HDAC2. Saudi J. Biol. Sci. 2020, 27, 975–984. [Google Scholar] [CrossRef]
  44. Colapietro, A.; Mancini, A.; D’Alessandro, A.M.; Festuccia, C. Crocetin and crocin from saffron in cancer chemotherapy and chemoprevention. Anti Cancer Agents Med. Chem. 2019, 19, 38–47. [Google Scholar] [CrossRef] [PubMed]
  45. Yu, L.; Li, J.; Xiao, M. Picrocrocin exhibits growth inhibitory effects against SKMEL- 2 human malignant melanoma cells by targeting JAK/ STAT5 signaling pathway, cell cycle arrest and mitochondrial mediated apoptosis. J. BUON 2018, 23, 1163–1168. [Google Scholar] [PubMed]
  46. Hoshyar, R.; Mollaei, H. A comprehensive review on anticancer mechanisms of the main carotenoid of saffron, crocin. J. Pharm. Pharmacol. 2017, 69, 1419–1427. [Google Scholar] [CrossRef] [Green Version]
  47. Lambrianidou, A.; Koutsougianni, F.; Papapostolou, I.; Dimas, K. Recent advances on the anticancer properties of saffron (Crocus sativus L.) and its major constituents. Molecules 2020, 26, 86. [Google Scholar] [CrossRef]
  48. Chryssanthi, D.G.; Lamari, F.N.; Iatrou, G.; Pylara, A.; Karamanos, N.K.; Cordopatis, P. Inhibition of breast cancer cell proliferation by style constituents of different Crocus species. Anticancer Res. 2007, 27, 357–362. [Google Scholar]
  49. Chen, S.S.; Gu, Y.; Lu, F.; Qian, D.P.; Dong, T.T.; Ding, Z.H.; Zhao, S.; Yu, Z.H. Antiangiogenic effect of crocin on breast cancer cell MDA-MB-231. J. Thorac. Dis. 2019, 11, 4464–4473. [Google Scholar] [CrossRef] [PubMed]
  50. Gismondi, A.; Serio, M.; Canuti, L.; Canini, A. Biochemical, antioxidant and antineoplastic properties of Italian saffron (Crocus sativus L.). Am. J. Plant Sci. 2012, 3, 1573–1580. [Google Scholar] [CrossRef] [Green Version]
  51. Chryssanthi, D.G.; Dedes, P.G.; Karamanos, N.K.; Cordopatis, P.; Lamari, F.N. Crocetin inhibits invasiveness of MDA-MB-231 breast cancer cells via downregulation of matrix metalloproteinases. Planta Med. 2011, 77, 146–151. [Google Scholar] [CrossRef]
  52. Escribano, J.; Alonso, G.L.; Coca-Prados, M.; Fernandez, J.A. Crocin, safranal and picrocrocin from saffron (Crocus sativus L.) inhibit the growth of human cancer cells in vitro. Cancer Lett. 1996, 100, 23–30. [Google Scholar] [CrossRef]
  53. Razavi, B.M.; Amanloo, M.A.; Imenshahidi, M.; Hosseinzadeh, H. The relaxant activity of safranal in isolated rat aortas is mediated predominantly via an endothelium-independent mechanism: Vasodilatory mechanism of safranal. J. Pharmacopunct. 2016, 19, 329–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Nanda, S.; Madan, K. The role of safranal and saffron stigma extracts in oxidative stress, diseases and photoaging: A systematic review. Heliyon 2021, 7, e06117. [Google Scholar] [CrossRef] [PubMed]
  55. Eroğlu, C.; Seçme, M.; Bağcı, G.; Dodurga, Y. Assessment of the anticancer mechanism of ferulic acid via cell cycle and apoptotic pathways in human prostate cancer cell lines. Tumour Biol. 2015, 36, 9437–9446. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, X.; Lin, D.; Jiang, R.; Li, H.; Wan, J.; Li, H. Ferulic acid exerts antitumor activity and inhibits metastasis in breast cancer cells by regulating epithelial to mesenchymal transition. Oncol. Rep. 2016, 36, 271–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kim, J.K.; Park, S.U. A recent overview on the biological and pharmacological activities of ferulic acid. Excli J. 2019, 18, 132–138. [Google Scholar]
  58. Son, B.K.; Choi, Y.S.; Sohn, Y.W. Anticancer effect of ferulic acid on cultured human skin melanoma cells. J. Exp. Biomed. Sci. 2006, 12, 457–461. [Google Scholar]
