Interaction of eight steroid derivatives with VEGFR-1 using a theoretical model

Maria López-Ramos1, Lauro Figueroa‑Valverde1, Magdalena Alvarez-Ramirez2, Marcela Rosas‑Nexticapa2, María Virginia Mateu-Armad Maria2 & Regina Cauich-Carrillo3

1 Laboratory of Pharmaco-Chemistry, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Av. Agustín Melgar s/n, Col Buenavista C.P. 24039 Campeche, Camp., México

2 Faculty of Nutrition, University Veracruzana, Médicos y Odontologos s/n C.P. 91010, Unidad del Bosque Xalapa Veracruz, México

3 University Autonomous of Quintana Roo State, Campus Chetumal, Av Erik Paolo Martinez s/n esq. Av. 4 de marzo, Col. Magisteterial, C.P. 77039, México

Correspondence: Figueroa-Valverde Lauro, Pharmacochemistry Laboratory, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Av. Agustín Melgar s/n, Col Buenavista C.P. 24039 Campeche, Camp., México. E-mail: lfiguero@uacam.mx

 

Received: December 14, 2023                     DOI: 10.14295/bjs.v3i3.523

Accepted: February 23, 2024                      URL: https://doi.org/10.14295/bjs.v3i3.523

 

Abstract

Some vascular endothelial growth factor receptor-1 (VEGFR-1) inhibitors drugs have been used to cancer cells; however, their interaction with VEGFR-1 is very confusing. The objective of this research was to evaluate the possible interaction of eight steroid derivatives with VEGFR-1 surface using 3hgn protein, cabozantinib, pazopanib, regorafenib, and sorafenib as theoretical tools in DockingServer program. The results showed some differences in the interaction of the steroid derivatives (1-8) with the 3hng protein surface such as i) differences in the number of amino acids; ii) different position of some amino acids compared to cabozantinib, pazopanib, regorafenib, and sorafenib. Besides, the inhibition constant (Ki) for steroid derivatives 1, 3, 6 and 8 was lower compared to cabozantinib and sorafenib drugs. In addition, other data display that Ki for steroid analogs 1, 3, 4, 6, 7 and 8 was lower compared with pazopanib and regorafenib. In conclusion, all these data suggest that steroid derivatives 1, 3, 4, 6, 7 and 8 could act as VEGFR-1 inhibitors and this phenomenon could be translated as good compounds to treat cancer cells.

Keywords: cancer, steroid, VEGFR-1, docking, theoretical model.

Interação de oito derivados de esteróides com VEGFR-1 utilizando um modelo teórico

Resumo

Alguns medicamentos inibidores do receptor 1 do fator de crescimento endotelial vascular (VEGFR-1), têm sido usados para células cancerígenas, no entanto, a sua interação com o VEGFR-1 é muito confusa. O objetivo desta pesquisa foi avaliar a possível interação de oito derivados de esteroides com a superfície do VEGFR-1 utilizando proteína 3hgn, cabozantinibe, pazopanibe, regorafenibe e sorafenibe como ferramentas teóricas no programa DockingServer. Os resultados mostraram algumas diferenças na interação dos derivados esteroides (1-8) com a superfície da proteína 3hng, tais como i) diferenças no número de aminoácidos; ii) posição diferente de alguns aminoácidos em comparação com cabozantinibe, pazopanibe, regorafenibe e sorafenibe. Além disso, a constante de inibição (Ki) para os derivados esteroides 1, 3, 6 e 8 foi menor em comparação com os medicamentos cabozantinibe e sorafenibe. Além disso, outros dados mostram que o Ki para os análogos de esteroides 1, 3, 4, 6, 7 e 8 foi menor em comparação com o pazopanibe e o regorafenibe. Em conclusão, todos estes dados sugerem que os derivados esteroides 1, 3, 4, 6, 7 e 8 poderiam actuar como inibidores do VEGFR-1 e este fenómeno poderia ser traduzido como bons compostos para tratar células cancerígenas.

Palavras-chave: câncer, esteróide, VEGFR-1, acoplamento, modelo teórico.

 

1. Introduction

The endothelium exerts a wide variety of functions, including the control of vascular function, the blood fluidity, permeability of biomolecules, and others (Vane et al., 1990; Bouis et al., 2001). It is noteworthy that endothelial cells in response to tissue injury or hypoxic conditions can develop new vessels through a differentiation process called angiogenesis (Ge et al., 2021; Lee et al., 2021); this phenomenon can be regulated through several biochemical factors, such as vascular endothelial growth factor and fibroblast growth factor (Mezu-Ndubuisi; Maheshwari, 2021).

Several studies indicate that vascular endothelial growth factor can interact with some endothelial cell surface receptors such as VEGF-R1, VEGF-R2 and VEGF-R3 which to indirectly regulate the formation of new blood vessels under normal conditions (Rahini et al, 2000; Shibuya, 2006). However, some studies suggest that VEGF-R1, VEGFR2 and VEGF-R3 activation can be involved in cancer cell growth (Carmeliet, 2005; Zhao et al., 2022). For example, a study showed that vascular endothelial growth factor can induce the proliferation of lymphatic vessels in patients with primary gastric cancer through VEGFR-3 activation (Yonemur et al 2001).

Other data indicate that VEGFR-2 may be expressed in carcinoid cancer cells, this phenomenon play an important role in tumor growth and metastasis (Silva et al., 2011). Besides, a study showed that VEGFR-2 and VEGFR-3 can be expressed in ovarian cancer patients using Western-blotting methods (Klasa-Mazurkiewicz et al., 2011). Other reports displayed that both VEGFR-1 and VEGFR-2 receptors are expressed in bladder squamous cell carcinoma cell line using an immunoblot analysis (Kopparapu et al., 2013). Besides, a study showed that VEGFR-1 can regulate epidermal growth factor receptor to promote proliferation in colon cancer cells using Western immunoblotting (Nagano et al., 2019).

