Next Article in Journal
Investigation on the Combined Effect of Hydroxypropyl Beta-Cyclodextrin (HPβCD) and Polysorbate in Monoclonal Antibody Formulation
Next Article in Special Issue
Evaluating the Role of Neddylation Modifications in Kidney Renal Clear Cell Carcinoma: An Integrated Approach Using Bioinformatics, MLN4924 Dosing Experiments, and RNA Sequencing
Previous Article in Journal
Development and Validation of a Capillary Zone Electrophoresis–Tandem Mass Spectrometry Method for Simultaneous Quantification of Eight β-Lactam Antibiotics and Two β-Lactamase Inhibitors in Plasma Samples
Previous Article in Special Issue
Advances in IL-7 Research on Tumour Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Potential Antitumor Activity of Combined Lycopene and Sorafenib against Solid Ehrlich Carcinoma via Targeting Autophagy and Apoptosis and Suppressing Proliferation

by
Thanaa A. El-Masry
1,*,†,
Maysa M. F. El-Nagar
1,*,†,
Nageh A. El Mahdy
1,
Fatemah A. Alherz
2,
Reham Taher
1 and
Enass Y. Osman
1
1
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
2
Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2024, 17(4), 527; https://doi.org/10.3390/ph17040527
Submission received: 28 February 2024 / Revised: 13 April 2024 / Accepted: 14 April 2024 / Published: 19 April 2024
(This article belongs to the Special Issue Adjuvant Therapies for Cancer Treatment)

Abstract

:
An FDA-approved kinase inhibitor called sorafenib (SOR) is used to treat primary kidney and liver cancer as well as to stop the spread of advanced breast cancer. Side effects from SOR, such as palmar–plantar erythrodysesthesia syndrome, can negatively impact an individual’s quality of life. There are a lot of data supporting the importance of lycopene (LYC) in preventing cancer. The antitumor properties of the combination of sorafenib and lycopene were examined in this study. A viability test against MDA-MB-231 was used to assess the anticancer efficacy of sorafenib, lycopene, and their combination in vitro. Moreover, a cell cycle analysis and Annexin-V/PI double staining were performed by using flow cytometry. In addition, the protein level of JNK-1, ERK-1, Beclin-1, P38, and P53 of the MDA-MB-231 cell line was estimated using ELISA kits. In addition, mice with SEC were divided into four equal groups at random (n = 10) to investigate the possible processes underlying the in vivo antitumor effect. Group IV (SEC-SOR-LYC) received SOR (30 mg/kg/day, p.o.) and LYC (20 mg/kg/day, p.o.); Group I received the SEC control; Group II received SEC-SOR (30 mg/kg/day, p.o.); and Group III received SEC-LYC (20 mg/kg/day, p.o.). The findings demonstrated that the combination of sorafenib and lycopene was superior to sorafenib and lycopene alone in causing early cell cycle arrest, suppressing the viability of cancer cells, and increasing cell apoptosis and autophagy. Likewise, the combination of sorafenib and lycopene demonstrated inhibition of the levels of Bcl-2, Ki-67, VEGF, IL-1β, and TNF-α protein. Otherwise, the quantities of the proteins BAX, P53, and caspase 3 were amplified. Furthermore, the combined treatment led to a substantial increase in TNF-α, caspase 3, and VEGF gene expression compared to the equivalent dosages of monotherapy. The combination of sorafenib and lycopene enhanced apoptosis and reduced inflammation, as seen by the tumor’s decreased weight and volume, hence demonstrating its potential anticancer effect.

1. Introduction

A medical diagnosis called a tumor is defined by an unnaturally high rate of cell division. Approximately 85% of all human cancers are solid tumors. They are the second most common cause of death [1]. According to the Global Cancer Statistics 2020 study, there were 10 million cancer-related deaths and 19.3 million newly diagnosed cancer patients [2]. The most frequent malignancy among women is breast cancer, of which there were more than 2 million new cases [3].
An Ehrlich tumor develops from a spontaneous adenocarcinoma of the mouse breast and becomes ascites via successive intraperitoneal transfers. The Ehrlich ascites model is widely employed in experimental cancer research because of its capacity to generate cancerous cells and its precision in forecasting survival duration [4]. A localized inflammatory response takes place in solid Ehrlich carcinoma (SEC), which increases the vascular permeability over time. Tumor growth is dependent on the development of severe edema, increasing ascetic fluid production and cellular migration, both of which can be brought on by these responses [5,6].
Radiation therapy, which employs high-energy radiation to destroy cancer cells, and surgery are two breast cancer treatment options. Chemotherapeutic medications, such as cisplatin, shrink or kill cancer cells while also potentially affecting rapidly proliferating normal healthy cells. In addition, hormonal therapy is utilized to prevent cancer cells from receiving the growth hormones they require, such as tamoxifen. Other therapeutic options include targeted drug therapy such as monoclonal antibodies and kinase inhibitors [7].
Although anticancer technology has advanced significantly, there is still no effective therapy for malignant disorders. The most common method used for cancer treatment is chemotherapy, whose main goal is to eradicate cancer cells while sparing nearby healthy cells [8]. Cytotoxic medications, however, lack specificity in practice, which can result in systemic toxicity and serious side effects such as mouth ulcers, hair loss, nephrotoxicity, lung toxicity, cardiotoxicity, bone marrow suppression, and skin changes [7,8]. As a result, the need to assess novel, potent anticancer medications is constant.
Significant advancements in treatment have been made recently, such as stem cell therapy, targeted therapy, nanoparticles, natural antioxidants, radionics, and ferroptosis-based therapy [8]. From a study of clinical trials, it was shown that cancer medicine has made remarkable strides recently toward more useful, accurate, and minimally invasive cancer therapies [8]. The terms stem cell, targeted treatment, immunotherapy, gene therapy, and natural antioxidant are now the most frequently used since they show great promise and effectiveness [9,10,11,12].
Sorafenib (SOR) is an oral, broad-spectrum, small molecule used for the treatment of different types of cancer (liver, thyroid, and kidney) [13]. It is a multikinase inhibitor that dose-dependently inhibits numerous kinases, including VEGFR, PDGFR, and others [13]. It also inhibits proliferation, apoptotic effects, and angiogenesis [14]. Unfortunately, SOR has side effects like rashes, fatigue, diarrhea, hair loss, nausea, and other serious side effects that impair the quality of life, such as hand–foot syndrome (palmar–plantar erythrodysesthesia) starting 5–6 weeks after the start of treatment [15]. An adjuvant for sorafenib therapy may lessen adverse effects while simultaneously boosting antitumor activity [16].
Phytochemicals like lycopene (LYC), in combination with anticancer drugs, could affect the reserves of tumor growth and the metastasis of tumors, and lycopene can be an adjuvant applicant for the management of cancer by sorafenib [16,17]. LYC is a lipophilic natural chemical with an acyclic and tetraterpene hydrocarbon component that includes 11 conjugated and 2 non-conjugated double bonds [18]. It can be found in nature as a structure with all transforms. In addition to tomatoes, it is the most prevalent carotenoid in apricots, melons, papayas, grapes, peaches, watermelons, and cranberries [18]. A multitude of functions, including antioxidation [19], anti-inflammation [20], immunomodulation [20], improved gap junctional communication [21], induction of phase II enzymes [22], inhibition of cell proliferation [23], anticancer [24,25], anti-angiogenesis [24,25,26], and anti-metastasis [27,28,29], have been demonstrated by studies involving lycopene.
Thus, the purpose of this study was to determine if using LYC as an adjuvant therapy with SOR may have anticancer effects.

2. Results

2.1. In Vitro Study

2.1.1. Sorafenib, Lycopene, and Their Combination Demonstrated Anticancer Efficacy Using the MTT Viability Test

SOR, LYC, and their combination’s inhibitory effects on the MDA-MB-231 cell line (breast cancer) are displayed in Figure 1, with IC50 values of 21.33 ± 2.25, 169.17 ± 6.94, and 16.94 ± 2.81 μM, respectively. A total of 10 to 40 μM of SOR was utilized; the lowest concentration (10 μM) showed a maximum viability of 79.69 ± 11.99% and a lesser inhibitory impact of 20.31 ± 1.63%, and the highest concentration (40 μM) showed the highest inhibition of 61.68 ± 1.89% (Figure 1A).
LYC was utilized at concentrations ranging from 20 to 180 μM in the same setting. The lowest concentration (20 μM) showed the maximum viability of 90.70 ± 1.77% and the lowest inhibitory effect of 9.29 ± 0.55%, while the highest LYC (180 μM) recorded the highest inhibition of 55.18 ± 1.62% (Figure 1B).
Additionally, SEC-SOR-LYC was applied in amounts that ranged from 6.25 to 100 μM. Of these, the lowest concentration (6.25 μM) had the highest viability (76.07 ± 8.01%) and the lowest inhibition effect (23.92 ± 2.22%), with the highest inhibition (69.18 ± 10.08%) occurring at the highest concentration (100 μg/mL) (Figure 1C).

2.1.2. Cell Cycle Analysis

Figure 2A–D display the cell cycle analysis of the MDA-MB-231 cell line using flow cytometry at various cell cycle stages (G0/G1, S, and G2/M) on the IC50 concentration detected by MTT viability tests of LYC, SOR, and their combination (IC50 = 21.33 ± 2.25, 169.17 ± 6.94 and 16.94 ± 2.81 μM, respectively; incubation for 48 h). G2/M phase cell cycle arrest was seen in cells treated with SOR, rising from 29.5% in untreated cells to 21.7% in treated cells. Treatment with LYC revealed G2/M phase cell cycle arrest in the same setting, increasing from 29.5% in untreated cells to 25.8% in treated cells. Additionally, cells treated with SOR-LYC showed a severe cell cycle arrest in the G2/M phase, changing from 29.5% of untreated cells to 6.6% of treated cells.