  59. Rajendra, P.N.; Karthikeyan, A.; Karthikeyan, S.; Reddy, B.V. Inhibitory effect of caffeic acid on cancer cell proliferation by oxidative mechanism in human HT-1080 fibrosarcoma cell line. Mol. Cell Biochem. 2011, 349, 11–19. [Google Scholar] [CrossRef]
  60. Khan, F.A.; Maalik, A.; Murtaza, G. Inhibitory mechanism against oxidative stress of caffeic acid. J. Food Drug Anal. 2016, 24, 695–702. [Google Scholar] [CrossRef]
  61. Gold-Smith, F.; Fernandez, A.; Bishop, K. Mangiferin and Cancer: Mechanisms of Action. Nutrients 2016, 8, 396. [Google Scholar] [CrossRef] [Green Version]
  62. Mei, S.; Ma, H.; Chen, X. Anticancer and anti-inflammatory properties of mangiferin: A review of its molecular mechanisms. Food Chem. Toxicol. 2021, 149, 111997. [Google Scholar] [CrossRef]
  63. Morozkina, S.N.; Nhung Vu, T.H.; Generalova, Y.E.; Snetkov, P.P.; Uspenskaya, M.V. Mangiferin as new potential anti-cancer agent and mangiferin-integrated polymer systems—A novel research direction. Biomolecules 2021, 11, 79. [Google Scholar] [CrossRef]
  64. Li, H.; Huang, J.; Yang, B.; Xiang, T.; Yin, X.; Peng, W.; Cheng, W.; Wan, J.; Luo, F.; Li, H.; et al. Mangiferin exerts antitumor activity in breast cancer cells by regulating matrix metalloproteinases, epithelial to mesenchymal transition, and β-catenin signaling pathway. Toxicol. Appl. Pharmacol. 2013, 272, 180–190. [Google Scholar] [CrossRef] [PubMed]
  65. Guha, S.; Ghosal, S.; Chattopadhyay, U. Antitumor, immunomodulatory and anti-HIV effect of mangiferin, a naturally occurring glucosylxanthone. Chemotherapy 1996, 42, 443–451. [Google Scholar] [CrossRef] [PubMed]
  66. Rajendran, P.; Ekambaram, G.; Sakthisekaran, D. Protective role of mangiferin against benzo(a)pyrene induced lung carcinogenesis in experimental animals. Biol. Pharm. Bull. 2008, 31, 1053–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Rajendran, P.; Ganapathy, E.; Sakthisekaran, D. Cytoprotective effect of mangiferin on benzo(a)pyrene-induced lung carcinogenesis in swiss albino mice. Basic Clin. Pharmacol. Toxicol. 2008, 103, 137–142. [Google Scholar] [CrossRef]
  68. Gundogdu, G.; Dodurga, Y.; Elmas, L.; Tasci, S.Y.; Karaoglan, E.S. Investigation of the anticancer mechanism of isoorientin isolated from Eremurus Spectabilis leaves via cell cycle pathways in HT-29 human colorectal adenocarcinoma cells. Eurasian J. Med. 2018, 50, 168–172. [Google Scholar] [CrossRef]
  69. Ye, T.; Su, J.; Huang, C.; Yu, D.; Dai, S.; Huang, X.; Chen, B.; Zhou, M. Isoorientin induces apoptosis, decreases invasiveness, and downregulates VEGF secretion by activating AMPK signaling in pancreatic cancer cells. OncoTargets Ther. 2016, 9, 7481–7492. [Google Scholar] [CrossRef] [Green Version]
  70. Anilkumar, K.; Reddy, G.V.; Azad, R.; Yarla, N.S.; Dharmapuri, G.; Srivastava, A.; Kamal, M.A.; Pallu, R. Evaluation of anti-inflammatory properties of isoorientin isolated from tubers of Pueraria tuberosa. Oxid. Med. Cell. Longev. 2017, 2017, 5498054. [Google Scholar] [CrossRef] [Green Version]
  71. Kim, T.H.; Ku, S.K.; Lee, I.C.; Bae, J.S. Anti-inflammatory effects of kaempferol-3-O-sophoroside in human endothelial cells. Inflamm. Res. 2012, 61, 217–224. [Google Scholar] [CrossRef]
  72. Iriti, M.; Kubina, R.; Cochis, A.; Sorrentino, R.; Varoni, E.M.; Kabała-Dzik, A.; Azzimonti, B.; Dziedzic, A.; Rimondini, L.; Wojtyczka, R.D. Rutin, a quercetin glycoside, restores chemosensitivity in human breast cancer cells. Phytother. Res. 2017, 31, 1529–1538. [Google Scholar] [CrossRef]
  73. Drewa, G.; Schachtschabel, D.O.; Pałgan, K.; Grzanka, A.; Sujkowska, R. The influence of rutin on the weight, metastasis and melanin content of B16 melanotic melanoma in C57BL/6 mice. Neoplasma 1998, 45, 266–271. [Google Scholar]