On the other hand, it is important to mention that some drugs can act as VEGF receptor inhibitors to treat cancer cells; for example, a study showed that pazopanib act as VEGF receptors non-selective inhibitor which has been approved for the treatment of multiple histological subtypes of soft tissue sarcoma (Lee et al., 2019). Other study display that regorafenib (VEGF-R1, -R2, -R3 inhibitor) can confers an overall survival benefit in patients with refractory metastatic colorectal cancer (Bekaii-Saab et al., 2019).

Other data indicate that regorafenib (VEGF receptors non selective inhibitor) has been used to treat Gastric Cancer (Pavlakis et al., 2016); however, regorafenib can induce adaptive resistance of colorectal cancer cells via inhibition of vascular endothelial growth factor receptor (Tamida et al., 2017). Furthermore, a study showed that the administration of sorafenib (VEGF receptors inhibitor) can prolong survival in patients with advanced hepatocellular carcinoma (Campani et al., 2020).

All of these data indicate that several drugs can be used to treat cancer; however, some of these drugs can induce acquired resistance which can increase the risk of death in worldwide due to this clinical pathology (Lo et al., 2015; Mir et al., 2017; Bruix et al., 2017). In the search for a therapeutic alternative to reduce the acquired resistance induced by some drugs, the drug vandetanib was used as VEGFR-1 inhibitor (Bianco et al., 2008), which is a predisposing factor involved in the acquired resistance induced to some anticancer drugs (Mezquita et al., 2016; Atzori et al., 2020).

These data indicate that several drugs have been used for try of cancer cells through inhibiting VEGFR-1; however, its interaction with this biomolecule is not clear. Analyzing all these data the aim of this study was to evaluate the possible interaction of eight steroid-derivatives with VEGF-R1 using 3hng protein (Tresaugues et al., 2013) cabozantinib (Kelley et al., 2022), pazopanib (Shiri et al., 2022), regorafenib (Zhang et al., 2019) and sorafenib (Stăncioiu et al., 2022) as theoretical tools in DockingServer program (Seidel t al., 2017).

 

2. Materials and Methods

2.1 Methodology general

Steroid derivatives (Figure 1) were used to evaluate their possible interaction with VEGF-R1 as follows:

 

Figure 1. Chemical structure of steroid derivatives (1-8). Source: Authors, 2024.

 

2.2 Name International Union of Pure and Applied Chemistry (IUPAC)

1 = 2-hydroxy-methylene-5-cholestan-3-one Barthakur,

2 = 16-dehydropregnenolone acetate (Saikia et al., 2015)

3 = Acetic acid 17-bromo-16-formyl-10,13-dimethyl-2,3,4,7,8, 9,10,11,12,13,14,15-dodecahydro-1H-cyclopenta[a]phenanthren-3-yl ester (Gogoi et al., 2012).

4 = [(10R,14S)-20-methoxy-10,14-dimethyl-16-azahexacyclo [12.11.0.02,11.05,10.015,24.017,22]pentacosa-4,15(24),16,18,20,22-hexaen-7-yl] ace tate (Gogoi et al., 2012).

5 = 17-Chloro-3-methoxy-13-methyl-7,8,9,11,12,13,14,15-octa-hydro-6H-cyclopenta[a]phenanthrene-16-carbaldehyde (Baji et al., 2016).

6 = (1S,2S,11S,14S)-7-methoxy-14-methyl-16-azahexacyclo- [12.11.0.02,11.05,10.015,24.017,22]pentacosa-5(10),6,8,15(24),16,18, 20,22-octaene. (Baji et al., 2016).

7 = 7-{1-[4-(3-Hydroxy-2-methyl-propyl)-3-methyl-isoxazol-5-yl]-ethyl}-4a,6a-dimethyl-icosahydro-pentaleno[2,1-a]phenanthrene-2,8- diol (Hernández-Linares et al., 2011).

8 = 2-Bromo-3-hydroxy-13-methyl-6,7,8,9,11,12,13,14,15,16-decahydro-cyclopenta[a]phenanthren-17-one (Barthakur et al., 2009).

 

2.3 Pharmacophore model

3D pharmacophore model for steroid derivatives (1 to 8) was evaluated using LigandScout 4.08 software (Temml et al., 2014).

 

2.4 Protein-Ligand

The interaction of steroid derivatives (1-8) with 3hng protein (PDB DOI:  https://doi.org/10.2210/pdb3HNG/pdb) was determined using cabozantinib, pazopanib, regorafenib and sorafenib as controls in DockingServer program (Seidel et al., 2017).

 

2.5 Pharmacokinetics parameter

Pharmacokinetic factors for steroid derivatives 1, 3, 4, 6, 7 and 8 were determined using the SwissADME software (Mahanthesh et al., 2020).

 

2.6 Toxicology analysis

Toxicology evaluation for steroid derivatives 1, 3, 4, 6, 7 and 8 were determined using the Gussar software (Lagunin et al., 2011).

 

3. Results

3.1 Pharmacophore model

Figure 2 shows a pharmacophore model for furanone and their derivatives (1 to 31) using the LigandScout 4.0 program. The results displayed different types of hydrogen bond donors (HBD) and hydrogen bond acceptors (HBA) and lipophilic areas.

 


Figure 2. Pharmacophore model for steroid derivatives (1-8). Note: Visualized with LigandScout 4.4 program. HBD = hydrogen bond donors (green), hydrogen bond acceptors (red), halogen bond donor (pink). Source: Authors, 2024.