2.1.3. Assay of Apoptosis

Figure 3A–D reveal apoptotic cell populations of the MDA-MB-231 cell line using Annexin-V/PI double staining on the IC50 concentration detected via an MTT viability test of LYC, SOR, and their combination (IC50 = 21.33 ± 2.25, 169.17 ± 6.94 and 16.94 ± 2.81 μM, respectively; incubation for 48 h). The MDA-MB-231 cell line treated with SOR-LYC showed an increase in apoptotic cells from 10.8% in untreated cells to 20.3% in the treated cells.

2.1.4. Determination of JNK-1, ERK-1, Beclin-1, P38 and P53 Content

Figure 4A–E expose the protein level of JNK-1, ERK-1, Beclin-1, P38 and P53 of the MDA-MB-231 cell line using (ELISA) kits on the IC50 concentration detected via an MTT viability test of LYC, SOR, and their combination (IC50 = 21.33 ± 2.25, 169.17 ± 6.94 and 16.94 ± 2.81 μM, respectively; incubation for 48 h). The MDA-MB-231 cell line treated with SOR-LYC displayed an increase in the JNK-1, ERK-1, Beclin-1, P38, and P53 content compared to SOR and LYC treatment alone (Figure 4A–E).

2.2. In Vivo Study

2.2.1. Measurement of Survival Rate

The mouse survival rate was followed during the experiment and described using the Kaplan–Meier survival curve. Treatments with SOR showed a 2.22% increase in the survival rate, while the LYC treatment exhibited an increase in survival by 3.99% compared to the SEC-Control group (Figure 5). Furthermore, the SEC-SOR-LYC group exhibited a higher rise in survival rate by 11.11% compared to the SEC-Control (Figure 5) at the end of the experiment.

2.2.2. Effect of SOR, LYC, and Their Combination on Tumor Volume and Weight

SEC-LYC and SEC-SOR significantly decreased the tumor tissue volume by 43% and 52%, respectively, compared to the SEC-Control. Additionally, in the SEC-SOR-LYC group, the tumor tissue volume decreased significantly by 64.3% compared to the SEC-Control and by 30.1% and 28.8% compared to the SEC-SOR and SEC-LYC groups, respectively (Figure 6A).
SEC-LYC and SEC-SOR significantly decreased the tumor weight by 62% and 64%, respectively, related to the SEC-Control. As well, in the SEC-SOR-LYC group, the tumor tissue weight decreased significantly by 78% compared to the SEC-Control (Figure 6B).

2.2.3. Effect of SOR, LYC, and Their Combination on TNF-α and VEGF Content

SEC-SOR significantly decreased the tumor tissue TNF-α content by 12.5% compared to the SEC-Control. Additionally, in the SEC-SOR-LYC group, the TNF-α content decreased significantly by 34% compared to the SEC-Control and by 32% compared to the SEC-LYC group (Figure 7A).
The VEGF content of tumor tissue was considerably reduced by 18% in the case of SEC-SOR and 19% in the case of SEC-LYC in comparison to the SEC-Control group. The SEC-SOR-LYC group had a notably reduced amount of VEGF in the tumor tissue by 38% in contrast to the SEC-Control group and by 15% and 25% when compared to the SEC-SOR and SEC-LYC groups alone (Figure 7B).

2.2.4. Determination of TNF-α, VEGF, and Caspase 3 Gene Expression

The TNF-α gene expression was dramatically reduced by 20% in the SEC-SOR group and by 35.27% in the SEC-LYC group as compared to the control group. Likewise, the TNF-α gene expression was considerably reduced by 73.2% in the SEC-SOR-LYC group (Figure 8A).
Comparing SEC-LYC and SEC-SOR to the SEC-Control group, in the same setting, VEGF gene expression was dramatically reduced by 59% and 49%, respectively. When comparing the SEC-Control group to the SEC-SOR-LYC group, there was a substantial 86% decrease in VEGF gene expression (Figure 8B).
In terms of caspase 3 gene expression, in the SEC-LYC group it dramatically increased by 1.58-fold and in the SEC-SOR group, the gene was considerably amplified by 1.31-fold in contrast to the SEC-Control group. Similarly, in the SEC-SOR-LYC group, the caspase 3 gene expression was dramatically increased by 2.36-fold (Figure 8C) in comparison to the control group.

2.2.5. Histopathological Assessment

Figure 9 shows the histopathological examination (a, b, c, and d). A strong mitotic activity and a strong proliferative activity of cancerous cells were seen in the SEC-Control group (Figure 9a). Conversely, the SEC-SOR group exhibited a reduction in neoplastic cells and a rise in degenerative and necrotic lesions (Figure 9b). Additionally, the SEC-LYC group demonstrated a reduction in malignant cells and an increase in degenerative and necrotic lesions (Figure 9c). Additionally, the necrotic alterations inside the neoplastic cells were significantly more prevalent in the SEC-SOR-LYC group (Figure 9d).

2.2.6. Immunohistochemical Assessment

The SEC-Control group had high staining for the inflammatory marker, IL-1β, the antiapoptotic marker, Bcl-2, and the proliferation marker, Ki-67, according to immunohistochemical labeling. Conversely, in comparison to the SEC-Control group, several treatments demonstrated a statistically significant reduction in immunoreactivity (Figures S1A–C and S2). Furthermore, apoptotic markers (caspase 3, BAX, and P53) demonstrated a noteworthy decay in immunoreactivity in the SEC-Control group, whereas a noteworthy increase in immunoreactivity was seen with various treatments in comparison to the SEC-Control group (Figures S1D–F and S2).