  74. Caparica, R.; Júlio, A.; Araújo, M.E.M.; Baby, A.R.; Fonte, P.; Costa, J.G.; Santos de Almeida, T. Anticancer activity of rutin and its combination with ionic liquids on renal cells. Biomolecules 2020, 10, 233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Satari, A.; Ghasemi, S.; Habtemariam, S.; Asgharian, S.; Lorigooini, Z. Rutin: A flavonoid as an effective sensitizer for anticancer therapy; insights into multifaceted mechanisms and applicability for combination therapy. Evid. Based Complement. Altern. Med. 2021, 2021, 9913179. [Google Scholar] [CrossRef] [PubMed]
  76. Pinzaru, I.; Chioibas, R.; Marcovici, I.; Coricovac, D.; Susan, R.; Predut, D.; Georgescu, D. Rutin exerts cytotoxic and senescence-inducing properties in human melanoma cells. Toxics 2021, 9, 226. [Google Scholar] [CrossRef] [PubMed]
  77. Elsayed, H.E.; Ebrahim, H.Y.; Mohyeldin, M.M.; Siddique, A.B.; Kamal, A.M.; Haggag, E.G.; El Sayed, K.A. Rutin as a novel c-met inhibitory lead for the control of triple negative breast malignancies. Nutr. Cancer 2017, 69, 1256–1271. [Google Scholar] [CrossRef]
  78. Chen, F.; Chen, X.; Yang, D.; Che, X.; Wang, J.; Li, X.; Zhang, Z.; Wang, Q.; Zheng, W.; Wang, L.; et al. Isoquercitrin inhibits bladder cancer progression in vivo and in vitro by regulating the PI3K/Akt and PKC signaling pathways. Oncol. Rep. 2016, 36, 165–172. [Google Scholar] [CrossRef] [Green Version]
  79. Orfali Gd Duarte, A.C.; Bonadio, V.; Martinez, N.P.; de Araújo, M.E.; Priviero, F.B.; Carvalho, P.O.; Priolli, D.G. Review of anticancer mechanisms of isoquercitin. World J. Clin. Oncol. 2016, 7, 189–199. [Google Scholar] [CrossRef]
  80. Chen, Q.; Li, P.; Li, P.; Xu, Y.; Li, Y.; Tang, B. Isoquercitrin inhibits the progression of pancreatic cancer in vivo and in vitro by regulating opioid receptors and the mitogen-activated protein kinase signalling pathway. Oncol. Rep. 2014, 33, 840–848. [Google Scholar] [CrossRef] [Green Version]
  81. Bauer, D.; Mazzio, E.; Soliman, K.F.A. Whole transcriptomic analysis of apigenin on TNFα immuno-activated MDA-MB-231 breast cancer cells. Cancer Genom. Proteom. 2019, 16, 421–431. [Google Scholar] [CrossRef] [Green Version]
  82. Kabała-Dzik, A.; Rzepecka-Stojko, A.; Kubina, R.; Iriti, M.; Wojtyczka, R.D.; Buszman, E.; Stojko, J. Flavonoids, bioactive components of propolis, exhibit cytotoxic activity and induce cell cycle arrest and apoptosis in human breast cancer cells MDA-MB-231 and MCF-7-a comparative study. Cell Mol. Biol. 2018, 64, 1–10. [Google Scholar] [CrossRef] [Green Version]
  83. Chen, D.; Landis-Piwowar, K.R.; Chen, M.S.; Dou, Q.P. Inhibition of proteasome activity by the dietary flavonoid apigenin is associated with growth inhibition in cultured breast cancer cells and xenografts. Breast Cancer Res. 2007, 9, R80–R88. [Google Scholar] [CrossRef] [Green Version]
  84. Ghițu, A.; Schwiebs, A.; Radeke, H.H.; Avram, S.; Zupko, I.; Bor, A.; Pavel, I.Z.; Dehelean, C.A.; Oprean, F.; Bojin, C.; et al. A comprehensive assessment of apigenin as an antiproliferative, proapoptotic, antiangiogenic and immunomodulatory phytocompound. Nutrients 2019, 11, 858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Smiljkovic, M.; Stanisavljevic, D.; Stojkovic, D.; Petrovic, I.; Marjanovic Vicentic, J.; Popovic, J.; Golic Grdadolnik, S.; Markovic, D.; Sankovic-Babice, S.; Glamoclija, J.; et al. Apigenin-7-O-glucoside versus apigenin: Insight into the modes of anticandidal and cytotoxic actions. Excli J. 2017, 16, 795–807. [Google Scholar] [PubMed]