 

3.2 Ligand-protein complex

Table 1 and Figure 3 shows the different aminoacid residues involved in the interaction of steroid derivatives (1 to 8), cabozantinib, pazopanib, regorafenib and sorafenib with 3hng protein surface. However, only aminoacid residue Ala874 is bound to 3hng protein surface compared with steroid derivatives 2 to 8), cabozantinib, pazopanib, regorafenib and sorafenib.

 

Table 1. Aminoacid residues involved in the interaction of Cabozantinib (I), Pazopanib (II); Regorafenib (III), Sorafenib (IV) and steroid derivatives (1 to 8) with 3hng protein surface.

Compound

Aminoacid residues

Cabozantinib

Val841; Ala859; Lys861; Glu878; Ile881; Leu882; Val892; Val907; Val909; Cys1018; His1020; Leu1029; Ile1038; Cys1039; Asp1040; Phe1041

Pazopanib

Leu833; Glu878; Leu882; Val892; Val909; Tyr911; Cys912; His1020; Leu1029; Cys1039; Asp1040; Phe1041

Regorafenib

Val841; Ala859; Lys861; Glu878; Leu882; Ile885; Ile881; Val892; Val907; Val909; Cys912; Leu1013; Cys1018; Ile1019; His1020; Leu1029; Asp1040; Phe1041

Sorafenib

Glu878; Ile881; Leu882; Val891; Val892; Leu1013; Ile1019; His1020; Arg1021; Ile1038; Cys1039; Asp1040

1

Ala874; Glu878; Ile881, Leu882; Ile885; Val891; Val892; Val909, Leu1013; Cys1018; His1020; Ile1038; Asp1040

2

Leu833; Val841; Ala859; Lys861; Glu878; Leu882; Val892; Val909; Tyr911; Leu1029; Cys1039; Asp1040; Phe1041

3

Val841; Ala859; Lys861, Glu878; Leu882; Ile885; Val892; Leu1013; Cys1018; His1020; Cys1039; Asp1040

4

Leu833; Val841; Lys861; Glu878; Leu882; Val892; Val909; Leu1013; His1020; Leu1029; Cys1039; Asp1040; Phe1041

5

Glu878; Ile881; Leu882; Val891; Val892; Leu1013; Cys1018; His1020; Arg1021; Ile1038; Asp 1040

6

Asp807; Glu878; Ile881; Leu882; Val892; Val909; His1020 ; Arg1021; Cys 039; Asp1040

7

Asp807; Val841; Lys861; Glu878 Ile881; Leu882; Val892; Val909; His 1020; Arg1021; Leu1029; Cys1039;  Asp1040; Phe1041

8

Asp807; Glu878; Ile881; Leu882; Val891; Val892; Cys1018; His1020; Arg1021; Asp1040

Source: Authors, 2024.

 

Other data showed differences in energies levels for steroid derivatives (1 to 8) compared to cabozantinib, pazopanib, regorafenib and sorafenib (Table 2). Besides, inhibition constant (Ki) for 6 was lower compared with steroid derivatives (1-5, 7 and 8), cabozantinib, pazopanib, regorafenib and sorafenib. In addition, the Ki for steroid derivatives 1, 3, 4, 7 and 8 was lower compared to pazopanib, regorafenib and sorafenib.

 

Table 2. Thermodynamic parameters involved in the interaction of for steroid derivatives (1-8), Cabozantinib, Pazopanib, Regorafenib, Sorafenib with 3hng protein surface.

Compound

A

B

C

D

E

F

Cabozantinib

-7.70

2.28

-8.77

-0.18

–8.95

1000.65

Pazopanib

–8.76

380.77

–10.15

–0.11

–10.26

999.38

Regorafenib

–5.05

198.17

–6.84

–0.09

–6.93

1004.77

Sorafenib

–7.03

7.03

–8.19

–0.23

–8.42

922.58

1

-7.67

2.37

-8.77

-0.09

-8.86

895.72

2

-5.88

49.22

-6.76

-0.04

-6.80

779.63

3

-7.46

3.42

-8.33

-0.02

-8.34

818.61

4

-6.95

8.11

-7.73

-0.10

-7.83

1000.54

5

-8.31

815.92

-8.70

-0.20

-8.90

718.19

6

-7.88

1.68

-8.13

-0.05

-8.18

820.06

7

-10.73

13.56

-12.71

-0.11

-12.82

1049.24

8

-7.92

1.56

-8.16

-0.06

-8.22

635.57

Note: A = Est: Free Energy of Binding (kcal/mol); B = Inhibition Constant, Ki (mM); C = vdW + Hbond + desolv Energy (kcal/mol); D = Electrostatic Energy (kcal/mol); E = Total Intermolec. Energy (kcal/mol); F = Interact. Surface. Source: Authors, 2024.  

 


Figure 3. Aminoacid residues involved in the interaction of steroid derivatives (1-8) with 3hng protein surface. Note: Visualized with DockingServer program. Source: Authors, 2024.

 

3.3 Lipophilicity analysis

The results (Table 3) showed that steroid derivative 1 could have a higher degree of Lipophilicity compared to compounds 2-8; however, compound 8 showed lower Lipophilicity compared to steroid derivatives 1-7.

 

Table 3. Lipophilicity degree for steroid derivatives (1 to 8) using several theoretical models.

Compound

ilogP

XlogP3

WlogP

MlogP

Silicost-IT

LogPo/w consensus

1

4.82

9.36

7.33

5.50

6.62

681

2

3.46

4.80

5.01

4.22

4.36

4.31

3

3.40

4.50

5.34

4.33

4.77

4.47

4

4.33

6.02

6.15

4.84

5.74

5.41

5

3.27

4.73

4.85

4.04

5.05

4.39

6

3.94

6.34

5.81

5.09

5.99

5.44

7

4.17

6.64

5.88

4.47

5.37

5.31

8

3.03

3.82

4.58

4.06

4.50

4.00

Source: Authors, 2024.