3. Discussion

About two million new instances of breast cancer are diagnosed each year, making it the second most common disease globally [2]. Currently, one in eight cancer diagnoses are breast cancer, and there are 2.3 million new instances of the disease in both sexes combined [2]. In 2020, it accounted for 25% of all female cancer cases and was by far the most often diagnosed cancer in women. The incidence of this disease has been increasing worldwide, particularly in developing countries [30]. In 2020, an estimated 685,000 women lost their lives to breast cancer, making up about 16% of all female cancer fatalities. The World Health Organization (WHO) has introduced the International Breast Cancer Program as a reaction to the insufficient public health attention to this development in previous decades [2,30]. The goal of the WHO and collaborators is to lower the death rate from breast cancer by promoting early detection, appropriate treatment, and patient care. To achieve this, they will coordinate sustainable efforts to improve outcomes and include global partners. A solid grasp of the worldwide trends and variances in the illness burden is essential to the success of these initiatives.
Although SOR as a single agent has shown good efficiency in breast cancer [31], its survival advantages in large randomized phase III studies remain low, and its clinical effects are mostly limited to transitory tumor stabilization [32]. Moreover, it is associated with substantial unpleasant side effects, and drug resistance frequently develops, highlighting the need for additional effective combinations that could minimize the SOR dosage and the incidence of side effects [33]. Thus, the goal of this investigation was to ascertain if using LYC as an adjuvant therapy with SOR in vitro and vivo may have any anticancer effects.
Lycopene (LYC) is a carotenoid existent in a variety of plants, particularly in ripe red fruits and vegetables [18]. It suppresses the proliferation and carcinogenesis of cancer cells in several organs [34]. Additionally, it has anti-inflammatory, antioxidant, and antiproliferative properties [35]. LYC exerts its effects through direct processes, such as regulating signaling, halting cell cycle progression, causing apoptosis, and altering certain enzymes and antioxidants [36]. These features let LYC inhibit the metastasis, invasion, and angiogenesis of diverse cancer cells. LYC inhibits the phosphorylation of tumor-associated proteins [37].
The findings of an in vitro study demonstrated that the combination of sorafenib and lycopene was superior to sorafenib and lycopene alone in causing early cell cycle arrest, suppressing the viability of cancer cells and increasing cell apoptosis. Moreover, the MDA-MB-231 cell line treated with SOR-LYC exhibited an increase in the autophagy and apoptosis markers JNK-1, ERK-1, Beclin-1, P38, and P53 as compared to SOR and LYC treatment alone. These results were accordant with many other studies that demonstrated and anticancer activity of sorafenib and lycopene [16,38,39,40].
The family of mitogen-activated protein kinases (MAPKs) includes many signaling pathways that respond to oncogenic mutational events and control proliferation, apoptosis, and aerobic glycolysis. There are three well-characterized subfamilies of MAPKs: p38 kinases, c-Jun N-terminal kinases (JNKs), and extracellular signal-regulated kinases (ERKs) [41]. Every MAPK signal is activated by a three-tier kinase module, wherein an MAP3K phosphorylates and initiates an MAP2K, which then phosphorylates and initiates an MAPK as well. MAPKs regulate a variety of cellular responses, including survival, differentiation, proliferation, and cell death, once they are activated [41,42]. Of the three kinds of MAPKs, it has recently been demonstrated that ERKs and JNKs control the activity of important metabolic regulators, which in turn control the redirection of energy to glycolysis in both malignant and highly proliferative cells [43].
Beclin-1 is a crucial autophagy effector that plays a significant part in the apoptotic pathway crosstalk. It has been demonstrated that JNK activation causes Bcl-2 phosphorylation, breaks down the Bcl-2/Beclin 1 complex, and encourages Beclin-1 release to trigger autophagy [44].
Cell cycle arrest and cell death are primarily mediated by p53, which also controls the response to DNA damage [45]. P53 is a crucial transcription regulator; it has been demonstrated that p53 activates both the intrinsic and extrinsic apoptotic pathways. Because of its intricate function in controlling both autophagy and apoptosis, p53 is a significant yet challenging target for cancer treatment [46]. Therapeutics that target p53 to promote apoptosis have the potential to eradicate cancer cells. As an example, treatment with resveratrol activates MAPKs in prostate cancer cells, phosphorylating p53 at S15 and initiating p53-dependent apoptosis [47].
Furthermore, some parameters like the survival rate, tumor volume and weight, and protein and gene expression for TNF-α, IL-1β, VEGF, Ki-67, Bcl2, BAX, P53, and caspase 3 were measured in an in vivo study on the tumor tissue. The results showed that the tumor volume and weight exhibited a significant decrease in all groups alone or in combination, which leads to an increase in survival rate and is a good sign of antitumor effects; however, the tumor volume and weight significantly decreased in the combination group compared to LYC or SOR alone and the controls [48,49].
One of the main characteristics of cancer is inflammation, which is essential for the development, spread, invasion, and angiogenesis of the disease [50].
The incredibly versatile cytokine tumor necrosis factor alfa (TNF-α) is essential for immunological homeostasis, inflammation, and host defense. It can have a variety of effects, including angiogenesis, necrosis, apoptosis, immune cell activation, differentiation, and cell migration, depending on the biological environment. In addition to being extremely important for tumor formation and progression, these pathways are also very relevant for tumor immune surveillance [32]. In the current study, TNF-α significantly decreased in the combination groups compared to the LYC or SOR groups and the control group, which agreed with many studies [15,51,52].
In the same context, one common biomarker linked with inflammation, interleukin 1 beta (IL-1β), is crucial for the diagnosis and treatment of breast cancer [53]. Data both in vitro and in vivo have demonstrated that IL-1β in particular drives immunosuppression, increases angiogenesis and local tumor formation, and encourages the migration and invasion of cancer cells. It also causes a more aggressive cancer phenotype [54,55,56]. Consequently, the outcome clarifies why there was a greater drop in IL-1β in the combination group than in the treatment group alone.
Numerous cells produce VEGF, a vascular permeability factor and signal protein that induces angiogenesis, the response to growth factors, cell permeability, and the formation of blood vessels [57]. The present data indicate that VEGF significantly decreased in the combination group compared to the LYC or SOR groups and the control group, similar to the findings in [58,59,60].
In the same manner, given its strong correlation with the proliferation and growth of tumor cells, Ki-67 is widely used as a proliferation marker for breast cancer. Furthermore, compared to normal tissue, malignant tissues with poorly differentiated tumor cells express much more Ki-67. In the current study, the combination group showed a decline in Ki-67 content in comparison to LYC or SOR alone, and the control group had comparable outcomes to [61,62].
Apoptosis is characterized as a process of planned cell death that preserves the tissue integrity by eliminating unstable cells. The key participants in the setting of triggering the apoptotic cascades are caspase 3, caspase 9, P53, and BAX, and Bcl2 functions as an antiapoptotic mediator. Apoptosis is downregulated and cell growth is upregulated in cancer, which promotes tumor development and proliferation [63]. The ongoing rise in tumor volume in the untreated group in the current data supports this.
In the same manner, during apoptosis, caspase-3 is known as the executioner caspase. Dysregulated apoptosis is a characteristic that sets human cancers apart [64]. In the two main pathways that initiate apoptosis, caspase-3 is essential: the extrinsic [also known as the death receptor (DR)] pathway is initiated by the ligand binding of members of the DR superfamily, which then activates caspase-8 and caspase-3; the intrinsic (also known as the mitochondrial) pathway is initiated by the mitochondria’s release of cytochrome c, which subsequently generates Apaf-1 and cytochrome c. Our study’s findings, which are consistent with [20,65,66,67], showed that in the combination groups, caspase-3 was reduced more than LYC or SOR alone and the control group.
Both apoptosis and autophagy serve as anticancer pathways. Reduced cell death as a result of defective apoptosis is a prevalent characteristic in the onset and spread of cancer. At first, autophagy was further recognized as a tumor suppressor pathway as it aids in the breakdown of carcinogenic chemicals in healthy cells [68]. Since then, though, it has been demonstrated that autophagy plays a far more intricate function in cancer. Since autophagy proteins are involved in type II cell death, autophagy has been associated with additional responsibilities in tumor suppression. It has been demonstrated that type II cell death affects several cancer therapies via a particular autophagy protein [68].
The activation of JNKs leads to cell proliferation or apoptosis and is dependent on the cell type, the nature of the death stimulus, the duration of its activation, and the activities of other signaling pathways [69]. In the absence of NF-κB activation, enhanced JNK activation contributes to TNF-α-induced apoptosis. JNK promotes apoptosis via different mechanisms. Activated JNK translocates to the nucleus and transactivates c-Jun and other transcription factors (e.g., p53), which further transactivate various pro-apoptotic genes, such as Fas-L, Bak, and p53-upregulated modulator of apoptosis [70].
The protein Beclin-1 was initially identified as interacting with the Bcl-2 antiapoptotic proteins. Bcl-2 or Beclin 1 defects impact both autophagy and apoptosis [71]. For example, elevated Beclin-1 expression might liberate BAK/BAX from Bcl-2 to encourage apoptosis, whereas a low Bcl-2 expression could lead to excessive autophagy dependent on Beclin-1. It has been established that Beclin 1 overexpression increases the apoptosis that anticancer medicines produce in cervical cancer cells, making the cancer cells more susceptible to chemotherapy treatments [72]. It has also been observed that Bcl-2 expression inhibition raises Beclin 1 levels and causes breast cancer cells to die [73].
Because Bcl-2 directly binds to Beclin-1 and BAX/BAK to control both mitochondria-dependent apoptosis and Beclin-1-induced autophagy, any medication that suppresses Bcl-2 would possess the capacity to increase apoptosis and autophagy [74].
Additionally, c-Jun N-terminal protein kinase 1 (JNK-1)-mediated Bcl-2 phosphorylation and stressors like hunger control the connection between Beclin-1 and Bcl-2 [44]. Caspases can also control Beclin-1’s function in the interaction between autophagy and apoptosis. Beclin-1 cleavage by caspases lowers Beclin-1 levels in cells, which in turn lowers autophagy levels [75].

4. Materials and Methods

4.1. Drugs and Reagents

Sorafenib was bought as a white powder from BOC Sciences (Shirley, NY, USA). Lycopene was bought as a red powder from Shaanxi Zhengsheng Kangyuan Bio-Medical Co. (Shaanxi, China). PEG and DMSO were bought from Loba Chemie (Mumbai, India). The analytical purity of every other chemical was quite high.

4.2. In Vitro Study

4.2.1. Sorafenib, Lycopene, and Their Combination Demonstrated Anticancer Efficacy Using the MTT Viability Test

The cell line MDA-MB-231 (breast cancer) utilized in this investigation was donated by the National Cancer Institute in Cairo, Egypt. The tumor cells were suspended in the medium at a concentration of 5 × 104 cells/well. Sorafenib (5 µM, 10 µM, 15 µM, 20 µM, 30 µM, and 40 µM), lycopene (20 µM, 60 µM, 100 µM, 140 µM, and 180 µM), and their combination (6.25 µM, 12.5 µM, 25 µM, 50 µM, and 100 µM) were introduced to the cells after 48 h of exponential development [40]. The Vibrant® MTT Cell Proliferation Assay Kit, V-13154, was then added and the mixture was incubated for 4 h at 37 °C. The absorbance was determined using a microplate reader (ELx 800, Bio-Tek Instruments Inc., Winooski, VT, USA) set at 540 nm [76].
The optical density of the treated cells (A) and the untreated cells (B) is represented by the formula for the rate of inhibition (%): (A/B) × 100.
Additionally, GraphPad Prism software (San Diego, CA, USA) was used to calculate IC50.

4.2.2. Analysis of Cell Cycle

The MDA-MB-231 cell line was used to examine the cell cycle distribution using flow cytometry at various cell cycle stages (G0/G1, S, and G2/M) of the IC50 concentration, detected via an MTT viability test of sorafenib, lycopene, and their combination (incubation for 48 h). Exponentially developing cells were plated on a well plate and treated with sorafenib, lycopene, and their combination (incubation for 48 h).
The cell cycle was then determined using flow cytometry (BD AccuriTM C6 plus Flow Cytometer). AccuriTM C6 software (Version 1.6, BD Biosciences, Franklin Lakes, NJ, USA) was utilized to ascertain the proportion of cells in every cell cycle phase [77].

4.2.3. Assay of Apoptosis

The MDA-MB-231 cell line was used to examine the effect of apoptotic cell populations on the IC50 concentrations, detected via an MTT viability test, of sorafenib, lycopene, and their combination (incubation for 48 h). Cells that were undergoing exponential development were put onto a well plate and stimulated by sorafenib, lycopene, and their combination (incubation for 48 h). Annexin-V labeled with a fluorescent dye (Annexin V-FITC, BD Biosciences, Franklin Lakes, NJ, USA) was used to determine the apoptosis. Lastly, just before the BD AccuriTM C6 Plus Flow Cytometer was utilized for analysis, 5 μL of propidium iodide solution was added [78].