  86. Hanske, L.; Loh, G.; Sczesny, S.; Blaut, M.; Braune, A. The bioavailability of apigenin-7-glucoside is influenced by human intestinal microbiota in rats. J. Nutr. 2009, 139, 1095–1102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Bouzaiene, N.; Chaabane, F.; Sassi, A.; Chekir-Ghedira, L.; Ghedira, K. Effect of apigenin-7-glucoside, genkwanin and naringenin on tyrosinase activity and melanin synthesis in B16F10 melanoma cells. Life Sci. 2016, 144, 80–85. [Google Scholar] [CrossRef]
  88. Lan, H.; Hong, W.; Fan, P.; Qian, D.; Zhu, J.; Bai, B. Quercetin inhibits cell migration and invasion in human osteosarcoma cells. Cell Physiol. Biochem. 2017, 43, 553–567. [Google Scholar] [CrossRef] [Green Version]
  89. Hashemzaei, M.; Far, A.D.; Yari, A.; Heravi, R.E.; Tabrizian, K.; Taghdisi, S.M.; Sadegh, S.E.; Tsarouhas, K.; Kouretas, D.; Tzanakakis, G.; et al. Anticancer and apoptosis-inducing effects of quercetin in vitro and in vivo. Oncol. Rep. 2017, 38, 819–828. [Google Scholar] [CrossRef] [Green Version]
  90. Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of biological importance of quercetin: A bioactive flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar]
  91. Liu, M.; Yang, S.; Jin, L.; Hu, D.; Wu, Z.; Yang, S. Chemical constituents of the ethyl acetate extract of Belamcanda chinensis (L.) DC roots and their antitumor activities. Molecules 2012, 17, 6156–6169. [Google Scholar] [CrossRef]
  92. Xu, D.; Hu, M.J.; Wang, Y.Q.; Cui, Y.L. Antioxidant activities of quercetin and its complexes for medicinal application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef] [Green Version]
  93. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Aslam Gondal, T.; Saeed, F.; Imran, A.; Shahbaz, M.; Tsouh Fokou, P.V.; Umair Arshad, M.; Khan, H.; et al. Kaempferol: A key emphasis to its anticancer potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [Green Version]
  94. Gao, Y.; Yin, J.; Rankin, G.O.; Chen, Y.C. Kaempferol induces G2/M cell cycle arrest via checkpoint kinase 2 and promotes apoptosis via death receptors in human ovarian carcinoma A2780/CP70 Cells. Molecules 2018, 23, 1095. [Google Scholar] [CrossRef] [Green Version]
  95. Huang, H.C.; Lin, C.L.; Lin, J.K. 1,2,3,4,6-penta-O-galloyl-β-Dglucose, quercetin, curcumin and lycopene induce cell-cycle arrest in MDA-MB-231 and BT474 cells through downregulation of Skp2 protein. J. Agric. Food Chem. 2011, 59, 6765–6775. [Google Scholar] [CrossRef]
  96. Yanqiu, H.; Linjuan, C.; Jin, W.; Hao, H.; Shi, Y.; Xue, G.; Ren, H. The effects of quercetin and kaempferol on multidrug resistance and the expression of related genes in human erythroleukemic K562/A cells. Afr. J. Biotechnol. 2011, 10, 13399–13406. [Google Scholar] [CrossRef]
  97. Yang, M.; Li, W.Y.; Xie, J.; Wang, Z.L.; Wen, Y.L.; Zhao, C.C.; Tao, L.; Li, L.F.; Tian, Y.; Sheng, J. Astragalin inhibits the proliferation and migration of human colon cancer HCT116 cells by regulating the NF-κB sSignaling pathway. Front. Pharmacol. 2021, 12, 639256. [Google Scholar]
  98. Wang, Z.; Lv, J.; Li, X.; Lin, Q. The flavonoid Astragalin shows anti-tumor activity and inhibits PI3K/AKT signaling in gastric cancer. Chem. Biol. Drug Des. 2021, 98, 779–786. [Google Scholar] [CrossRef] [PubMed]
  99. Hu, J.; Zhang, Y.; Jiang, X.; Zhang, H.; Gao, Z.; Li, Y.; Fu, R.; Li, L.; Li, J.; Cui, H.; et al. ROS-mediated activation and mitochondrial translocation of CaMKII contributes to Drp1-dependent mitochondrial fission and apoptosis in triple-negative breast cancer cells by isorhamnetin and chloroquine. J. Exp. Clin. Cancer Res. 2019, 38, 225. [Google Scholar] [CrossRef]
  100. Cai, F.; Zhang, Y.; Li, J.; Huang, S.; Gao, R. Isorhamnetin inhibited the proliferation and metastasis of androgen-independent prostate cancer cells by targeting the mitochondrion-dependent intrinsic apoptotic and PI3K/Akt/mTOR pathway. Biosci. Rep. 2020, 40, BSR20192826. [Google Scholar] [CrossRef] [Green Version]