 

3.4 Pharmacokinetic parameters

Table 4 shows the theoretical pharmacokinetic parameters for the steroid derivatives (1-8). The results showed that the gastrointestinal absorption of compound 1 could be lower compared to 3, 4, 6, 7 and 8. Furthermore, the metabolism of steroid derivatives involves different Cyp´s for each steroid derivative.

 

Table 4. Pharmacokinetic parameters for steroids derivatives (1 to 8) using SwissADME program.

Compound

GI absorption

BBB Permeant

P-gp substrate

Cyp1A2 inhibitor

Cyp 2C19 inhibitor

Cyp2C9 inhibitor

Cyp2D6 inhibitor

Cyp3A4 inhibitor

1

Low

No

No

No

No

Yes

No

No

3

High

Yes

No

No

No

Yes

No

No

4

High

No

No

No

No

No

No

Yes

6

High

No

No

No

No

No

Yes

No

7

High

No

Yes

No

No

No

No

No

8

High

Yes

Yes

Yes

No

No

Yes

No

Note: Cyp = P450 family; GI absorption = gastrointestinal absoption; PPB = plasma protein binding; vd = volume distribution; T1/2 = medium live; CL = clearance; Fu = Fraction unbound un plasms; BBB = barrier blood brain. Source: Authors, 2024.

 

3.5 Toxicology analysis

Table 5 showed that compound 1 requires higher doses through intraperitoneal, intravenous, oral and subcutaneous routes of administration to produce a certain degree of toxicity compared to steroid derivatives (2-8).

 

Table 5. Theoretical toxicity analysis produced by steroid derivatives.

Compound

Rat IP LD50

(mgkg)

Rat IV LD50

(mgkg)

Rat Oral LD50

(mgkg)

Rat SC LD50

(mgkg)

1

1587.00   

70.40   

4094.00   

3477.00   

12

1143.00   

11.88   

2407.00   

1618.00   

14

472.70   

4.91   

1085.00   

307.30   

19

1552.00   

1.16   

1218.00   

1222.00   

22

512.30   

11.85   

11.85    

397.40   

Note: IP - Intraperitoneal route of administration; IV - Intravenous route of administration; Oral - Oral route of administration; SC - Subcutaneous route of administration. Source: Authors, 2024.

 

4. Discussion

In the literature there are several reports on computer-aided drug design (Macalino et al., 2015; Hassan-Baig et al., 2016); These methods are used to predict the biological activity produced by several drugs on some biomolecule; In this way, in this research a theoretical study was carried out to evaluate the activity of steroid derivatives on (VEGFR-1) using some tools such as;

 

4.1 Pharmacophore model

Pharmacophore models are used to define the chemical characteristics of one or more molecules with the same biological activity; in this way pharmacophore is used as a theoretical tool to develop some compounds with therapeutic purposes (Wang et al., 2017). For example, some studies developed a C6-substituted steroid pharmacophore-based strategy to identify new aromatase inhibitors using HipHop pharmacophore model (Neves et al., 2009). Other data showed the pharmacophore for testosterone, estradiol and androstenedione using Discovery Studio 2.0 program (Saxena et al., 2016).

Besides, a report showed the identification of inhibitors of the steroid sulfate transporter using Catalyst-Pharmacophore Model (Grosser et al., 2016). Recently, a pharmacophore for a steroid derivative was developed using the LigandScout program (Figueroa-Valverde et al., 2023). The aim of this study, several pharmacophores were designed for eight steroid derivatives using Ligandscout software. In the Figure 2 are showed different pharmacophore for eight steroid derivatives; it is noteworthy that characteristics of each pharmacophore depends on the functional groups involved in the chemical structure of steroid derivatives, which can be hydrogen bond donors (HBD), hydrogen bond acceptors (HBA), halogen bond donor (XBD), rings, aromatics and hydrophobic areas.

 

4.2 Ligand-protein complex

The interactions of biomolecules (protein-protein and small molecules with macromolecules) are essential to produce different biological activities such as signal transduction, physiological regulation, genetic transcription and enzymatic activity. In the search for some system which can predict protein-protein interactions, several methods have been used such as NGPINT (Banerjee et al., 2021), MEGADOCK (Matsuzaki et al., 2013), ProKSim (Khruschev et al., 2013) and others. Furthermore, to evaluate the interaction of small molecules with macromolecules, other types of methods are used to determine the ligand-protein complex formation. For example, PyPLIF (Radifar et al., 2013), PLIP (Salentin et al., 2015), LIGPLOT (Wallace et al., 1995), Autodock (Forli et al., 2016), DockingServer (Figueroa-Valverde et al., 2023).

In this study, the interaction of steroid derivatives with vascular endothelial growth factor receptor-1 (VEGFR-1) was evaluated using 3hng protein, cabozantinib, pazopanib, regorafenib and sorafenib as theoretical tools in DockingServer program. The results (Table 1, Figure 3) showed differences in the number of amino acid residues involved in the interaction of the steroid derivatives with the surface of the 3hng protein compared to cabozantinib, pazopanib, regorafenib and sorafenib. Furtheremore, the steroid derivative (compound 1) may possibly interact with Ala874 aminoacid residue compared to cabozantinib, pazopanib, regorafenib and sorafenib and steroid derivatives 2 to 8.