4.2.4. Determination of JNK-1, ERK-1, Beclin-1, P38 and P53 Contents

The effects of the protein level of JNK-1, ERK-1, Beclin-1, P38, and P53 of the MDA-MB-231 cell line was estimated using ELISA kits on the IC50 concentration detected via an MTT viability test of sorafenib, lycopene, and their combination (incubation for 48 h). The JNK-1 Elisa kit was purchased from Assay Genie Co. (Windsor Place, Dublin, Ireland), Cat No. (RTFI00924), ERK-1 was purchased from Assay Genie Co, Cat No. (RTFI00754), and Beclin-1 was bought from CUSABIO. Co. (Fannin, Houston, TX, USA), Cat No. (CSB-EL002658RA). Furthermore, P38 and P53 ELISA kits were produced by Assay Genie Co, Cat No. (RTFI00943) and CUSABIO. Co, Cat No. (CSB-E08336r), respectively.

4.3. In Vivo Study

4.3.1. Animals

Swiss albino adult female mice weighing 20–25 g were provided by Tanta University’s National Cancer Institute in Tanta, Egypt. Mice were housed in plastic cages under standard laboratory conditions, which included a 12 h light–dark cycle, 5% relative humidity, and a temperature of 25 degrees Celsius. All of the mice were acclimated for one week before the experiment and were allowed unlimited access to normal food and water. The Tanta University Faculty of Pharmacy’s Ethical and Animal Care Committees approved all experimental procedures (TP/RE/08/22M-0034).

4.3.2. Induction of Solid Tumors in Mice

References for Ehrlich ascites (EAC) or a solid kind of cancer that is easier to grow in suspension within the mouse peritoneum were used [79]. In Swiss albino female mice, the Ehrlich ascites carcinoma (EAC) cells were cultured through several intraperitoneal (IP) passages [58]. The Pharmacology and Experimental Oncology Unit at Cairo University’s National Cancer Institute kindly donated the EAC cells.
To create a solid tumor (SEC) on the right flank of the mice, 1 × 106 live EAC cells were injected subcutaneously. There was a discernible solid tumor after around 12 days [80].

4.3.3. Experimental Design

Ten mice with SEC were divided into four equal groups at random. Group I (SEC-Control) included mice given DMSO, PEG, and saline as a vehicle; Group II (SEC-SOR) included mice given SOR (30 mg/kg/day, p.o.) [81]; Group III (SEC-LYC) included mice given LYC (20 mg/kg/day, p.o.) [82]; and Group IV (SEC-SOR-LYC) included mice given SOR (30 mg/kg/day, p.o.) and LYC (20 mg/kg/day, p.o.). On the twelfth day of SEC induction, a daily oral administration of the vehicle, SOR, LYC, and SEC-SOR-LYC was initiated, and it was continued for another 21 days. In the end, all of the mice were euthanized, and the euthanasia was performed by cervical dislocation, agreeing to the American Veterinary Medical Association’s (AVMA) recommendations for the Euthanasia of Animals (2020 Edition). After removal, the tumors were sectioned. One portion was stored in 10% formalin for the histopathological examination, while the other was stored for further biochemical research in a freezer at −80 °C.

4.3.4. Survival Rate Measurement

During the study, the mice’s survival rate was followed and described using the Kaplan–Meier survival curve, which was intended using the following formula: survival rate = number of surviving mice in a group/total number of mice during the experiment (21 days) after 1, 2 and 3.

4.3.5. Tumor Weight and Volume

After the tumor tissue was cautiously removed, an accurate electronic balance (ADAM®, Maidstone Road, Kingston, Milton Keynes, MK, UK) was used to weigh the sample. From the 12th day till the end of the experiment, a Vernier digital caliper was used to measure the tumor mass (Anyi Instrument Co., Tieshan Road, Qixing Strict, Guilin, China). The tumor volume was then calculated using tumor volume (mm3) = 0.52 AB2, where A and B are the lengths of the main and minor axis lengths, respectively [83].

4.3.6. Determination of TNF-α and VEGF Contents

After pulverizing the tumor tissue in 10 milliliters of ice-cold phosphate-buffered saline (PBS, pH 7.4), the resulting mixture was centrifuged for 15 min at 3000 revolutions per minute. After separation, the supernatant’s levels of VEGF and TNF-α were measured. Shanghai Sunred Biological Technology Co., Ltd. (Hutai Road, Baoshan District, Shanghai, China) supplied the enzyme-linked immunosorbent assay (ELISA) kits, which were utilized following the manufacturer’s instructions, to ascertain the amounts of VEGF and TNF-α in tumor tissues.

4.3.7. Determination of TNF-α, VEGF, and Caspase 3 Gene Expressions

Using beta-actin as a housekeeping gene in qRT-PCR, the relative gene expression of TNF-α, VEGF, and caspase 3 was determined. Table 1 shows an assortment of primer sequences. The TRIzol reagent (15596026) from Life Technologies, Van Allen Way, Carlsbad, CA, USA, was used to extract total RNA. The QuantiTects Reverse Transcription Kit (Qiagen, Germantown, MD, USA) was used to perform the reverse transcription procedure. Primers, complementary DNA amplicons, and Syber green master mix (Maxima SYBR Green/qPCR Master Mix, Thermo Fisher Scientific, Third Avenu, Waltham, MA, USA) were included in the reaction mixtures. The fold change in gene expression concerning the calibrator control group was calculated using the Livak technique [84].

4.3.8. Histopathological Examination

Tumor tissue sections were sliced into 3–5 µm-thick sections and stained with hematoxylin and eosin (H&E) [89]. The characteristics of the histopathological findings were investigated using an Olympus CX21 light microscope (Tokyo, Japan).

4.3.9. Immunohistochemical Examination

The methods for immunohistochemical staining were carried out in accordance with [89]. The antibodies used were P53 (Cat# MA5-12557, 1:300 dilution); Bax (Invitrogen, Van Allen Way, Carlsbad, CA, USA, Cat# MA5-13-0300, 1:100); Bcl2 (Abcam, Kendall Sq, Cambridge, MA, USA, Cat# ab182858, dilution 1:500); caspase 3 (Invitrogen, Cat# PA5-77887, dilution 1/100); Ki-67 (Dako, Stevens Creek Blvd, Santa Clara, CA, USA, Cat# M7248, dilution 1/100); and IL-1b (Cat# P420B, 1:200 dilution). Slides were visualized using a DAB kit, and Mayer’s hematoxylin was used as a counterstain after that. Using Image-J software (version 1.54 D, Java 1.8, 0_354), the staining labeling indices of Bax, caspase 3, and P53 were evaluated and shown as a percentage of positive expression in a total of 1000 cells/8 HPF.

4.3.10. Statistical Analysis

For all statistical studies, Graph Pad Prism 9.5.1 was used. The means ± SD of the data were calculated, and p < 0.05 was chosen as the threshold for statistical significance. Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings.

5. Conclusions

In conclusion, the combination of sorafenib with lycopene showed an anticancer impact in both in vitro and vivo studies through targeting autophagy and apoptosis and suppressing proliferation. Both apoptosis and autophagy serve as anticancer mechanisms. The reduced cell death as a result of defective apoptosis is a typical characteristic in the development of cancer. Since autophagy aids in the breakdown of carcinogenic chemicals in normal cells, it was first thought to be a tumor-suppressing process. Since then though, it has been demonstrated that autophagy plays a far more intricate function in cancer. Because autophagy proteins are involved in tumor suppression, autophagy has associated with new tasks. Also, combining lycopene and sorafenib resulted in more effective remodeling of inflammatory markers. Moreover, the combination therapy demonstrated superiority in impeding the advancement of cancer and restoring critical gene expression levels. Consequently, the combination of sorafenib with lycopene is a valuable anticancer therapy for better treatment results.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph17040527/s1, Figure S1. (A) IL-1β expression (200×, scale bar=100 μm), (B) Bcl-2 expression (200×, scale bar = 100 μm), (C) Ki-67 expression (200×, scale bar = 100 μm), (D) Caspase 3 expression (200×, scale bar = 100 μm, (E) BAX expression (200×, scale bar = 100 μm), (F) P53 expression (200×, scale bar = 100 μm). Figure S2. Immunohistochemical percent of positive area/8 HPF using Image-J software; version 1.54 D, Java 1.8,0_354 (NIH, USA) of (a) IL-1β, (b) Bcl2, (c) Ki-67, (d) Caspase 3, (e) BAX and (f) P5.

Author Contributions

Conceptualization, N.A.E.M., T.A.E.-M., M.M.F.E.-N. and E.Y.O.; methodology, M.M.F.E.-N., E.Y.O. and R.T.; validation, N.A.E.M., T.A.E.-M., M.M.F.E.-N., E.Y.O., F.A.A. and R.T.; formal analysis, N.A.E.M., T.A.E.-M., M.M.F.E.-N., E.Y.O., F.A.A. and R.T.; investigation, N.A.E.M., T.A.E.-M., M.M.F.E.-N., E.Y.O., F.A.A. and R.T.; resources, N.A.E.M., T.A.E.-M., M.M.F.E.-N., E.Y.O., F.A.A. and R.T.; data curation, M.M.F.E.-N. and R.T.; writing—original draft preparation, M.M.F.E.-N. and R.T.; review and editing, N.A.E.M., T.A.E.-M., M.M.F.E.-N., E.Y.O., F.A.A. and R.T.; funding acquisition, F.A.A. All authors have read and agreed to the published version of the manuscript.

Funding

Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2024R322), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.