  101. Rahman, A.U.; Nasim, S.; Baig, I.; Jalil, S.; Orhan, I.; Sener, B.; Choudhary, M.I. Anti-inflammatory isoflavonoids from the rhizomes of Iris germanica. J. Ethnopharmacol. 2003, 86, 177–180. [Google Scholar] [CrossRef]
  102. Jun, H.J.; Hoang, M.H.; Lee, J.W.; Yaoyao, J.; Lee, J.H.; Lee, D.H.; Lee, H.J.; Seo, W.D.; Hwang, B.Y.; Lee, S.J. Iristectorigenin B isolated from Belamcanda chinensis is a liver X receptor modulator that increases ABCA1 and ABCG1 expression in macrophage RAW 264.7 cells. Biotechnol. Lett. 2012, 34, 2213–2221. [Google Scholar] [CrossRef]
  103. Lim, H.; Park, B.K.; Shin, S.Y.; Kwon, Y.S.; Kim, H.P. Methyl caffeate and some plant constituents inhibit age-related inflammation: Effects on senescence-associated secretory phenotype (SASP) formation. Arch. Pharm. Res. 2017, 40, 524–535. [Google Scholar] [CrossRef]
  104. Han, T.; Cheng, G.; Liu, Y.; Yang, H.; Hu, Y.T.; Huang, W. In vitro evaluation of tectoridin, tectorigenin and tectorigenin sodium sulfonate on antioxidant properties. Food Chem. Toxicol. 2012, 50, 409–414. [Google Scholar] [CrossRef] [PubMed]
  105. Kang, K.; Lee, S.B.; Jung, S.H.; Cha, K.H.; Park, W.D.; Sohn, Y.C.; Nho, C.W. Tectoridin, a poor ligand of estrogen receptor alpha, exerts its estrogenic effects via an ERK-dependent pathway. Mol. Cells 2009, 27, 351–357. [Google Scholar] [CrossRef] [PubMed]
  106. Xiong, Y.; Yang, Y.; Yang, J.; Chai, H.; Li, Y.; Yang, J.; Jia, Z.; Wang, Z. Tectoridin, an isoflavone glycoside from the flower of Pueraria lobata, prevents acute ethanol-induced liver steatosis in mice. Toxicology 2010, 276, 64–72. [Google Scholar] [CrossRef] [PubMed]
  107. Mousavi, S.Z.; Bathaie, S.Z. Historical uses of saffron: Identifying potential new avenues for modern research. Avicenna J. Phytomed. 2011, 1, 57–66. [Google Scholar]
  108. WHO. Monographs on Selected Medicinal Plants; World Health Organization: Barcelona, Spain, 2007; Volume 3, pp. 126–135. [Google Scholar]
  109. Srivastava, J.K.; Gupta, S. Extraction, characterization, stability and biological activity of flavonoids isolated from Chamomile flowers. Mol. Cell Pharmacol. 2009, 1, 138–153. [Google Scholar] [CrossRef]
  110. Xiao, J. Review dietary flavonoid aglycones and their glycosides: Which show better biological significance? Crit. Rev. Food Sci. Nutr. 2017, 57, 1874–1905. [Google Scholar]
  111. Marksa, M.; Radušienė, J.; Jakštas, V.; Ivanauskas, L.; Marksienė, R. Development of an HPLC post-column antioxidant assay for Solidago canadensis radical scavengers. Nat. Prod. Res. 2015, 30, 536–543. [Google Scholar] [CrossRef]
  112. International Conference on Harmonization (ICH). Validation of Analytical Procedures: Text and Methodology; Q2 (R1); ICH Secretariat: Geneva, Switzerland, 2005; p. 17. [Google Scholar]
Figure 1. The QbD approach to quality assurance in Crocus perianth drugs production. Adopted by [5,6,19].
Figure 1. The QbD approach to quality assurance in Crocus perianth drugs production. Adopted by [5,6,19].
Scipharm 90 00019 g001
Figure 2. Risk assessment of C. sativus perianth extracts obtaining using the Ishikawa method.
Figure 2. Risk assessment of C. sativus perianth extracts obtaining using the Ishikawa method.
Scipharm 90 00019 g002
Figure 3. DoE of obtaining C. sativus crude extracts with potential anticancer activity [14,17,25,30,31].
Figure 3. DoE of obtaining C. sativus crude extracts with potential anticancer activity [14,17,25,30,31].
Scipharm 90 00019 g003
Figure 4. Herb MaRS criteria for chosen of Q-markers for Crocus perianth raw material and crude extracts.
Figure 4. Herb MaRS criteria for chosen of Q-markers for Crocus perianth raw material and crude extracts.