This phenomenon could condition the biological activity of steroid derivatives; however, it is important to mention that other type of thermodynamic factors could be involved. For this reason, in this study thermodynamic parameters (Table 2) for steroid derivatives, cabozantinib, pazopanib, regorafenib and sorafenib were evaluated using DockingServer program. The results showed differences in the energies levels for steroid derivatives compared with cabozantinib, pazopanib, regorafenib and sorafenib. Other data showed that inhibition constant (Ki) for 6 was lower compared with steroid derivatives (1-5, 7 and 8), cabozantinib, pazopanib, regorafenib and sorafenib. Besides, the Ki for steroid derivatives 1, 3, 4, 7 and 8 was lower compared to pazopanib, regorafenib and sorafenib. All these data indicate that steroid derivatives 1, 3, 4, 6, 7 and 8 could act as 3hng protein inhibitors, this phenomenon could be translated as vascular endothelial growth factor receptor 1 (VEGFR-1) inhibition which could be involved in some cancer cell growth processes.

 

4.3 Lipophilicity analysis

In the literature there are reports on the determination of lipophilicity degree of several compounds using different methods such as ilogP (Daina et al., 2014), XlogP (Zhong et al., 2018), WlogP (Daina et al., 2016), MlogP (Chui, 2010), Silicost-IT (Shahryari et al., 2021). Furthermore, other studies showed that SwissADME software can be used to determine the lipophilicity degree (LogPo/w consensus) of several drugs. Analyzing these data, in this study, SwissADME program was used to calculate the lipophilicity degree of steroid derivatives [33]. The results (Table 3) display that compound 1 have higher lipophilicity degree compared with other steroid derivatives; this phenomenon could condition some changes in pharmacokinetic process.

 

4.4 Pharmacokinetic parameters

There are several methods such as PK/PD (Derendorf et al., 1999), PKMP (Shah, 2022), PBPK (Kanacher et al., 2020), PkQuest (Levitt et al., 2002) have been used to characterize the effectiveness and safety of medications. In this research, SwissADME (Mahanthesh et al., 2020) was used to determinate some pharmacokinetic parameters for steroid derivatives 1, 3, 4, 6, 7 and 8. The results showed that; i) possibly the absorption of compound 1 could be lower compared to 3, 4, 6, 7 and 8; and ii) steroid derivatives could be metabolized through different Cyp's. This phenomenon could depend on the chemical characteristics of each steroid derivative, which may result in the generation of a beneficial or toxic metabolite.

 

4.5 Toxicology analysis

For several years, several computational tools such as ProTox-II (Banerjee et al., 2018), STopTox (Borba et al., 2022), GUSAR (Lagunin et al., 2011) have been used to predict toxicity degree of new compounds with biological activity. For this reason, in this research, the possible toxicity produced by steroid derivatives (1-8) was determined using the GUSAR software. The results display that compound 1 requires higher doses through intraperitoneal, intravenous, oral and subcutaneous routes of administration to produce a certain degree of toxicity compared to steroid derivatives. These data suggest that toxicity could depends on the following parameters; i) the dose administered; ii) the different routes of administration; and iii) the chemical characteristics of each steroid derivative.

 

5. Conclusions

Theoretical models used in this study are suitable for the following reasons: i) develop a pharmacophore model for steroid derivatives that allows analyzing their interaction with 3hng protein surface; ii) analyze the thermodynamic parameters involved in the interaction of steroidal derivatives with the 3hng protein surface; ii) Analyze both pharmacokinetic and toxicological aspects that can determine the biological activity of each steroid derivative. All these data suggest that steroid derivatives 1, 3, 4, 6, 7 and 8 could be a good alternative as VEGFR-1inhibitors to decrease cancer cells growth.

 

6. Authors’ Contributions

López-Ramos Maria: reading, writing and evaluating data. Figueroa Valverde Lauro: article writing, data evaluation and corrections. Alvarez-Ramirez Magdalena: corrections, software usage, data analysis. Rosas Nexticapa Marcela: corrections, data analysis, writing and layout corrections. Mateu-Armad Maria Virginia: outline of topics, writing, translation and corrections. Cauich-Carrillo Regina: translation, corrections, submission and publication.

 

7. Conflicts of Interest

No conflicts of interest.

 

8. Ethics Approval

Not applicable.

 

9. References

Atzori, M., Ceci, C., Ruffini, F., Trapani, M., Barbaccia, M., Tentori, L., D’Atri, S., Lacal, P. M., Graziani, G. (2020). Role of VEGFR1 in melanoma acquired resistance to the BRAF inhibitor vemurafenib. Journal of Cellular and Molelcular Medicine, 24(1): 465-475. https://doi.org/10.1111/jcmm.14755

Banerjee, P., Eckert, A., Schrey, A., & Preissner, R. (2018). ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Research, 46(W1), W257-W263. https://doi.org/10.1093/nar/gky318

Banerjee, S., Velásquez-Zapata, V., Fuerst, G., Elmore, J., & Wise, R. (2021). NGPINT: a next-generation protein–protein interaction software. Briefings in Bioinformatics, 22(4), 1-14. https://doi.org/10.1093/bib/bbaa351

Bekaii-Saab, T., Ou, F., Ahn, D., Boland, P., Ciombor, K., & Heying, E. (2019). Regorafenib dose-optimisation in patients with refractory metastatic colorectal cancer (ReDOS): a randomised, multicentre, open-label, phase 2 study. The Lancet Oncology, 20(8), 1070-1082. https://doi.org/10.1016/S1470-2045(19)30272-4

Bianco, R., Rosa, R., Damiano, V., Daniele, G., Gelardi, T., & Garofalo, S. (2008). Vascular endothelial growth factor receptor-1 contributes to resistance to anti-epidermal growth factor receptor drugs in human cancer cells. Clinical Cancer Research, 14(16), 5069-5080.