Institutional Review Board Statement

This in vivo study was accepted by the Research Ethics Committee (Faculty of Pharmacy, Tanta University, Egypt) and it conformed with the guidelines of The Council for International Organizations of Medical Sciences (CIOMS) (Protocol code: TP/RE/08/22M-0034) on date 8/2022.

Informed Consent Statement

This in vivo study was approved by the Research Ethics Review Board (ERB), approval no. (Protocol code: TP/RE/08/22M-0034).

Data Availability Statement

Data are contained within the article.

Acknowledgments

Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2024R322), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Dai, X.; Cheng, H.; Bai, Z.; Li, J. Breast Cancer Cell Line Classification and Its Relevance with Breast Tumor Subtyping. J. Cancer 2017, 8, 3131–3141. [Google Scholar] [CrossRef] [PubMed]
  4. Frajacomo, F.T.; de Souza Padilha, C.; Marinello, P.C.; Guarnier, F.A.; Cecchini, R.; Duarte, J.A.; Deminice, R. Solid Ehrlich carcinoma reproduces functional and biological characteristics of cancer cachexia. Life Sci. 2016, 162, 47–53. [Google Scholar] [CrossRef] [PubMed]
  5. Couch, F.J.; Shimelis, H.; Hu, C.; Hart, S.N.; Polley, E.C.; Na, J.; Hallberg, E.; Moore, R.; Thomas, A.; Lilyquist, J.; et al. Associations between cancer predisposition testing panel genes and breast cancer. JAMA Oncol. 2017, 3, 1190–1196. [Google Scholar] [CrossRef] [PubMed]
  6. Michailidou, K.; Lindström, S.; Dennis, J.; Beesley, J.; Hui, S.; Kar, S.; Lemacon, A.; Soucy, P.; Glubb, D.; Rostamianfar, A.; et al. Association analysis identifies 65 new breast cancer risk loci. Nature 2017, 551, 92–94. [Google Scholar] [CrossRef] [PubMed]
  7. Momenimovahed, Z.; Salehiniya, H. Epidemiological Characteristics of and Risk Factors for Breast Cancer in the World. Breast Cancer Targets Ther. 2019, 11, 151–164. [Google Scholar] [CrossRef] [PubMed]
  8. Debela, D.T.; Muzazu, S.G.; Heraro, K.D.; Ndalama, M.T.; Mesele, B.W.; Haile, D.C.; Kitui, S.K.; Manyazewal, T. New approaches and procedures for cancer treatment: Current perspectives. SAGE Open Med. 2021, 9, 20503121211034366. [Google Scholar] [CrossRef]
  9. Wang, H.; He, X. Nanoparticles for targeted drug delivery to cancer stem cells and tumor. Methods Mol. Biol. 2018, 1831, 59–67. [Google Scholar] [CrossRef]
  10. Zhao, Z.; Zheng, L.; Chen, W.; Weng, W.; Song, J.; Ji, J. Delivery strategies of cancer immunotherapy: Recent advances and future perspectives. J. Hematol. Oncol. 2019, 12, 126. [Google Scholar] [CrossRef]
  11. Sung, Y.K.; Kim, S.W. Recent advances in the development of gene delivery systems. Biomater. Res. 2019, 23, 8. [Google Scholar] [CrossRef]
  12. Vaiserman, A.; Koliada, A.; Zayachkivska, A.; Lushchak, O. Nanodelivery of natural antioxidants: An anti-aging perspective. Front. Bioeng. Biotechnol. 2020, 7, 447. [Google Scholar] [CrossRef]
  13. Tang, W.; Chen, Z.; Zhang, W.; Cheng, Y.; Zhang, B.; Wu, F.; Wang, Q.; Wang, S.; Rong, D.; Reiter, F.P.; et al. The Mechanisms of Sorafenib Resistance in Hepatocellular Carcinoma: Theoretical Basis and Therapeutic Aspects. Signal Transduct. Target Ther. 2020, 5, 87. [Google Scholar] [CrossRef] [PubMed]
  14. Yokota, S.; Yonezawa, T.; Momoi, Y.; Maeda, S. Sorafenib Inhibits Tumor Cell Growth and Angiogenesis in Canine Transitional Cell Carcinoma. J. Vet. Med. Sci. 2022, 84, 666–674. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, J.-C.; Wang, J.-C.; Pan, Y.-X.; Yi, M.-J.; Chen, J.-B.; Wang, X.-H.; Fu, Y.-Z.; Zhang, Y.-J.; Xu, L.; Chen, M.-S.; et al. Preventive Effect of Celecoxib in Sorafenib-Related Hand-Foot Syndrome in Hepatocellular Carcinoma Patients, a Single-Center, Open-Label, Randomized, Controlled Clinical Phase III Trial. Am. J. Cancer Res. 2020, 10, 1467–1476. [Google Scholar]
  16. Chan, Y.P.; Chuang, C.H.; Lee, I.; Yang, N.C. Lycopene in Combination with Sorafenib Additively Inhibits Tumor Metastasis in Mice Xenografted with Lewis Lung Carcinoma Cells. Front. Nutr. 2022, 9, 886988. [Google Scholar] [CrossRef]
  17. Lin, M.T.; Lin, C.L.; Lin, T.Y.; Cheng, C.W.; Yang, S.F.; Lin, C.L.; Wu, C.-C.; Hsieh, Y.-H.; Tsai, J.-P. Synergistic effect of fisetin combined with sorafenib in human cervical cancer HeLa cells through activation of death receptor-5 mediated caspase-8/caspase-3 and the mitochondria-dependent apoptotic pathway. Tumour Biol. 2016, 37, 6987–6996. [Google Scholar] [CrossRef] [PubMed]
  18. Khan, U.M.; Sevindik, M.; Zarrabi, A.; Nami, M.; Ozdemir, B.; Kaplan, D.N.; Selamoglu, Z.; Hasan, M.; Kumar, M.; Alshehri, M.M.; et al. Lycopene: Food sources, biological activities, and human health benefits. Oxid. Med. Cell. Longev. 2021, 2021, 2713511. [Google Scholar] [CrossRef]
  19. Miller, N.J.; Sampson, J.; Candeias, L.P.; Bramley, P.M.; Rice-Evans, C.A. Antioxidant activities of carotenes and xanthophylls. FEBS. Lett. 1996, 384, 240–242. [Google Scholar] [CrossRef]
  20. Huang, C.S.; Chuang, C.H.; Lo, T.F.; Hu, M.L. Antiangiogenic effects of lycopene through immunomodulation of cytokine secretion in human peripheral blood mononuclear cells. J. Nutr. Biochem. 2013, 24, 428–434. [Google Scholar] [CrossRef]
  21. Stahl, W.; Von Laar, J.; Martin, H.D.; Emmerich, T.; Sies, H. Stimulation of gap junctional communication: Comparison of acycloretinoic acid and lycopene. Arch. Biochem. Biophys. 2000, 373, 1271–1274. [Google Scholar] [CrossRef] [PubMed]
  22. Talalay, P. Chemoprotection against cancer by induction of phase 2 enzymes. BioFactors 2000, 12, 5–11. [Google Scholar] [CrossRef] [PubMed]
  23. Levy, J.; Bosin, E.; Feldman, B.; Giat, Y.; Miinster, A.; Danilenko, M.; Sharoni, Y. Lycopene is a more potent inhibitor of human cancer cell proliferation than either alpha-carotene or beta-carotene. Nutr. Cancer 1995, 24, 257–266. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, C.M.; Yen, Y.T.; Huang, C.S.; Hu, M.L. Growth inhibitory efficacy of lycopene and β-carotene against androgen-independent prostate tumor cells xenografted in nude mice. Mol. Nutr. Food Res. 2011, 55, 606–612. [Google Scholar] [CrossRef] [PubMed]
  25. Tang, F.Y.; Pai, M.H.; Wang, X.D. Consumption of lycopene inhibits the growth and progression of colon cancer in a mouse xenograft model. J. Agric. Food Chem. 2011, 59, 9011–9021. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, M.L.; Lin, Y.H.; Yang, C.M.; Hu, M.L. Lycopene inhibits angiogenesis both in vitro and in vivo by inhibiting the MMP-2/uPA system through VEGFR2-mediated PI3K-Akt and ERK/p38 signaling pathways. Mol. Nutr. Food Res. 2012, 56, 889–899. [Google Scholar] [CrossRef] [PubMed]
  27. Huang, C.S.; Shih, M.K.; Chuang, C.H.; Hu, M.L. Lycopene inhibits cell migration and invasion and upregulates Nm23-H1 in a highly invasive hepatocarcinoma, SK-Hep-1 cells. J. Nutr. 2005, 135, 2119–2123. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, C.S.; Liao, J.W.; Hu, M.L. Lycopene inhibits experimental metastasis of human hepatoma SK-Hep-1 cells in athymic nude mice. J. Nutr. 2008, 138, 538–543. [Google Scholar] [CrossRef] [PubMed]
  29. Jhou, B.Y.; Song, T.Y.; Lee, I.; Hu, M.L.; Yang, N.C. Lycopene inhibits metastasis of human liver adenocarcinoma SK-Hep-1 cells by downregulation of NADPH oxidase 4 protein expression. J. Agric. Food Chem. 2017, 65, 12893–12903. [Google Scholar] [CrossRef]
  30. Heer, E.; Harper, A.; Escandor, N.; Sung, H.; McCormack, V.; Fidler-Benaoudia, M.M. Global Burden and Trends in Premenopausal and Postmenopausal Breast Cancer: A Population-Based Study. Lancet Glob. Health 2020, 8, e1027–e1037. [Google Scholar] [CrossRef]
  31. Bronte, G.; Andreis, D.; Bravaccini, S.; Maltoni, R.; Cecconetto, L.; Schirone, A.; Farolfi, A.; Fedeli, A.; Serra, P.; Donati, C.; et al. Sorafenib for the Treatment of Breast Cancer. Expert Opin. Pharmacother. 2017, 18, 621–630. [Google Scholar] [CrossRef] [PubMed]
  32. Gao, J.J.; Shi, Z.Y.; Xia, J.F.; Inagaki, Y.; Tang, W. Sorafenib-Based Combined Molecule Targeting in Treatment of Hepatocellular Carcinoma. World J. Gastroenterol. 2015, 21, 12059–12070. [Google Scholar] [CrossRef] [PubMed]
  33. Zhu, Y.J.; Zheng, B.; Wang, H.Y.; Chen, L. New Knowledge of the Mechanisms of Sorafenib Resistance in Liver Cancer. Acta Pharmacol. Sin. 2017, 38, 614–622. [Google Scholar] [CrossRef] [PubMed]
  34. Kapała, A.; Szlendak, M.; Motacka, E. The Anti-Cancer Activity of Lycopene: A Systematic Review of Human and Animal Studies. Nutrients 2022, 14, 5152. [Google Scholar] [CrossRef] [PubMed]
  35. Qi, W.J.; Sheng, W.S.; Peng, C.; Xiaodong, M.; Yao, T.Z. Investigating into Anti-Cancer Potential of Lycopene: Molecular Targets. Biomed. Pharmacother. 2021, 138, 111546. [Google Scholar] [CrossRef] [PubMed]
  36. Mirahmadi, M.; Azimi-Hashemi, S.; Saburi, E.; Kamali, H.; Pishbin, M.; Hadizadeh, F. Potential Inhibitory Effect of Lycopene on Prostate Cancer. Biomed. Pharmacother. 2020, 129, 110459. [Google Scholar] [CrossRef] [PubMed]
  37. Marzocco, S.; Singla, R.K.; Capasso, A. Multifaceted Effects of Lycopene: A Boulevard to the Multitarget-Based Treatment for Cancer. Molecules 2021, 26, 5333. [Google Scholar] [CrossRef] [PubMed]
  38. Yang, S.; Zhang, B.; Gong, X.; Wang, T.; Liu, Y.; Yang, S. In vivo biodistribution, biocompatibility, and efficacy of sorafenib-loaded lipid-based nanosuspensions evaluated experimentally in cancer. Int. J. Nanomed. 2016, 11, 12329–12343. [Google Scholar] [CrossRef]
  39. Yao, X.; Zhao, C.; Yin, H.; Wang, K.; Gao, J.-J. Synergistic antitumor activity of sorafenib and artesunate in hepatocellular carcinoma cells. Acta Pharmacol. Sin. 2020, 41, 1609–1620. [Google Scholar] [CrossRef]