Scipharm 90 00019 g004
Figure 5. The chromatogram of C. sativus perianth fingerprints obtained by HPLC-DAD method at 270 nm (line 1), 310 nm (line 2), and 440 nm (line 3): mangiferin (A), isoorienthin (B), rutin (C), ferulic acid (D), isoquercitrin (E), tectoridin (F), crocin 4 (G), apigenin-7-glucoside (H), quercetin (I), nigricin (J), iristectorignin B (K), kaempferol (L).
Figure 5. The chromatogram of C. sativus perianth fingerprints obtained by HPLC-DAD method at 270 nm (line 1), 310 nm (line 2), and 440 nm (line 3): mangiferin (A), isoorienthin (B), rutin (C), ferulic acid (D), isoquercitrin (E), tectoridin (F), crocin 4 (G), apigenin-7-glucoside (H), quercetin (I), nigricin (J), iristectorignin B (K), kaempferol (L).
Scipharm 90 00019 g005
Figure 6. Chemical structure of selected quality markers and some major compounds of C. sativus perianth.
Figure 6. Chemical structure of selected quality markers and some major compounds of C. sativus perianth.
Scipharm 90 00019 g006
Figure 7. EC50 values of C. sativus perianth extracts against MDA-MB-231 and IGR39 cell lines after 72 h, * p < 0.05, n = 3.
Figure 7. EC50 values of C. sativus perianth extracts against MDA-MB-231 and IGR39 cell lines after 72 h, * p < 0.05, n = 3.
Scipharm 90 00019 g007
Table 1. Failure mode and effect analysis of Crocus sativus perianth extract manufacturing process.
Table 1. Failure mode and effect analysis of Crocus sativus perianth extract manufacturing process.
Risk Area, CCP/QCPFailure ModePotential Cause or Route of FailureDetection or Control MethodRisk AnalysisCorrection Action
SPDRPN
Herbal raw material
QPT
  • Cultivation
  • Seed material
  • Harvest
  • Post-harvest material
Poor cultivation management, inappropriate soil and irrigation; physical properties; long duration of handling and transporting, warm and humid condition; poor personal hygiene in collection; wrong handling by personnel.Compliance with GACP principles of all cultivation processes; certified suppliers with HACCP program; documenting;
visual inspection; botanical identification; soil analysis; metal detector; microbiological analysis.
54480Rejection, sorting, instructions to supplier
43336
54480
54560
Primary processing
CCP
  • Extract content
  • Water and ash content
  • Average size
  • Proportion of powdered
Improper control of temperature and time of drying.
operator error, poor development; material variation.
Calibration of thermometer and timer, maintenance program; personnel training; visual control; monitoring; microbiological analysis; chemical analysis.545100Re-dry, sorting out; repair and replace damaged equipment
53575
43224
43224
Extraction agent CCP
  • Type
  • Concentration
  • Quality
  • Speed of throughput
Uniformity of the extractant concentration; content of extractive compounds; ethanol concentration; operator’s error.Alcoholometry; monitoring; chemical analysis; calibration; temperature control. 54480Instruction to operator
545100
545100
53345
Production process CCP
  • Extraction process type
  • Extraction time
  • Extraction temperature
  • Extraction pressure
  • Batch size
Poor monitoring; operator’s error, equipment failure; machine failure, poor development; improper control of temperature and time, improper sealing of system; uniformity of the raw material.Calibration of thermometer and timer, maintenance program, personnel training; monitoring; visual control; temperature control.54360Repair and replace damaged equipment
54360
54480
54360
54360
Equipment
QPT
  • Filling quality
  • Filling level
  • Static pressure
  • Related operation (filtration, drying)
Defective devices for extraction, evaporation and drying.All equipment should be easily cleaned to minimize contamination; calibration of equipment, personnel training; monitoring; visual control; temperature control.4218Repair and replace damaged equipment
3216
3216
3216
Personal
QPT
  • Work safety
  • Working procedure
  • Staff qualification
  • Trainings
Human error; poor personnel hygiene, wrong handling by personnel.All persons having contact with raw materials should observe a strict level of personal hygiene; personnel training; monitoring; visual control.4218Training, instruction to operator
43224
53230
43224
Adapted according to [29]: S—severity of excursion = 1 (low), 5 (high); P—probability of occurrence = 1 (low), 5 (high); D—detection of probability = 1 (easy), 5 (hard); risk priority number RPN = S × O × D. 1–29 low risk, 30–59 medium risk, 60–125 high risk; CCP, critical control point; QCP, quality control point. The rank for risk quantification of the S, P, and D parameter is presented in Supplementary Table S3.
Table 2. Composition of Crocus sativus perianth and relevant Herb MaRS score based on potential anticancer action.
Table 2. Composition of Crocus sativus perianth and relevant Herb MaRS score based on potential anticancer action.
CompoundActivityHerb MaRS Ranking *Reference
All crocinsAnticancer, cytotoxic, antioxidant, neuroprotective, retinal damage protection, antidepressant, anti-Alzheimer, hypolipidemic, anti-inflammatory.5[41,42,43,44,45,46,47,48,49,50,51]
PicrocrocinAnticancer, antineoplastic, antioxidant.4[45,52]
SafranalAntitussive, anticonvulsant, antioxidant, antianxiety, antidepressant, antinociceptive, anti-ischemia.3[53,54]
Ferulic acidAnticancer, anti-inflammatory, antioxidant, antibacterial, antidiabetic. 5[55,56,57,58]
Caffeic acidAnticancer, antioxidant, anti-inflammatory.5[59,60]
MangiferinAnticancer, antiviral, anti-inflammatory, antidiabetic, antitumor, lipometabolism regulating, cardioprotective, antihyperuricemic, neuroprotective, antioxidant, antipyretic, analgesic, antibacterial, immunomodulatory.5[61,62,63,64,65,66,67]
IsoorientinAnticancer, anti-inflammatory, QS inhibitor, antinociceptive, gastroprotective.5[68,69,70]
Kaempferol-3-O-sophorosideAntiinflammatory, antitumor, antioxidative, antiallergic, antidiabetic. 4[71]
RutinAnticancer, anti-inflammatory, QS inhibitor, antibacterial, antiprotozoal, antitumor, antiallergic, antiviral, cytoprotective, vasoactive, hypolipidaemic, antiplatelet, antispasmodic, antihypertensive.5[72,73,74,75,76,77]
IsoquercitrinAnticancer, antioxidant, antiproliferative, anti-inflammatory, anti-hypertensive, antidiabetic. 5[78,79,80]
ApigeninAnticancer, antiallergic, anti-inflammatory, antioxidant, antimutagenic, anticarcinogenic.5[81,82,83,84]
Apigenin-7-O-glucoside Cytotoxic effect, antifungal, anticancer, antiproliferative.5[85,86,87]
Quercitin and its derivativesAnticancer, antiviral, antiprotozoal, antimicrobial, antiallergic, anti-inflammatory. 5[88,89,90,91]
Kaempferol and its derivativesAntioxidant, anti-inflammatory, antimicrobial, anticancer, cardioprotective, neuroprotective, antidiabetic. 5[92,93,94,95,96]
Astragalin Anticancer, anti-inflammatory, antioxidant, neuroprotective.4[97,98]
Isoramnetin Anticancer, cardiovascular, cerebrovascular protection, anti-inflammatory, antioxidant.4[99,100]
NigricinHigh anti-inflammatory activity.3[101]
Iristectorigenin BLiver X receptor modulator, anti-inflammatory, antioxidant. 4[102,103]
TectoridinAnticancer, anti-inflammatory, antioxidant, hepatoprotectivy, hypoglycemic, antiallergic, anaphylaxis inhibitory. 5[104,105,106]
* The ranking score ranges from 0 to 5, with 0 being the least and 5 being the most suitable.
Table 3. Calibration curves of the reference standard compounds.
Table 3. Calibration curves of the reference standard compounds.
CompoundCalibration Curve aCorrelation Coefficient r2 (n = 6)Linear Range (μg/mL)RSD, %LoD b (ng/mL)LoQ c (ng/mL)
1Mangiferin f(x) = 29,263.5x + 13,863.90.9997950.28–145.001.32310940
2Isoorientinf(x) = 26,559.9x + 2849.650.9999960.73–92.851.41824
3Ferulic acidf(x) = 54,955.4x − 638.3450.9999590.44–56.501.603080
4Rutinf(x) = 16,072.5x + 1499.730.9998790.16–20.241.0796290
5Isoquercitrinf(x) = 24,139.7x + 3904.440.9998940.35–44.561.0273220
6Crocin f(x) = 3789.03x + 220.8360.9995881.15–147.201.28100300
7Tectoridin f(x) = 76,104.4x + 114,1520.9995800.51–260.000.55130400
8Astragalinf(x) = 20,536.0x + 1618.680.9999870.37–47.701.0190270
9Apigenin-7-glucosidef(x) = 38,477.5x + 4025.410.9999250.25–32.000.8253160
10Quercetinf(x) = 39,349.5x + 1454.470.9998500.16–20.080.673190
11Kaempferolf(x) = 29,888.8x + 1814.270.9999240.14–18.320.9037110
12Iristectorigenin Bf(x) = 109,562x + 68,062.70.9996810.23–120.000.8550150
13Nigricin f(x) = 89,415.4x + 103,2880.9994040.35–181.000.3040130
14Safranalf(x) = 39,230.1x – 11,887.20.9995291.33–42.561.35120360
15Caffeic acidf(x) = 57,646.8x − 3853.480.9999220.72–91.921.562060
16Apigeninf(x) = 50,138.3x + 5722.970.9998890.2–25.760.532580
a concentration of compound (mg/mL); y, peak area; b LOD, limit of detection (S/N = 3); c LOQ, limit of quantification (S/N = 10).