Borba, J., Alves, V., Braga, R., Korn, D., Overdahl, K., Silva, A. (2022). STopTox: An in silico alternative to animal testing for acute systemic and topical toxicity. Environmental Health Perspectives, 130(2), 027012. https://doi.org/10.1289/EHP9341

Bouïs, D., Hospers, G., Meijer, C. (2001). Endothelium in vitro: a review of human vascular endothelial cell lines for blood vessel-related research. Angiogenesis, 4, 91-102. https://doi.org/10.1023/A:1012259529167

Bruix, J., Qin, S., Merle, P., Granito, A., Huang, Y. H., & Bodoky, G. (2016). Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet, 389: 56-66. https://doi.org/10.1016/S0140-6736(16)32453-9

Campani, C., Rimassa, L., Personeni, N., & Marra, F. (2020). Angiogenesis inhibitors for advanced hepatocellular carcinoma: in search for the right partner. Annals of Translational Medicine, 8(22), 1532. https://doi.org/10.21037%2Fatm-20-3788

Carmeliet, P. (2005). VEGF as a key mediator of angiogenesis in cancer. Oncology, 69, 4-10. http://dx.doi.org/10.1159%2F000088478

Chui, C. (2010). The LogP and MLogP models for parallel image processing with multi-core microprocessor. Proceendings of the Sympposium on Information and Communication Technology, 23-27. https://doi.org/10.1145/1852611.1852616

Daina, A., Michielin, O., & Zoete, V. (2014). iLOGP: a simple, robust, and efficient description of n-octanol/water partition coefficient for drug design using the GB/SA approach. Journal of Chemical Information and Modeling, 54(12), 3284-3301. https://doi.org/10.1021/ci500467k

Daina, A., & Zoete, V. (2016). A boiledegg to predict gastrointestinal absorption and brain penetration of small molecules. ChemMedChem, 11(11), 1117-1121. https://doi.org/10.1002/cmdc.201600182

Derendorf, H., & Meibohm, B. (1999). Modeling of pharmacokinetic/pharmacodynamic (PK/PD) relationships: concepts and perspectives. Pharmaceutical Research, 16, 176-185. https://doi.org/10.1023/A:1011907920641

Figueroa-Valverde, L., Rosas-Nexticapa, M., Alvarez-Ramirez, M., López-Ramos, M., Díaz-Cedillo, F., & Mateu-Armad, M. (2023). Evaluation of biological activity exerted by Dibenzo [b, e] Thiophene-11 (6H)-One on left ventricular pressure using an isolated rat heart model. Drug Research, 73(05), 263-270. DOI: 10.1055/a-1995-6351

Figueroa-Valverde, L., Rosas-Nexticapa, M., Montserrat, M., Díaz-Cedillo, F., López-Ramo, M., & Alvarez-Ramirez, M. (2023). Synthesis and theoretical interaction of 3-(2-oxabicyclo [7.4. 0] trideca-1 (13), 9, 11-trien-7-yn-12-yloxy)-steroid deriva-tive with 17β-hydroxysteroid dehydrogenase enzyme surface. Biointerface Research in Applied Chemistry, 13(3), 1-11. https://doi.org/10.33263/BRIAC133.266

Forli, S., Huey, R., Pique, M., Sanner, M., Goodsell, D., & Olson, A. (2016). Computational protein–ligand docking and virtual drug screening with the AutoDock suite. Nature Protocols, 11(5), 905-919. https://doi.org/10.1038/nprot.2016.051

Ge, L., Xun, C., & Li, W. (2021). Extracellular vesicles derived from hypoxia preconditioned olfactory mucosa mesenchymal stem cells enhance angiogenesis via miR-612. Journal of Nanobiotechnology, 19, 1-23. https://doi.org/10.1186/s12951-021-01126-6

Grosser, G., Baringhaus, K., Döring, B., Kramer, W., Petzinger, E., & Geyer, J. (2016). Identification of novel inhibitors of the steroid sulfate carrier ‘sodium-dependent organic anion transporter’SOAT (SLC10A6) by pharmacophore modelling. Molecular and Cellular Endocrinology, 428, 133-141. https://doi.org/10.1016/j.mce.2016.03.028

Hassan-Baig, M., Ahmad, K., Roy, S., Mohammad-Ashraf, J., Adil, M., Haris-Siddiqui, M., & Choi, I. (2016). Computer aided drug design: success and limitations. Current Pharmaceutical Design, 22(5), 572-581.

Kanacher, T., Lindauer, A., Mezzalana, E., Michon, I., Veau, C., & Mantilla, J. (2020). A physiologically-based pharmacokinetic (PBPK) model network for the prediction of CYP1A2 and CYP2C19 drug–drug–gene interactions with fluvoxamine, omeprazole, s-mephenytoin, moclobemide, tizanidine, mexiletine, ethinylestradiol, and caffeine. Pharmaceutics, 12(12), 1191. https://doi.org/10.3390/pharmaceutics12121191

Kelley, R., Rimassa, L., & Cheng, A. (2022). Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312): a multicentre, open-label, randomised, phase 3 trial. The Lancet Oncology, 23, 995-1008. https://doi.org/10.1016/S1470-2045(22)00326-6

Khruschev, S., Abaturova, A., Diakonova, A., Ustinin, D., Zlenko, D., & Fedorov, V. (2013). Multi-particle Brownian dynamics software ProKSim for protein-protein interactions modeling. Computer Research and Modeling, 5(1), 47-64. https://doi.org/10.20537/2076-7633-2013-5-1-47-64

Klasa-Mazurkiewicz, D., Jarząb, M., Milczek, T., Lipińska, B., & Emerich, J. (2011). Clinical significance of VEGFR-2 and VEGFR-3 expression in ovarian cancer patients. Polish Journal of Pathology, 62(1), 31-40.