  40. Abdu, S.; Juaid, N.; Amin, A.; Moulay, M.; Miled, N. Effects of Sorafenib and Quercetin Alone or in Combination in Treating Hepatocellular Carcinoma: In Vitro and In Vivo Approaches. Molecules 2022, 27, 8082. [Google Scholar] [CrossRef]
  41. Cargnello, M.; Roux, P.P. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 120–183. [Google Scholar] [CrossRef]
  42. Raman, M.; Chen, W.; Cobb, M.H. Differential regulation and properties of MAPKs. Oncogene 2007, 26, 12100–12112. [Google Scholar] [CrossRef]
  43. Papa, S.; Choy, P.M.; Bubici, C. The ERK and JNK pathways in the regulation of metabolic reprogramming. Oncogene 2019, 38, 2223–2240. [Google Scholar] [CrossRef]
  44. Wei, Y.; Pattingre, S.; Sinha, S.; Bassik, M.; Levine, B. JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol. Cell 2008, 30, 678–688. [Google Scholar] [CrossRef]
  45. Crighton, D.; Woiwode, A.; Zhang, C.; Mandavia, N.; Morton, J.P.; Warnock, L.J.; Milner, J.; White, R.J.; Johnson, D.L. P53 represses RNA polymerase III transcription by targeting TBP and inhibiting promoter occupancy by TFIIIB. EMBO J. 2003, 22, 2810–2820. [Google Scholar] [CrossRef]
  46. Maiuri, M.C.; Zalckvar, E.; Kimchi, A.; Kroemer, G. Self-eating and self-killing: Crosstalk between autophagy and apoptosis. Nature reviews. Mol. Cell Biol. 2007, 8, 741–752. [Google Scholar] [CrossRef]
  47. Lin, H.Y.; Shih, A.; Davis, F.B.; Tang, H.Y.; Martino, L.J.; Bennett, J.A.; Davis, P.J. Resveratrol-induced serine phosphorylation of p53 causes apoptosis in a mutant p53 prostate cancer cell line. J. Urol. 2002, 168, 748–755. [Google Scholar] [CrossRef] [PubMed]
  48. Fendrich, V.; Maschuw, K.; Rehm, J.; Buchholz, M.; Holler, J.P.; Slater, E.P.; Bartsch, D.K.; Waldmann, J. Sorafenib inhibits tumor growth and improves survival in a transgenic mouse model of pancreatic islet cell tumors. Sci. World J. 2012, 2021, 529151. [Google Scholar] [CrossRef] [PubMed]
  49. Connolly, P.F.; Jäger, R.; Fearnhead, H.O. New roles for old enzymes: Killer caspases as the engine of cell behavior changes. Front. Physiol. 2014, 5, 149. [Google Scholar] [CrossRef]
  50. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 1246–1274. [Google Scholar] [CrossRef]
  51. Cha, J.H.; Kim, W.K.; Ha, A.W.; Kim, M.H.; Chang, M.J. Anti-inflammatory effect of lycopene in SW480 human colorectal cancer cells. Nutr. Res. Pract. 2017, 11, 120–196. [Google Scholar] [CrossRef] [PubMed]
  52. Mekuria, A.N.; Tura, A.K.; Hagos, B.; Sisay, M.; Abdela, J.; Mishore, K.M.; Motbaynor, B. Anti-Cancer Effects of Lycopene in Animal Models of Hepatocellular Carcinoma: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2020, 11, 1306. [Google Scholar] [CrossRef] [PubMed]
  53. Idris, A.; Ghazali, N.B.; Koh, D. Interleukin 1β—A Potential Salivary Biomarker for Cancer Progression? Biomark. Cancer 2015, 7, 25–29. [Google Scholar] [CrossRef] [PubMed]
  54. Gelfo, V.; Romaniello, D.; Mazzeschi, M.; Sgarzi, M.; Grilli, G.; Morselli, A.; Manzan, B.; Rihawi, K.; Lauriola, M. Roles of il-1 in cancer: From tumor progression to resistance to targeted therapies. Int. J. Mol. Sci. 2020, 21, 6009. [Google Scholar] [CrossRef] [PubMed]
  55. Mantovani, A.; Dinarello, C.A.; Molgora, M.; Garlanda, C. Interleukin-1 and related cytokines in the regulation of inflammation and immunity. Immunity 2019, 50, 1278–1295. [Google Scholar] [CrossRef] [PubMed]
  56. Das, S.; Shapiro, B.; Vucic, E.A.; Vogt, S.; Bar-Sagi, D. Tumor cell-derived IL1β promotes desmoplasia and immune suppression in pancreatic cancer. Cancer Res. 2020, 80, 12088–12101. [Google Scholar] [CrossRef] [PubMed]
  57. Johnson, K.E.; Wilgus, T.A. Vascular Endothelial Growth Factor and Angiogenesis in the Regulation of Cutaneous Wound Repair. Adv. Wound Care 2014, 3, 1247–1661. [Google Scholar] [CrossRef]
  58. Abd-Alhaseeb, M.M.; Zaitone, S.A.; Abou-El-Ela, S.H.; Moustafa, Y.M. Olmesartan Potentiates the Anti-Angiogenic Effect of Sorafenib in Mice Bearing Ehrlich’s Ascites Carcinoma: Role of Angiotensin. PLoS ONE 2014, 9, e85891. [Google Scholar] [CrossRef] [PubMed]
  59. Brose, M.S.; Frenette, C.T.; Keefe, S.M.; Stein, S.M. Management of Sorafenib-related adverse events: A clinician’s perspective. Semin. Oncol. 2014, 41, S1–S16. [Google Scholar] [CrossRef]
  60. Ishihara, S.; Onoda, N.; Noda, S.; Asano, Y.; Tauchi, Y.; Morisaki, T.; Kashiwagi, S.; Takashima, T.; Ohira, M. Sorafenib inhibits vascular endothelial cell proliferation stimulated by anaplastic thyroid cancer cells regardless of BRAF mutation status. Int. J. Oncol. 2019, 55, 1069–1076. [Google Scholar] [CrossRef]
  61. Alferraly, I.T.; Munir, D.; Putra, I.B.; Sembiring, R.J. Correlation of Ki-67 Expression as Tumor Cell Proliferation Activity Marker with Cutaneous Squamous Cell Carcinoma Grading. Maced. J. Med. Sci. 2019, 7, 3384–3386. [Google Scholar] [CrossRef] [PubMed]
  62. Sun, X.; Kaufman, P.D. Ki-67: More than a proliferation marker. Chromosoma 2018, 127, 175–186. [Google Scholar] [CrossRef] [PubMed]
  63. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, Pyroptosis, and Apoptosis: An Intricate Game of Cell Death. Cell Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
  64. Pu, X.; Storr, S.J.; Zhang, Y.; Rakha, E.A.; Green, A.R.; Ellis, I.O.; Martin, S.G. Caspase-3 and caspase-8 expression in breast cancer: Caspase-3 is associated with survival. Apoptosis 2017, 22, 1257–1368. [Google Scholar] [CrossRef] [PubMed]
  65. Youssef, M.M.; Tolba, M.F.; Badawy, N.N.; Liu, A.W.; El-Ahwany, E.; Khalifa, A.E.; Zada, S.; Abdel-Naim, A.B. The novel combination of sorafenib and biochanin—A synergistically enhances the anti-proliferative and pro-apoptotic effects on hepatocellular carcinoma cells. Sci. Rep. 2016, 6, 30717. [Google Scholar] [CrossRef]
  66. Busche, S.; John, K.; Wandrer, F.; Vondran, F.W.R.; Lehmann, U.; Wedemeyer, H.; Essmann, F.; Schulze-Osthoff, K.; Bantel, H. BH3-only protein expression determines hepatocellular carcinoma response to sorafenib-based treatment. Cell Death Dis. 2021, 12, 736. [Google Scholar] [CrossRef] [PubMed]
  67. Lee, J.; Lim, J.W.; Kim, H. Lycopene Inhibits IL-6 Expression by Upregulating NQO1 and HO-1 via Activation of Nrf2 in Ethanol/Lipopolysaccharide-Stimulated Pancreatic Acinar Cells. Antioxidants 2022, 11, 519. [Google Scholar] [CrossRef] [PubMed]
  68. Su, Z.; Yang, Z.; Xu, Y.; Chen, Y.; Yu, Q. Apoptosis, autophagy, necroptosis, and cancer metastasis. Mol. Cancer 2015, 14, 48. [Google Scholar] [CrossRef] [PubMed]
  69. Jing, L.; Anning, L. Role of JNK activation in apoptosis: A double-edged sword. Cell Res. 2005, 15, 36–42. [Google Scholar] [CrossRef]
  70. Dhanasekaran, D.N.; Reddy, E.P. JNK signaling in apoptosis. Oncogene 2008, 27, 6245–6251. [Google Scholar] [CrossRef]
  71. Liang, X.H.; Kleeman, L.K.; Jiang, H.H.; Gordon, G.; Goldman, J.E.; Berry, G.; Herman, B.; Levine, B. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J. Virol. 1998, 72, 8586–8596. [Google Scholar] [CrossRef] [PubMed]
  72. Sun, Y.; Liu, J.H.; Jin, L.; Lin, S.M.; Yang, Y.; Sui, Y.X.; Shi, H. Over-expression of the Beclin1 gene upregulates chemosensitivity to anti-cancer drugs by enhancing therapy-induced apoptosis in cervix squamous carcinoma CaSki cells. Cancer Lett. 2010, 294, 204–210. [Google Scholar] [CrossRef] [PubMed]
  73. Akar, U.; Chaves-Reyez, A.; Barria, M.; Tari, A.; Sanguino, A.; Kondo, Y.; Kondo, S.; Arun, B.; Lopez-Berestein, G.; Ozpolat, B. Silencing of Bcl-2 expression by small interfering RNA induces autophagic cell death in MCF-7 breast cancer cells. Autophagy 2008, 4, 669–679. [Google Scholar] [CrossRef] [PubMed]
  74. Oltersdorf, T.; Elmore, S.W.; Shoemaker, A.R.; Armstrong, R.C.; Augeri, D.J.; Belli, B.A.; Bruncko, M.; Deckwerth, T.L.; Dinges, J.; Hajduk, P.J.; et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumors. Nature 2005, 435, 677–681. [Google Scholar] [CrossRef] [PubMed]
  75. Rohn, T.T.; Wirawan, E.; Brown, R.J.; Harris, J.R.; Masliah, E.; Vandenabeele, P. Depletion of Beclin-1 due to proteolytic cleavage by caspases in the Alzheimer’s disease brain. Neurobio. Dis. 2011, 43, 68–78. [Google Scholar] [CrossRef] [PubMed]
  76. Mosmann, T. Rapid Colorimetric Assay for Cellular Growth and Survival: Application to Proliferation and Cytotoxicity Assays. J. Immun. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef] [PubMed]
  77. D’yakonov, V.A.; Makarov, A.A.; Dzhemileva, L.U.; Ramazanov, I.R.; Makarova, E.K.; Dzhemilev, U.M. Natural Trienoic Acids as Anticancer Agents: First Stereoselective Synthesis, Cell Cycle Analysis, Induction of Apoptosis, Cell Signaling and Mitochondrial Targeting Studies. Cancers 2021, 13, 1808. [Google Scholar] [CrossRef] [PubMed]
  78. Nie, T.; Wu, H.; Wong, K.H.; Chen, T. Facile Synthesis of Highly Uniform Selenium Nanoparticles Using Glucose as the Reductant and Surface Decorator to Induce Cancer Cell Apoptosis. J. Mater. Chem. B 2016, 4, 2351–2358. [Google Scholar] [CrossRef] [PubMed]
  79. Ozaslan, M.; Karagoz, I.D.; Kilic, I.H.; Guldur, M.E. Ehrlich Ascites Carcinoma. Afr. J. Biotechnol. 2011, 10, 2375–2378. [Google Scholar]
  80. Mishra, S.; Tamta, A.K.; Sarikhani, M.; Desingu, P.A.; Kizkekra, S.M.; Pandit, A.S.; Kumar, S.; Khan, D.; Raghavan, S.C.; Sundaresan, N.R. Subcutaneous Ehrlich Ascites Carcinoma Mice Model for Studying Cancer-Induced Cardiomyopathy. Sci. Rep. 2018, 8, 5599. [Google Scholar] [CrossRef]
  81. Gupta, S.K.; Trivedi, D.; Srivastava, S.; Joshi, S.; Halder, N.; Verma, S.D. Lycopene Attenuates Oxidative Stress Induced Experimental Cataract Development: An in Vitro and in Vivo Study. Nutrition 2003, 19, 794–799. [Google Scholar] [CrossRef] [PubMed]
  82. Mandal, P.K.; Roy, R.G.; Samaria, A. Oxidative Stress: Glutathione and Its Potential to Protect Methionine-35 of Aβ Peptide from Oxidation. ACS Omega 2022, 26, 27052–27061. [Google Scholar] [CrossRef] [PubMed]