Table 4. Compound content (mg/g) in C. sativus perianth and its crude extracts.
Table 4. Compound content (mg/g) in C. sativus perianth and its crude extracts.
CompoundRetention Time, min/λ, nmRaw MaterialCrocus Perianth Extracts
WaterHydroethanolic
Mangiferin13.83/2701.060 ± 0.7511.091 ± 0.0140.885 ± 0.010
Isoorientin16.74/3107.389 ± 0.3690.668 ± 0.112-
Kaempherol-3-O-sophoroside17.94/3102.329 ± 0.114--
Rutin20.46/310160.45 ± 7.80581.157 ± 0.58065.785 ± 1.089
Ferulic acid22.74/3100.025 ± 0.080.045 ± 0.0040.247 ± 0.003
Isoquercitrin24.76/3502.704 ± 0.1041.785 ± 0.0041.322 ± 0.026
Tectoridin31.34/2702.230 ± 0.0931.423 ± 0.0030.921 ± 0.070
Apigenin-7-O-glucoside31.41/3408.114 ± 0.3872.587 ± 1.587-
trans-Crocin 438.00/4402.662 ± 0.1133.788 ± 0.0150.203 ± 0.003
trans-Crocin 240.59/4402.299 ± 0.109--
Quercetin43.63/3100.481 ± 0.0190.229 ± 0.5400.253 ± 0.003
cis-Crocin 447.07/4400.591 ± 0.028--
cis-Crocin 348.36/4401.356 ± 0.0570.809 ± 0.361-
Nigricin48.94/2700.117 ± 0.0150.052 ± 1.1370.099 ± 0.022
Iristectorigenin B49.15/2700.142 ± 0.050.139 ± 0.0120.142 ± 0.006
Kaempferol49.43/3100.916 ± 0.0310.820 ± 0.0031.018 ± 0.021
- = Not detected; values are presented as mean ± standard deviation from triplicate investigations. Statistical comparisons were performed using ANOVA test (p < 0.05).
Table 5. The radical scavenging activity of individual compounds expressed as TEAC (mmol/g) between C. sativus perianth extracts using the ABTS post-column assay.
Table 5. The radical scavenging activity of individual compounds expressed as TEAC (mmol/g) between C. sativus perianth extracts using the ABTS post-column assay.
CompoundRetention TimeWater ExtractHydroethanolic Extract
Mangiferin15.3415.76 ± 0.28128.13 ± 2.25
Isoorientin18.675.51 ± 0.10-
Rutin22.427.78 ± 0.143.77 ± 0.07
Ferulic acid23.779.12 ± 0.16110.15 ± 1.94
Tectoridin29.9015.16 ± 0.2711.60 ± 0.20
Quercetin44.3710.05 ± 0.18121.11 ± 2.13
Apigenin-7-O-glucoside46.826.13 ± 0.11-
Iristectorigenin B51.7113.47 ± 0.2423.06 ± 0.41
Nigricin52.885.66 ± 0.103.04 ± 0.05
Total88.64 ± 1.56400.86 ± 7.05
- = No activity in ABTS post-column assay. Trolox equivalent (0.3995 µmol/g) antioxidant capacity (TEAC) was used to express antioxidant activity of Crocus extracts; values are presented as mean ± standard deviation from triplicate investigations. Statistical comparisons were performed using ANOVA test (p < 0.05).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mykhailenko, O.; Ivanauskas, L.; Bezruk, I.; Petrikaitė, V.; Georgiyants, V. Application of Quality by Design Approach to the Pharmaceutical Development of Anticancer Crude Extracts of Crocus sativus Perianth. Sci. Pharm. 2022, 90, 19. https://doi.org/10.3390/scipharm90010019

AMA Style

Mykhailenko O, Ivanauskas L, Bezruk I, Petrikaitė V, Georgiyants V. Application of Quality by Design Approach to the Pharmaceutical Development of Anticancer Crude Extracts of Crocus sativus Perianth. Scientia Pharmaceutica. 2022; 90(1):19. https://doi.org/10.3390/scipharm90010019

Chicago/Turabian Style

Mykhailenko, Olha, Liudas Ivanauskas, Ivan Bezruk, Vilma Petrikaitė, and Victoriya Georgiyants. 2022. "Application of Quality by Design Approach to the Pharmaceutical Development of Anticancer Crude Extracts of Crocus sativus Perianth" Scientia Pharmaceutica 90, no. 1: 19. https://doi.org/10.3390/scipharm90010019

Article Metrics

Back to TopTop