Kopparapu, P., Boorjian, S., Robinson, B., Downes, M., Gudas, L., & Mongan, N. (2013). Expression of VEGF and its receptors VEGFR1/VEGFR2 is associated with invasiveness of bladder cancer. Anticancer Research, 33(6), 2381-2390.

Lagunin, A., Zakharov, A., Filimonov, D., & Poroikov, V. (2011). QSAR modelling of rat acute toxicity on the basis of PASS prediction. Molecular Informatics, 30(23), 241-250. https://doi.org/10.1002/minf.201000151

Lee, A., Jones, R., & Huang, P. (2019). Pazopanib in advanced soft tissue sarcomas. Signal Transduction and Targeted Therapy, 4(1), 1-9. https://doi.org/10.1038/s41392-019-0049-6

Lee, H., Xu, Y., & He, L. (2021). Role of venous endothelial cells in developmental and pathologic angiogenesis. Circulation, 144, 1308-1322. https://doi.org/10.1161/CIRCULATIONAHA.121.054071

Levitt, D. (2002). PKQuest: capillary permeability limitation and plasma protein binding–application to human inulin, dicloxacillin and ceftriaxone pharmacokinetics. BMC Clinical Pharmacology, 2(1), 1-11. https://doi.org/10.1186/1472-6904-2-7

Lo, J., Lau, E., Ching, R., Cheng, B., Ma, M., Ng, I., & Lee, T. (2015). Nuclear factor kappa B–mediated CD47 upregulation promotes sorafenib resistance and its blockade synergizes the effect of sorafenib in hepatocellular carcinoma in mice. Hepatology, 62(2), 534-545. https://doi.org/10.1002/hep.27859

Macalino, S., Gosu, V., Hong, S., & Choi, S. (2015). Role of computer-aided drug design in modern drug discovery. Archives of Pharmacal Research, 38, 1686-1701. https://doi.org/10.1007/s12272-015-0640-5

Mahanthesh, M., Ranjith, D., Yaligar, R., Jyothi, R., Narappa, G., & Ravi, M. (2020). Swiss ADME prediction of phytochemicals present in Butea monosperma (Lam.) Taub. Journal of Pharmacognosy and Phytochemistry, 9(3), 1799-1809.

Matsuzaki, Y., Uchikoga, N., Ohue, M., Shimoda, T., Sato, T., & Ishida, T. (2013). MEGADOCK 3.0: a high-performance protein-protein interaction prediction software using hybrid parallel computing for petascale supercomputing environments. Source Code for Biology and Medicine, 8(1), 1-8. https://doi.org/10.1186/1751-0473-8-18

Mezquita, B., Pineda, E., Mezquita, J., Mezquita, P., Pau, M., CodonyServat, J. (2016). LoVo colon cancer cells resistant to oxaliplatin overexpress cMET and VEGFR1 and respond to VEGF with dephosphorylation of cMET. Molecular Carcinogenesis, 55(5), 411-419. https://doi.org/10.1002/mc.22289

Mezu-Ndubuisi, O., & Maheshwari, A. (2021). The role of integrins in inflammation and angiogenesis. Pediatric Research, 89, 1619-1626. https://doi.org/10.1038/s41390-020-01177-9

Mir, N., Jayachandran, A., Dhungel, B., Shrestha, R., & Steel, J. (2017). Epithelial-to-mesenchymal transition: A mediator of sorafenib resistance in advanced hepatocellular carcinoma. Current Cancer Drug Targets, 17(8), 698-706. https://doi.org/10.2174/1568009617666170427104356

Nagano, H., Tomida, C., Yamagishi, N., Teshima-Kondo, S. (2019). VEGFR-1 regulates EGF-R to promote proliferation in colon cancer cells. International Journal of Molecular Sciences, 20(22), 5608. https://doi.org/10.3390/ijms20225608

Neves, M., Dinis, T., Colomb, G., De-Melo., M. (2009). An efficient steroid pharmacophore-based strategy to identify new aromatase inhibitors. European Journal of Medicinal Chemistry, 44(10), 4121-4127. https://doi.org/10.1016/j.ejmech.2009.05.003

Pavlakis, N., Sjoquist, K., Martin, A., Tsobanis, E., Yip, S., Kang, Y. K., & Goldstein, D. (2016). Regorafenib for the treatment of advanced gastric cancer (INTEGRATE): a multinational placebo-controlled phase II trial. Journal of Clinical Oncology, 34(23), 2728-2735. https://doi.org/10.1200%2FJCO.2015.65.1901

Radifar, M., Yuniarti, N., & Istyastono, E. (2013). PyPLIF: Python-based protein-ligand interaction fingerprinting. Bioinformation, 9(6), 325-328. https://doi.org/10.6026%2F97320630009325

Rahimi, N., Dayanir, V., & Lashkari, K. (2000). Receptor chimeras indicate that the vascular endothelial growth factor receptor-1 (VEGFR-1) modulates mitogenic activity of VEGFR-2 in endothelial cells. Journal of Biological Chemistry, 275(22), 16986-16992. https://doi.org/10.1074/jbc.M000528200

Salentin, S., Schreiber, S., Haupt, V., Adasme, M., & Schroeder, M. (2015). PLIP: fully automated protein–ligand interaction profiler. Nucleic Acids Research, 43(W1), W443-W447. https://doi.org/10.1093/nar/gkv315