  83. Livak, K. Undefined Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT Method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef]
  84. Khan, H.A.; Abdelhalim, M.A.K.; Alhomida, A.S.; Al Ayed, M.S. Transient Increase in IL-1β, IL-6 and TNF-α Gene Expression in Rat Liver Exposed to Gold Nanoparticles. Genet. Mol. Res. 2013, 12, 5851–5857. [Google Scholar] [CrossRef] [PubMed]
  85. Peng, J.; Lai, Z.G.; Fang, Z.L.; Xing, S.; Hui, K.; Hao, C.; Jin, Q.; Qi, Z.; Shen, W.J.; Dong, Q.N.; et al. Dimethyloxalylglycine Prevents Bone Loss in Ovariectomized C57BL/6J Mice through Enhanced Angiogenesis and Osteogenesis. PLoS ONE 2014, 9, e112744. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, K.; Shi, Y.; Song, Y.; Wang, Y.; Liang, X.; Hu, Y.; Guan, X.; Cheng, J. p,p′-DDE Induces Apoptosis of Rat Sertoli Cells via a Fasl-Dependent Pathway. J. Biomed. Biotechnol. 2009, 2009, 181282. [Google Scholar] [CrossRef]
  87. Sisto, F.; Miluzio, A.; Leopardi, O.; Mirra, M.; Boelaert, J.R.; Taramelli, D. Differential Cytokine Pattern in the Spleens and Livers of BALB/c Mice Infected with Penicillium Marneffei: Protective Role of Gamma Interferon. Infect. Immun. 2003, 71, 465–473. [Google Scholar] [CrossRef] [PubMed]
  88. Kenerson, H.L.; Aicher, L.D.; True, L.D.; Yeung, R.S. Activated mammalian target of rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal tumors. Cancer Res. 2002, 62, 5645–5650. [Google Scholar]
  89. Khalil, R.; Shata, A.; Abd El-Kader, E.M.; Sharaf, H.; Abdo, W.S.; Amin, N.A.; Saber, S. Vildagliptin, a DPP-4 Inhibitor, Attenuates Carbon Tetrachloride-Induced Liver Fibrosis by Targeting ERK1/2, P38α, and NF-ΚB Signaling. Toxicol. Appl. Pharmacol. 2020, 407, 115246. [Google Scholar] [CrossRef]
Figure 1. SOR, LYC, and their combination effect on the MDA-MB-231 cell line using an MTT viability test (incubation for 48 h). (A) SOR (IC50 = 21.33 ± 2.25 μM), (B) LYC (IC50 = 169.17 ± 6.94 μM), and (C) SOR-LYC (IC50 = 16.94 ± 2.81 μM). Data are expressed as means ± SD, n = 3.
Figure 1. SOR, LYC, and their combination effect on the MDA-MB-231 cell line using an MTT viability test (incubation for 48 h). (A) SOR (IC50 = 21.33 ± 2.25 μM), (B) LYC (IC50 = 169.17 ± 6.94 μM), and (C) SOR-LYC (IC50 = 16.94 ± 2.81 μM). Data are expressed as means ± SD, n = 3.
Pharmaceuticals 17 00527 g001
Figure 2. Cell cycle analysis of the MDA-MB-231 cell line using flow cytometry at various cell cycle stages (G0/G1, S, and G2/M) (incubation for 48 h) on the IC50 concentration detected via an MTT viability test. (A) Control, (B) SOR (IC50 = 21.33 ± 2.25 μM), (C) LYC (IC50 = 169.17 ± 6.94 μM), and (D) SOR-LYC (IC50 = 16.94 ± 2.81 μM).
Figure 2. Cell cycle analysis of the MDA-MB-231 cell line using flow cytometry at various cell cycle stages (G0/G1, S, and G2/M) (incubation for 48 h) on the IC50 concentration detected via an MTT viability test. (A) Control, (B) SOR (IC50 = 21.33 ± 2.25 μM), (C) LYC (IC50 = 169.17 ± 6.94 μM), and (D) SOR-LYC (IC50 = 16.94 ± 2.81 μM).
Pharmaceuticals 17 00527 g002
Figure 3. Estimation of apoptotic cell populations of the MDA-MB-231 cell line using Annexin-V/PI double staining on the IC50 concentration detected via an MTT viability test. (A) Control, (B) SOR (IC50 = 21.33 ± 2.25 μM), (C) LYC (IC50 = 169.17 ± 6.94 μM), and (D) SOR-LYC (IC50 = 16.94 ± 2.81 μM). The lower right quadrant shows cells in apoptosis.
Figure 3. Estimation of apoptotic cell populations of the MDA-MB-231 cell line using Annexin-V/PI double staining on the IC50 concentration detected via an MTT viability test. (A) Control, (B) SOR (IC50 = 21.33 ± 2.25 μM), (C) LYC (IC50 = 169.17 ± 6.94 μM), and (D) SOR-LYC (IC50 = 16.94 ± 2.81 μM). The lower right quadrant shows cells in apoptosis.
Pharmaceuticals 17 00527 g003
Figure 4. Effect of SOR, LYC, and their combination on the protein level of (A) JNK-1, (B) ERK-1, (C) Beclin-1, (D) P38, and (E) P53 of the MDA-MB-231 cell line using (ELISA) kits on the IC50 concentration detected via an MTT viability test of LYC, SOR, and their combination (IC50 = 21.33 ± 2.25, 169.17 ± 6.94 and 16.94 ± 2.81 μM, respectively, incubation for 48 h). Data are expressed as means ± SD (n = 3). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SOR group, and # means significant versus the LYC group. Each group differed significantly from the others at p ≤ 0.05.
Figure 4. Effect of SOR, LYC, and their combination on the protein level of (A) JNK-1, (B) ERK-1, (C) Beclin-1, (D) P38, and (E) P53 of the MDA-MB-231 cell line using (ELISA) kits on the IC50 concentration detected via an MTT viability test of LYC, SOR, and their combination (IC50 = 21.33 ± 2.25, 169.17 ± 6.94 and 16.94 ± 2.81 μM, respectively, incubation for 48 h). Data are expressed as means ± SD (n = 3). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SOR group, and # means significant versus the LYC group. Each group differed significantly from the others at p ≤ 0.05.
Pharmaceuticals 17 00527 g004
Figure 5. The survival rate of mice in each group described by Kaplan–Meier survival plots. The survival rate of each group was calculated according to the formula: survival rate = number of surviving mice/total number of mice during the experiment (21 days) after 1, 2, and 3 weeks.
Figure 5. The survival rate of mice in each group described by Kaplan–Meier survival plots. The survival rate of each group was calculated according to the formula: survival rate = number of surviving mice/total number of mice during the experiment (21 days) after 1, 2, and 3 weeks.
Pharmaceuticals 17 00527 g005
Figure 6. Effect of SOR, LYC, and their combination on (A) Tumor volume and (B) Tumor weight. Data were expressed as mean ± SD (n = 10). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SEC-Control group, a means significant versus the SEC-SOR group, and b means significant versus the SEC-LYC group. Each group differed significantly from the others at p ≤ 0.05.
Figure 6. Effect of SOR, LYC, and their combination on (A) Tumor volume and (B) Tumor weight. Data were expressed as mean ± SD (n = 10). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SEC-Control group, a means significant versus the SEC-SOR group, and b means significant versus the SEC-LYC group. Each group differed significantly from the others at p ≤ 0.05.
Pharmaceuticals 17 00527 g006
Figure 7. Effect of SOR, LYC, and their combination on (A) TNF-α and (B) VEGF content. Data are expressed as means ± SD (n = 10). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SEC-Control group, a means significant versus the SEC-SOR group, and b means significant versus the SEC-LYC group. Each group differed significantly from the others at p ≤ 0.05.
Figure 7. Effect of SOR, LYC, and their combination on (A) TNF-α and (B) VEGF content. Data are expressed as means ± SD (n = 10). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SEC-Control group, a means significant versus the SEC-SOR group, and b means significant versus the SEC-LYC group. Each group differed significantly from the others at p ≤ 0.05.
Pharmaceuticals 17 00527 g007
Figure 8. Effect of SOR, LYC, and their combination on (A) TNF-α, (B) VEGF, and (C) caspase 3. Data are expressed as means ± SD (n = 3). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SEC-Control group, a means significant versus the SEC-SOR group, and b means significant versus the SEC-LYC group. Each group differed significantly from the others at p ≤ 0.05.
Figure 8. Effect of SOR, LYC, and their combination on (A) TNF-α, (B) VEGF, and (C) caspase 3. Data are expressed as means ± SD (n = 3). Tukey–Kramer multiple comparison tests were used after a one-way analysis of variance (ANOVA) of the data to examine and assess the findings. * means significant versus the SEC-Control group, a means significant versus the SEC-SOR group, and b means significant versus the SEC-LYC group. Each group differed significantly from the others at p ≤ 0.05.
Pharmaceuticals 17 00527 g008
Figure 9. Histopathological examination (200×, scale bar = 100 μm). (a) The SEC-Control group showed a high proliferative activity of neoplastic cells with a high mitotic index (black arrows). (b) The SEC- SOR group showed an increase in the degenerative and necrotic lesions (red arrows) with a decrease in the neoplastic cells (black arrows). (c) The SEC-LYC group showed an increase in the degenerative and necrotic lesions (red arrows) with a decrease in the neoplastic cells (black arrows). (d) The SEC-SOR-LYC group showed a marked perivascular neoplastic cells necrosis (N) (red arrows).
Figure 9. Histopathological examination (200×, scale bar = 100 μm). (a) The SEC-Control group showed a high proliferative activity of neoplastic cells with a high mitotic index (black arrows). (b) The SEC- SOR group showed an increase in the degenerative and necrotic lesions (red arrows) with a decrease in the neoplastic cells (black arrows). (c) The SEC-LYC group showed an increase in the degenerative and necrotic lesions (red arrows) with a decrease in the neoplastic cells (black arrows). (d) The SEC-SOR-LYC group showed a marked perivascular neoplastic cells necrosis (N) (red arrows).
Pharmaceuticals 17 00527 g009
Table 1. Primer sequences *.
Table 1. Primer sequences *.
GenePrimer Sequence (5′–3′)Reference
TNF-αF: CACCAGCTCTGAACAGATCATGA
R: TCAGCCCATCTTCTTCCAGATGGT
[85]
VEGFF: GGCTCTGAAACCATGAACTTTCT
R: GCAGTAGCTGCGCTGGTAGAC
[86]
Caspase 3F: GGAGTCTGACTGGAAAGCCGAA
R: CTTCTGGCAAGCCATCTCCTCA
[87]
Β-ActinF: GTG GGA ATT CGT CAG AAG GAC TCC TAT GTG
R: GAA GTC TAG AGC AAC ATA GCA CAG CTT CTC
[88]
* VEGF: vascular endothelial growth factor, TNF-α: tumor necrosis factor-alpha, Β-Actin: beta actin.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