Saxena, A., Devillers, J., Pery, A., Beaudouin, R., Balaramnavar, V., & Ahmed, S. (2014). Modelling the binding affinity of steroids to zebrafish sex hormone-binding globulin. SAR and QSAR in Environmental Research, 25(5), 407-421. https://doi.org/10.1080/1062936X.2014.909197

Seidel, T., Bryant, S., Ibis, G., Poli, G., & Langer, T. (2017). 3D Pharmacophore modeling techniques in computer-aided molecular design using ligandscout. Tutorials in Chemoinformatics, 279-309. https://doi.org/10.1002/9781119161110.ch20

Shah, A. (2022). Pharmacokinetic modeling program (PKMP): A software for PK/PD data analysis. Pharmacokinetics and Pharmacodynamics of Nanoparticulate Drug Delivery Systems, 101-139. https://doi.org/10.1007/978-3-030-83395-4_7

Shahryari, S., Mohammadnejad, P., Noghabi, K. (2021). Screening of anti-Acinetobacter baumannii phytochemicals, based on the potential inhibitory effect on OmpA and OmpW functions. Royal Society Open Science, 8(8), 201652. https://doi.org/10.1098/rsos.201652

Shibuya, M. (2006). Vascular endothelial growth factor receptor-1 (VEGFR-1/Flt-1): a dual regulator for angiogenesis. Angiogenesis, 4, 225-230. https://doi.org/10.1007/s10456-006-9055-8

Shiri, P., Ramezanpour, S., & Amani, A. (2022). A patent review on efficient strategies for the total synthesis of pazopanib, regorafenib and lenvatinib as novel anti-angiogenesis receptor tyrosine kinase inhibitors for cancer therapy. Molecular Diversity, 26, 2981-3002. https://doi.org/10.1007/s11030-022-10406-8

Silva, S., Bowen, K., Rychahou, P., Jackson, L., Weiss, H., & Lee, E. (2011). VEGFR2 expression in carcinoid cancer cells and its role in tumor growth and metastasis. International Journal of Cancer, 128(5), 1045-1056. https://doi.org/10.1002/ijc.25441

Stăncioiu, L., Gherman, A., & Brezeștean, I. (2022). Vibrational spectral analysis of Sorafenib and its molecular docking study compared to other TKIs. Journal of Molecular Structure, 1248, 131507. https://doi.org/10.1016/j.molstruc.2021.131507

Temml, V., Kaserer, T., Kutil, Z., Landa, P., Vanek, T., & Schuster, D. (2014). Pharmacophore modeling for COX-1 and-2 inhibitors with LigandScout in comparison to discovery studio. Future Medicinal Chemistry, 6(17), 1869-1881. https://doi.org/10.4155/fmc.14.114

Tomida, C., Nagano, H., Yamagishi, N., Uchida, T., Ohno, A., Hirasaka, K. (2017). Regorafenib induces adaptive resistance of colorectal cancer cells via inhibition of vascular endothelial growth factor receptor.  Journal of Medical Investigation, 64(3.4), 262-265. https://doi.org/10.2152/jmi.64.262

Tresaugues, L., Roos, A., Arrowsmith, C., Berglund, H., Bountra, C., Collins, R., Nordlund, P. (2009). Crystal structure of VEGFR1 in complex with N-(4-Chlorophenyl)-2-((pyridin-4-ylmethyl) amino) benzamide. 2013; RCSB Protein Data Bank. https://doi.org/10.2210/pdb3HNG/pdb

Vane, J., Änggård, E., & Botting. R. (1990). Regulatory functions of the vascular endothelium. The New England Journal of Medicine, 323, 27-36. DOI: 10.1056/NEJM199007053230106

Wallace, A., Laskowski, R., Thornton, J. (1995). LIGPLOT: a program to generate schematic diagrams of protein-ligand interactions. Protein Engineering, Design & Selection, 8(2), 127-134. https://doi.org/10.1093/protein/8.2.127

Wang, X., Shen, Y., Wang, S., Li, S., Zhang, W., Liu, X., Li, H. (2017). PharmMapper 2017 update: a web server for potential drug target identification with a comprehensive target pharmacophore database. Nucleic Acids Research, 45(W1), W356-W360. https://doi.org/10.1093/nar/gkx374

Yonemura, Y., Fushida, S., Bando, E., Kinoshita, K., Miwa, K., Endo, Y., & Sasaki, T. (2001). Lymphangiogenesis and the vascular endothelial growth factor receptor (VEGFR)-3 in gastric cancer. European Journal of Cancer, 37(7), 918-923. https://doi.org/10.1016/S0959-8049(01)00015-6

Zhang, Y., Wang, Y., & Lei, Z. (2019). Regorafenib antagonizes BCRP-mediated multidrug resistance in colon cancer. Cancer Letters, 442, 104-112. https://doi.org/10.1016/j.canlet.2018.10.032

Zhao, Y., Guo, S., & Deng, J. (2022). VEGF/VEGFR-targeted therapy and immunotherapy in non-small cell lung cancer: targeting the tumor microenvironment. International Journal of Biological Sciences, 18(9), 3845-3858. https://doi.org/10.7150%2Fijbs.70958

Zhong, T., Hao, Y., Yao, X., Zhang, S., Duan, X., Yin, Y., & Zhang, X. (2018). Effect of XlogP and Hansen solubility parameters on small molecule modified paclitaxel anticancer drug conjugates self-assembled into nanoparticles. Bioconjugate Chemistry, 29(2), 437-444. https://doi.org/10.1021/acs.bioconjchem.7b00767

 

Funding

Not applicable.

 

Institutional Review Board Statement

Not applicable.

 

Informed Consent Statement

Not applicable.

 

Copyrights

Copyright for this article is retained by the author(s), with first publication rights granted to the journal.

This is an open-access article distributed under the terms and conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/4.0/).