El-Masry, T.A.; El-Nagar, M.M.F.; El Mahdy, N.A.; Alherz, F.A.; Taher, R.; Osman, E.Y. Potential Antitumor Activity of Combined Lycopene and Sorafenib against Solid Ehrlich Carcinoma via Targeting Autophagy and Apoptosis and Suppressing Proliferation. Pharmaceuticals 2024, 17, 527. https://doi.org/10.3390/ph17040527

AMA Style

El-Masry TA, El-Nagar MMF, El Mahdy NA, Alherz FA, Taher R, Osman EY. Potential Antitumor Activity of Combined Lycopene and Sorafenib against Solid Ehrlich Carcinoma via Targeting Autophagy and Apoptosis and Suppressing Proliferation. Pharmaceuticals. 2024; 17(4):527. https://doi.org/10.3390/ph17040527

Chicago/Turabian Style

El-Masry, Thanaa A., Maysa M. F. El-Nagar, Nageh A. El Mahdy, Fatemah A. Alherz, Reham Taher, and Enass Y. Osman. 2024. "Potential Antitumor Activity of Combined Lycopene and Sorafenib against Solid Ehrlich Carcinoma via Targeting Autophagy and Apoptosis and Suppressing Proliferation" Pharmaceuticals 17, no. 4: 527. https://doi.org/10.3390/ph17040527

APA Style

El-Masry, T. A., El-Nagar, M. M. F., El Mahdy, N. A., Alherz, F. A., Taher, R., & Osman, E. Y. (2024). Potential Antitumor Activity of Combined Lycopene and Sorafenib against Solid Ehrlich Carcinoma via Targeting Autophagy and Apoptosis and Suppressing Proliferation. Pharmaceuticals, 17(4), 527. https://doi.org/10.3390/ph17040527

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop