Research Paper Volume 13, Issue 16 pp 20246—20257

Identification of effective natural PIK3CA H1047R inhibitors by computational study

Naimeng Liu1, , Xinhui Wang2, , Xuan Li3, , Xiaye Lv4, , Haoqun Xie5, , Zhen Guo5, , Jing Wang5, , Gaojing Dou1,5, , Ye Du1, , Dong Song1, ,

  • 1 Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
  • 2 Department of Oncology, First People’s Hospital of Xinxiang, Xinxiang, China
  • 3 Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
  • 4 Department of Hematology, Honghui Hospital, Xi’an Jiao Tong University, Xi’an, China
  • 5 Clinical College, Jilin University, Changchun, China

Received: May 20, 2021       Accepted: August 3, 2021       Published: August 20, 2021      

https://doi.org/10.18632/aging.203409
How to Cite

Copyright: © 2021 Liu et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer with a poor prognosis and a high recurrence rate. PIK3CA gene is frequently mutated in breast cancer, with PIK3CA H1047R as the hotspot mutation reported in TNBC. We used the ZINC database to screen natural compounds that could be structurally modified to develop drugs targeting the PIK3CA H1047R mutant protein in the PI3K pathway. The LibDock module showed that 2,749 compounds could strongly bind to the PIK3CA H1047R protein. Ultimately, the top 20 natural ligands with high LibDock scores were used for further analyses including assessment of ADME (absorption, distribution, metabolism, and excretion), toxicity, stability, and binding affinity. ZINC000004098448 and ZINC000014715656 were selected as the safest drug candidates with strong binding affinity to PIK3CA H1047R, no hepatotoxicity, less carcinogenicity, better plasma protein binding (PPB) properties, and enhanced intestinal permeability and absorption than the two reference drugs, PKI-402 and wortmannin. Moreover, their lower potential energies than those of PIK3CA H1047R confirmed the stability of the ligand-receptor complex under physiological conditions. ZINC000004098448 and ZINC000014715656 are thus safe and stable leads for designing drugs against PIK3CA H1047R as part of a targeted therapeutic approach for patients with TNBC.

Introduction

Triple-negative breast cancer (TNBC) is a highly aggressive molecular subtype of breast cancer, characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). According to the World Health Organization’s latest global cancer data, breast cancer has replaced lung cancer as the most common cancer worldwide. With a high recurrence rate and poor prognosis, TNBC accounts for nearly 10% to 20% of the new breast cancer cases worldwide [13]. Moreover, its 4-year survival rate is only 77%, despite intensive treatment strategies such as surgery and chemotherapy [4]. This highlights the need to develop new and effective treatment strategies.

The phosphoinositide-3 kinase (PI3K) pathway interacts with multiple pathways to perform numerous biological functions including proliferation, apoptosis, and metabolism. It is known to be frequently altered in human cancers, especially in breast cancer [5], with its overexpression associated with the activation of oncogenes such as PIK3CA and MTOR and inactivation of tumor suppressor genes such as PIK3R1 and PTEN [6]. Of these, PIK3CA mutation is the most common and independent event in breast cancer. According to the Cancer Genome Atlas (TCGA) and the Catalogue of Somatic Mutations in Cancer (COSMIC), the mutation rate of the PIK3CA gene is 36% in breast cancer [7]. A study by Jiang et al. reported PIK3CA as the most frequently mutated gene (18%) in refractory TNBC [8], indicating its association with poor outcome and drug resistance [9].

PIK3CA H1047R, the hotspot mutation in TNBC, maps to the kinase domain of the protein [10]. PIK3CA H1047R expression induces dedifferentiation of lineage-restricted epithelial cells into a multipotent stem-like state, which further stimulates the development of heterogeneous mixed-lineage tumors [11]. In addition, certain studies have reported that TNBC with a PIK3CA H1047R mutation is less likely to result in a pathologic complete response (pCR) following chemotherapy [12].

With increasing chemotherapeutic resistance and poor prognosis in patients with TNBC, selective inhibitors of H1047R mutated protein can provide more effective treatment. For example, PKI-402 (ZINC000049745945), a highly specific PI3K inhibitor, is known to hinder breast cancer cell proliferation and block metastasis [13] by suppressing the phosphorylation of PI3K, particularly phosphorylated Akt (p-Akt) at T308 [14]. Wortmannin (ZINC000001619592), another PI3K inhibitor, can kill tumor cells by inducing double-stranded DNA breaks. Computational studies have shown a strong binding affinity of wortmannin with H1047R protein [15, 16]. However, these drugs have certain limitations, for example, PKI-402 is effective only at a high dose and the tumor may reoccur shortly after the treatment [14].

Recent years have witnessed a surge in the use of several natural products as leads that can be structurally modified to develop new and effective drugs [17]. We assessed the inhibitory effect of two natural compounds and their potential for drug development against PIK3CA mutated TNBC using computational methods. In addition, we compared the absorption, distribution, metabolism, excretion (ADME) and toxicity of these compounds with those of PKI-402 and wortmannin using predictive analysis.

Results

Virtual screening of natural product database against PIK3CA H1047R

A total of 17,931 ligands were downloaded from the ZINC database. The chemical structure of PIK3CA H1047R (3HHM) was selected as the receptor to align these ligands. Of these, 2,794 compounds exhibited strong binding to PIK3CA H1047R protein. The top 20 ligands are listed in Table 1. PKI-402 (ZINC000049745945) and wortmannin (ZINC000001619592) were selected as two reference proteins (Figure 1).

Table 1. The top 20 ranked compounds with LibDock scores.

NumberCompoundsLIBDOCK score
1ZINC000014780951132.446
2ZINC000014763060131.304
3ZINC000100168592119.583
4ZINC000004098466119.396
5ZINC000085808802118.322
6ZINC000000899675118.184
7ZINC000004098004117.968
8ZINC000085808820117.475
9ZINC000014715656117.471
10ZINC000013378578117.305
11ZINC000001714287117.107
12ZINC000044417879116.723
13ZINC000006094124114.710
14ZINC000004098448114.209
15ZINC000001667453114.109
16ZINC000014819753112.782
17ZINC000014820552112.700
18ZINC000008234257112.644
19ZINC000000388657112.412
20ZINC000032840897112.136
Molecular structure of PIK3CA H1047R with the highlighted 1047Arg. (A) The initial molecular structure. (B) Structure after binding area surface was added. Blue represents positive charge and red represents negative charge. (C) The molecular structure of PIK3CA H1047R with wortmannin. (D) The molecular structure of PIK3CA H1047R with PKI-402.

Figure 1. Molecular structure of PIK3CA H1047R with the highlighted 1047Arg. (A) The initial molecular structure. (B) Structure after binding area surface was added. Blue represents positive charge and red represents negative charge. (C) The molecular structure of PIK3CA H1047R with wortmannin. (D) The molecular structure of PIK3CA H1047R with PKI-402.

Absorption, distribution, metabolism, and excretion and toxicity analyses

We used the ADME module of the Discovery Studio 4.5 software to study the pharmacologic properties of the top 20 selected compounds, PKI-402, and wortmannin (Table 2). Except for ZINC000044417879, the aqueous solubility of all compounds was predicted to be high. Half of the top 20 compounds had undefined blood–brain barrier (BBB) permeability, whereas others had medium or low permeability, except for ZINC000001714287 and ZINC000004098004. Nearly all compounds including the two targeted drugs effectively inhibited cytochrome P450 2D6 (CYP2D6), except for ZINC000032840897 and ZINC000004098448. Eleven compounds and PKI-402 did not exhibit hepatotoxicity, whereas 9 compounds and wortmannin showed an opposite effect. We next predicted the extent of intestinal permeability and drug absorption of these compounds. ZINC000044417879 and ZINC000008234257 showed the worst and poor intestinal permeability, respectively, and others exhibited a good level of permeability and absorption, except for PKI-402, ZINC000100168592, ZINC000004098466, and ZINC000032840897. An analysis of plasma protein-binding (PPB) properties predicted that 6 compounds had weak binding, similar to that of wortmannin and PKI-402.

Table 2. Adsorption, distribution, metabolism, and excretion properties of compounds.

NumberCompoundsSolubility levelBBB levelCYP2D6HepatotoxicityAbsorption levelPPB level
1ZINC000100168592341110
2ZINC000014819753221000
3ZINC000006094124331000
4ZINC000004098004211000
5ZINC000004098466241110
6ZINC000014820552221000
7ZINC000014780951341101
8ZINC000008234257341121
9ZINC000004098448220000
10ZINC000013378578341001
11ZINC000000388657321000
12ZINC000001667453341100
13ZINC000044417879141130
14ZINC000032840897340111
15ZINC000001714287211000
16ZINC000014763060241000
17ZINC000014715656221000
18ZINC000000899675341000
19ZINC000085808820431101
20ZINC000085808802431101
21wortmannin231101
22PKI-402341011
Abbreviations: BBB: blood-brain barrier; CYP2D6: cytochrome P-450 2D6; PPB: plasma protein binding.
Aqueous-solubility level: 0, extremely low; 1, very low, but possible; 2, low; 3, good.
BBB level: 0, very high penetrant; 1, high; 2, medium; 3, low; 4, undefined.
CYP2D6 level: 0, noninhibitor; 1, inhibitor.
Hepatotoxicity: 0, nontoxic; 1, toxic.
Human-intestinal absorption level: 0, good; 1, moderate; 2, poor; 3, very poor.
PPB: 0, absorbent weak; 1, absorbent strong.

We next screened the drugs for safety using the TOPKAT module based on the two-dimensional (2D) structures of 20 compounds and two reference drugs. The toxic effects of these compounds on humans and the environment were assessed using Ames mutagenicity test (Ames test), the developmental toxicity potential (DTP) test, and the U.S. National Toxicology Program (NTP) database rodent carcinogenicity test (Table 3). Twelve compounds were found to be non-mutagenic; in addition, 10 compounds in male mice, 14 compounds in female rats, and 9 compounds in male rats were non-carcinogenic. On the contrary, both PKI-402 and wortmannin were found to be carcinogenic in these three groups of animals.

Table 3. Toxic effects of compounds.

NumberCompoundsMouse NTPRat NTPAmesDTP
FemaleMaleFemaleMale
1ZINC00010016859200.9990010
2ZINC000014819753110.9980.92901
3ZINC0000060941240010.06800.881
4ZINC0000040980040100.12700.998
5ZINC0000040984660.0911111
6ZINC0000148205520010.6300.997
7ZINC0000147809510.98210100.016
8ZINC00000823425710.70500.37301
9ZINC00000409844800000.7531
10ZINC0000133785780100.9740.0460
11ZINC0000003886570.67700.0170.0090.9890.986
12ZINC0000016674530.86800.9420.9920.7061
13ZINC0000444178790.00100001
14ZINC00003284089700.045000.4350
15ZINC0000017142870.9270.4051001
16ZINC0000147630600.999100.99401
17ZINC00001471565600.0950011
18ZINC00000089967501010.4441
19ZINC00008580882000.0200.94600
20ZINC00008580880200.0200.94600
21wortmannin0.05411100
22PKI-402010.9880.9020.0440
Abbreviations: NTP: U.S. National Toxicology Program; DTP: developmental toxicity potential. NTP < 0.3 (noncarcinogen); > 0.8 (carcinogen). Ames < 0.3 (non-mutagen); > 0.8 (mutagen). DTP < 0.3 (nontoxic); > 0.8 (toxic).

These results showed ZINC000004098448 and ZINC000014715656 as potential candidate drugs with no hepatotoxicity, less carcinogenicity, better PPB properties, and enhanced intestinal permeability and absorption than other compounds and two reference drugs. Thus, ZINC000004098448 and ZINC000014715656 were selected as safe lead compounds for further studies (Figure 2).

Chemical structure of (A) ZINC000014715656 (B) ZINC000004098448 (C) PKI-402 (D) Wortmannin.

Figure 2. Chemical structure of (A) ZINC000014715656 (B) ZINC000004098448 (C) PKI-402 (D) Wortmannin.

Analysis of ligand binding and ligand pharmacophore

The pharmacophore study of the two candidate ligands, PKI-402, and wortmannin showed 14 features in ZINC000014715656 including 4 hydrogen bond (HB) acceptors, 6 hydrophobics, and 4 ring aromatics (Figure 3). ZINC000004098448 showed 20 features in ZINC000004098448, including 6 HB acceptors, 4 HB donors, 4 hydrophobics, and 6 ring aromatics. Wortmannin and PKI-402 showed 13 and 24 features, respectively.

Pharmacophores of (A) ZINC000014715656 (B) ZINC000004098448 (C) PKI-402 (D) Wortmannin.

Figure 3. Pharmacophores of (A) ZINC000014715656 (B) ZINC000004098448 (C) PKI-402 (D) Wortmannin.

We used the CHARMM force field-based CDOCKER method to study the binding affinity and mechanism between these compounds and 3HHM. ZINC000004098448 and ZINC000014715656 bound to 3HHM in the CDOCKER module served as the reference drugs; their potential binding energies are listed in Table 4. The CDOCKER potential energy of ZINC000014715656 (–43.6257 kcal/mol) was close to that of wortmannin (–41.5097 kcal/mol) and PKI-402 (–42.9796 kcal/mol). In addition, the CDOCKER potential energy of ZINC000004098448 (–51.3249 kcal/mol) was highly lower than those of reference drugs, showing the strong binding of 3HHM with ZINC000004098448 and ZINC000014715656 than PKI-402 and wortmannin.

Table 4. CDOCKER potential energy of compounds with PIK3CA H1047R.

Compounds-CDOCKER potential energy (kcal/mol)
ZINC00001471565643.6257
ZINC00000409844851.3249
wortmannin41.5097
PIK-40242.9796

Next, we conducted structural computational studies to obtain hydrogen bonds, π-related interactions, and the charge on these ligands (Figures 4, 5 and 6). ZINC000014715656 formed 4 pairs of hydrogen bonds (Table 5) between O10 of the compound and LYS802:HZ2 of 3HHM, H26 of the compound and SER774:OG of 3HHM, H36 of the compound and ASP810:OD1 of 3HHM, and H36 of the compound and GLU849:O of 3HHM. Eight pairs of π-related interactions are listed in Table 6. In addition, ZINC000004098448 formed 4 pairs of hydrogen bonds between LYS802:HZ2 of 3HHM and O16 of the ligand, VAL851:HN of 3HHM and O25 of the ligand, H38 of the ligand and ASP933:OD1 of 3HHM, and H41 of the ligand and GLU849:O of 3HHM. Six pairs of π-related interactions, including TYR836, ILE932, ILE848, ILE932, MET922, and ILE932 of 3HHM were found with ZINC000004098448. Wortmannin exhibited 6 hydrogen bonds with 3HHM and 4 π-related interactions with 3HHM, whereas PKI-402 displayed 1 hydrogen bond with 3HHM and 4 π-related interactions with 3HHM (Tables 5 and 6).

Schematic representation showing intermolecular interactions of predicted binding modes of (A) ZINC000014715656 with PIK3CA H1047R, (B) ZINC000004098448 with PIK3CA H1047R, (C) PKI-402 with PIK3CA H1047R, and (D) wortmannin with PIK3CA H1047R.

Figure 4. Schematic representation showing intermolecular interactions of predicted binding modes of (A) ZINC000014715656 with PIK3CA H1047R, (B) ZINC000004098448 with PIK3CA H1047R, (C) PKI-402 with PIK3CA H1047R, and (D) wortmannin with PIK3CA H1047R.

Schematic representation showing interactions between ligands and 3HHM. The binding area surface was added. Blue represents positive charge and red represents negative charge. Ligands are shown in sticks, with the structure around the ligand–receptor junction shown in thinner sticks. (A) ZINC000004098448–3HHM complex. (B) ZINC000014715656–3HHM complex.

Figure 5. Schematic representation showing interactions between ligands and 3HHM. The binding area surface was added. Blue represents positive charge and red represents negative charge. Ligands are shown in sticks, with the structure around the ligand–receptor junction shown in thinner sticks. (A) ZINC000004098448–3HHM complex. (B) ZINC000014715656–3HHM complex.

(A) The charge between the ZINC000014715656–3HHM surface. (B) The charge between the ZINC000004098448–3HHM surface. (C) Potential energy of ZINC000004098448 and ZINC000014715656, with average root-mean-square deviation. (D) RMSD of ZINC000004098448 and ZINC000014715656.

Figure 6. (A) The charge between the ZINC000014715656–3HHM surface. (B) The charge between the ZINC000004098448–3HHM surface. (C) Potential energy of ZINC000004098448 and ZINC000014715656, with average root-mean-square deviation. (D) RMSD of ZINC000004098448 and ZINC000014715656.

Table 5. Hydrogen bond interaction parameters of each compound with PIK3CA H1047R.

ReceptorCompoundDonor atomReceptor atomDistances (Å)
3HHMZINC000014715656LYS802:HZ2ZINC000014715656:O101.94
ZINC000014715656:H26SER774:OG3.05
ZINC000014715656:H32ASP810:OD13.03
ZINC000014715656:H36GLU849:O2.08
ZINC000004098448LYS802:HZ2ZINC000004098448:O161.86
VAL851:HNZINC000004098448:O252.12
ZINC000004098448:H38ASP933:OD12.11
ZINC000004098448:H41GLU849:O2.84
wortmanninLYS802:HZ1Molecule:O22.15
ASP933:HNMolecule:O312.38
SER774:HB2Molecule:O72.77
VAL850:HAMolecule:O232.29
ASP933:HAMolecule:O313.07
Molecule:H34SER774:OG2.54
PKI-402Molecule:H60ASP810:OD12.69

Table 6. π-Related interaction parameters of each compound with PIK3CA H1047R.

ReceptorCompoundDonor atomReceptor atomDistances (Å)
3HHMZINC000014715656VAL850ZINC0000147156565.10
VAL851ZINC0000147156565.45
ZINC000014715656MET9224.85
ZINC000014715656ILE9325.27
ZINC000014715656MET7725.30
ZINC000014715656ILE9325.14
ZINC000014715656ILE8484.86
ZINC000014715656ILE9324.25
ZINC000004098448TYR836ZINC0000040984485.83
ILE932ZINC0000040984485.30
ZINC000004098448ILE8484.78
ZINC000004098448ILE9325.02
ZINC000004098448MET9225.33
ZINC000004098448ILE9324.63
wortmanninILE932Molecule4.93
Molecule:C14ILE9324.36
Molecule:C25MET9224.42
Molecule:C25ILE9323.81
PKI-402ARG770Molecule5.37
ILE848Molecule4.37
ILE932Molecule5.22
MoleculeILE9325.23

Molecular dynamics simulation

We next used a molecular dynamics simulation module to study the stability of ligand–3HHM complexes under natural circumstances. Molecular docking experiments were conducted to evaluate the potential binding energies of these complexes and obtain the root mean square deviation (RMSD) curves (Figure 6). The trajectories of the two complexes reached the equilibrium after 30 ps and gradually stabilized with time. The hydrogen bonding and π-related interactions between the compounds and 3HHM contributed to the stability of these complexes. In conclusion, ZINC000004098448 and ZINC000014715656 could stably bind with 3HHM under natural circumstances.

Discussion

Triple-negative breast cancer is the most aggressive and highly invasive molecular subtype of breast cancer with a high recurrence rate and low median survival [4, 18]. The PI3K/AKT pathway is one of the most frequently altered pathways in TNBC [19]. The hyperactivation of the PI3K/AKT pathway due to mutations in PIK3CA gene results in cell cycle dysregulation and consequently tumorigenesis [2022]. Among these, PIK3CA H1047R is a hotspot mutation that maps to the kinase domain of the protein [10].

Targeted drug therapy along with radiotherapy has emerged as a recent therapeutic approach to treat different subtypes of breast cancer. Although numerous drugs are available against TNBC, only a few, such as PKI-402 and wortmannin, are known to target PIK3CA H1047R. In addition, shortcomings such as the high dose requirement of PKI-402 and recurrence within a short interval, limit their clinical applications [14].

We used different modules of the Discovery Studio 4.5 software to screen potential natural ligands for drug development. After 17,931 natural ligands were downloaded from the ZINC 15 database, the LibDock module showed that 2,749 compounds could strongly bind with PIK3CA H1047R. Finally, the top 20 natural ligands with high LibDock scores were used for further analyses.

The ADME and TOPKAT modules were used to predict and evaluate the pharmacologic properties of these 20 compounds. Two natural ligands, namely ZINC000004098448 and ZINC000014715656, were found to be non-hepatotoxic, water-soluble, and fulfilling the ADME criteria. The TOPKAT results showed that these two compounds had less carcinogenicity and could safely replace PKI-402 and wortmannin as targeted drugs.

The CDOCKER module, used to analyze the binding affinity and mechanism of ligands with 3HHM, revealed that ZINC000004098448 had lower potential energy than PKI-402 and wortmannin, whereas the potential energy of ZINC000014715656 was similar to those of reference drugs. Structural computational studies showed the formation of hydrogen bonds and π-related interactions by lead compounds and reference drugs. The results indicated that ZINC000004098448 and ZINC000014715656 had a higher binding affinity for 3HHM than PKI-402 and wortmannin.

Finally, a molecular dynamics simulation model was used to calculate RMSD and potential energy and assess the stability of compound–3HHM complexes. The trajectories of both ZINC000004098448–3HHM and ZINC000014715656–3HHM complexes reached the equilibrium after 30 ps and gradually stabilized with time, implying the stability of two complexes under physiological conditions. These findings indicated that ZINC000004098448 and ZINC000014715656 could be used to develop targeted therapy for patients with TNBC harboring PIK3CA H1047R mutation.

Conclusions

Compared to standard chemotherapy and surgery, molecular targeted therapies are more specific with lesser side effects. We conducted a comprehensive computational study to identify potential effective inhibitors of PIK3CA H1047R. Altogether, our results showed ZINC000004098448 and ZINC000014715656 as promising candidates to develop targeted therapy against PIK3CA mutated TNBC. However, further in vivo and in vitro studies are warranted before these drug candidates could enter the market for clinical applications.

Materials and Methods

Discovery studio 4.5 software and ligand database

Discovery Studio 4.5 software is used to simulate the systems of small molecules and macromolecules. It provides easy-to-use and valuable tools for protein simulation, optimization, and drug design. Discovery Studio is a visualization tool for viewing and analyzing protein and modeling data by integrating the storage and management of experimental data using modeling and simulation tools.

The database of natural products was downloaded from the ZINC database and lead compounds were screened. The ZINC database is a freely available toolset designed by the Irwin and Shoichet Laboratories, Department of Pharmaceutical Chemistry, University of California, San Francisco that provides access to commercially available compounds for virtual screening and ligand and pharmacophore identification [23].

Structure-based virtual screening using LibDock

We used LibDock to screen ligands with potential inhibitory activity against PIK3CA H1047R. The crystals of p110alpha H1047R mutant in complex with wortmannin (Protein Data Bank identifier: 3HHM), wortmannin inhibitor (Protein Data Bank identifier: ZINC000001619592), and PKI-402 inhibitor (Protein Data Bank identifier: ZINC000049745945) were downloaded from the ZINC database and the RCSB Protein Data Bank. The chemical structure of 3HHM is shown in Figure 2. Next, these protein complexes were imported to LibDock. For LibDock analysis, the proteins were prepared by removing the crystal water and other heteroatoms, followed by the addition of hydrogen and protonation, ionization, and energy minimization. The binding region of wortmannin to PIK3CA H1047R was selected as the binding site. The active site for docking was generated, and the LibDock scores of these compounds were ranked and listed [24].

Absorption, distribution, metabolism, and excretion and toxicity prediction

We used the ADME and TOPKAT modules to calculate the absorption, distribution, metabolism, and excretion (ADME) and the toxicity of these compounds. In addition, the safety and pharmacologic properties of these compounds were studied during the selection of natural ligands for PIK3CA H1047R [25].

CDOCKER for molecule docking and ligand pharmacophore prediction

Molecular docking was performed using the CDOCKER module, which is based on CHARMM force field. The compounds were prepared for docking by removing water molecules and adding hydrogen atoms to 3HHM protein because fixed water molecules can affect the conformation of the receptor–ligand complex [26].

Pharmacophore modeling was performed using feature mapping with predictive activity. This method uses a series of compounds with well-defined activity values for specific biological targets. Feature mapping can analyze five kinds of molecular patterns, including hydrophobic, hydrogen bond (HB) acceptor, HB donor, ring aromatic, and positive ion.

Molecular dynamics simulation

The molecule docking program was used to select the best conformation of ligand–3HHM complexes. The complexes were imported to molecular dynamics simulation program. Sodium chloride was added to the system to simulate the physiological environment. The CHARMM field force was used for energy minimization. Next, the trajectory was determined for potential energy and structural characteristics of compounds using Discovery Studio 4.5.

Abbreviations

TNBC: Triple-negative breast cancer; ADME: Absorption distribution metabolic excretion; ER: Estrogen receptor; PR: Progesterone receptor; HER2: Human epidermal growth factor receptor 2; PIK3: Phosphoinositide-3 kinase; TCGA: The Cancer Genome Atlas; COSMIC: The Catalogue of Somatic Mutations in Cancer; pCR: Pathologic complete response; BBB level: Blood brain barrier level; CYP2D6: Cytochrome P450 2D6 inhibition; PPB level: Plasma protein binding properties level; DTP: Developmental toxicity potential; NTP: National Toxicology Program dataset.

Author Contributions

Naimeng Liu contributed to manuscript writing, downloaded the datasets, and conducted bioinformatic analyses. Xuan Li and Ye Du revised the manuscript and figures according to reviewers’ comments. Haoqun Xie, Zhen Guo, and Xinhui Wang analyzed the results. Gaojing Dou, Jing Wang, and Xiaye Lv prepared the figures and tables. Dong Song supervised the study and contributed to the experiments and data analysis.

Conflicts of Interest

The authors declare no conflicts of interest related to this study.

Funding

This study was supported by the Science and technology Development Project of Jilin Province (Grant No.20200403083SF).

References

  • 1. Jiang YZ, Ma D, Suo C, Shi J, Xue M, Hu X, Xiao Y, Yu KD, Liu YR, Yu Y, Zheng Y, Li X, Zhang C, et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell. 2019; 35:428–40.e5. https://doi.org/10.1016/j.ccell.2019.02.001 [PubMed]
  • 2. Shen M, Jiang YZ, Wei Y, Ell B, Sheng X, Esposito M, Kang J, Hang X, Zheng H, Rowicki M, Zhang L, Shih WJ, Celià-Terrassa T, et al. Tinagl1 Suppresses Triple-Negative Breast Cancer Progression and Metastasis by Simultaneously Inhibiting Integrin/FAK and EGFR Signaling. Cancer Cell. 2019; 35:64–80.e7. https://doi.org/10.1016/j.ccell.2018.11.016 [PubMed]
  • 3. Vagia E, Mahalingam D, Cristofanilli M. The Landscape of Targeted Therapies in TNBC. Cancers (Basel). 2020; 12:916. https://doi.org/10.3390/cancers12040916 [PubMed]
  • 4. Howlader N, Cronin KA, Kurian AW, Andridge R. Differences in Breast Cancer Survival by Molecular Subtypes in the United States. Cancer Epidemiol Biomarkers Prev. 2018; 27:619–26. https://doi.org/10.1158/1055-9965.EPI-17-0627 [PubMed]
  • 5. Martínez-Sáez O, Chic N, Pascual T, Adamo B, Vidal M, González-Farré B, Sanfeliu E, Schettini F, Conte B, Brasó-Maristany F, Rodríguez A, Martínez D, Galván P, et al. Frequency and spectrum of PIK3CA somatic mutations in breast cancer. Breast Cancer Res. 2020; 22:45. https://doi.org/10.1186/s13058-020-01284-9 [PubMed]
  • 6. Yang SX, Polley E, Lipkowitz S. New insights on PI3K/AKT pathway alterations and clinical outcomes in breast cancer. Cancer Treat Rev. 2016; 45:87–96. https://doi.org/10.1016/j.ctrv.2016.03.004 [PubMed]
  • 7. Pascual J, Turner NC. Targeting the PI3-kinase pathway in triple-negative breast cancer. Ann Oncol. 2019; 30:1051–60. https://doi.org/10.1093/annonc/mdz133 [PubMed]
  • 8. Jiang YZ, Liu Y, Xiao Y, Hu X, Jiang L, Zuo WJ, Ma D, Ding J, Zhu X, Zou J, Verschraegen C, Stover DG, Kaklamani V, et al. Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: the FUTURE trial. Cell Res. 2021; 31:178–86. https://doi.org/10.1038/s41422-020-0375-9 [PubMed]
  • 9. Xing Y, Lin NU, Maurer MA, Chen H, Mahvash A, Sahin A, Akcakanat A, Li Y, Abramson V, Litton J, Chavez-MacGregor M, Valero V, Piha-Paul SA, et al. Phase II trial of AKT inhibitor MK-2206 in patients with advanced breast cancer who have tumors with PIK3CA or AKT mutations, and/or PTEN loss/PTEN mutation. Breast Cancer Res. 2019; 21:78. https://doi.org/10.1186/s13058-019-1154-8 [PubMed]
  • 10. Tzanikou E, Markou A, Politaki E, Koutsopoulos A, Psyrri A, Mavroudis D, Georgoulias V, Lianidou E. PIK3CA hotspot mutations in circulating tumor cells and paired circulating tumor DNA in breast cancer: a direct comparison study. Mol Oncol. 2019; 13:2515–30. https://doi.org/10.1002/1878-0261.12540 [PubMed]
  • 11. Koren S, Reavie L, Couto JP, De Silva D, Stadler MB, Roloff T, Britschgi A, Eichlisberger T, Kohler H, Aina O, Cardiff RD, Bentires-Alj M. PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature. 2015; 525:114–18. https://doi.org/10.1038/nature14669 [PubMed]
  • 12. Guo S, Loibl S, von Minckwitz G, Darb-Esfahani S, Lederer B, Denkert C. PIK3CA H1047R Mutation Associated with a Lower Pathological Complete Response Rate in Triple-Negative Breast Cancer Patients Treated with Anthracycline-Taxane-Based Neoadjuvant Chemotherapy. Cancer Res Treat. 2020; 52:689–96. https://doi.org/10.4143/crt.2019.497 [PubMed]
  • 13. Yuan G, Lian Z, Liu Q, Lin X, Xie D, Song F, Wang X, Shao S, Zhou B, Li C, Li M, Yao G. Phosphatidyl inositol 3-kinase (PI3K)-mTOR inhibitor PKI-402 inhibits breast cancer induced osteolysis. Cancer Lett. 2019; 443:135–44. https://doi.org/10.1016/j.canlet.2018.11.038 [PubMed]
  • 14. Mallon R, Hollander I, Feldberg L, Lucas J, Soloveva V, Venkatesan A, Dehnhardt C, Delos Santos E, Chen Z, Dos Santos O, Ayral-Kaloustian S, Gibbons J. Antitumor efficacy profile of PKI-402, a dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor. Mol Cancer Ther. 2010; 9:976–84. https://doi.org/10.1158/1535-7163.MCT-09-0954 [PubMed]
  • 15. Ihara M, Shichijo K, Takeshita S, Kudo T. Wortmannin, a specific inhibitor of phosphatidylinositol-3-kinase, induces accumulation of DNA double-strand breaks. J Radiat Res. 2020; 61:171–76. https://doi.org/10.1093/jrr/rrz102 [PubMed]
  • 16. Kumar DT, Doss CG. Investigating the Inhibitory Effect of Wortmannin in the Hotspot Mutation at Codon 1047 of PIK3CA Kinase Domain: A Molecular Docking and Molecular Dynamics Approach. Adv Protein Chem Struct Biol. 2016; 102:267–97. https://doi.org/10.1016/bs.apcsb.2015.09.008 [PubMed]
  • 17. Mondal S, Bandyopadhyay S, Ghosh MK, Mukhopadhyay S, Roy S, Mandal C. Natural products: promising resources for cancer drug discovery. Anticancer Agents Med Chem. 2012; 12:49–75. https://doi.org/10.2174/187152012798764697 [PubMed]
  • 18. Lopez-Ozuna VM, Hachim IY, Hachim MY, Lebrun JJ, Ali S. Prolactin modulates TNBC aggressive phenotype limiting tumorigenesis. Endocr Relat Cancer. 2019; 26:321–37. https://doi.org/10.1530/ERC-18-0523 [PubMed]
  • 19. Tokunaga E, Oki E, Egashira A, Sadanaga N, Morita M, Kakeji Y, Maehara Y. Deregulation of the Akt pathway in human cancer. Curr Cancer Drug Targets. 2008; 8:27–36. https://doi.org/10.2174/156800908783497140 [PubMed]
  • 20. Jouali F, Marchoudi N, Talbi S, Bilal B, El Khasmi M, Rhaissi H, Fekkak J. Detection of PIK3/AKT pathway in Moroccan population with triple negative breast cancer. BMC Cancer. 2018; 18:900. https://doi.org/10.1186/s12885-018-4811-x [PubMed]
  • 21. Vanhaesebroeck B, Guillermet-Guibert J, Graupera M, Bilanges B. The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev Mol Cell Biol. 2010; 11:329–41. https://doi.org/10.1038/nrm2882 [PubMed]
  • 22. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ, Willson JK, Markowitz S, Kinzler KW, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004; 304:554. https://doi.org/10.1126/science.1096502 [PubMed]
  • 23. Ren J, Huangfu Y, Ge J, Wu B, Li W, Wang X, Zhao L. Computational study on natural compounds inhibitor of c-Myc. Medicine (Baltimore). 2020; 99:e23342. https://doi.org/10.1097/md.0000000000023342 [PubMed]
  • 24. Zhong S, Wu B, Yang W, Ge J, Zhang X, Chen Z, Duan H, He Z, Liu Y, Wang H, Jiang Y, Zhang Z, Wang X, et al. Effective natural inhibitors targeting poly ADP-ribose polymerase by computational study. Aging (Albany NY). 2021; 13:1898–912. https://doi.org/10.18632/aging.103986 [PubMed]
  • 25. Ge J, Wang Z, Cheng Y, Ren J, Wu B, Li W, Wang X, Su X, Liu Z. Computational study of novel natural inhibitors targeting aminopeptidase N(CD13). Aging (Albany NY). 2020; 12:8523–35. https://doi.org/10.18632/aging.103155 [PubMed]
  • 26. Zhong S, Bai Y, Wu B, Ge J, Jiang S, Li W, Wang X, Ren J, Xu H, Chen Y, Zhao G. Selected by gene co-expression network and molecular docking analyses, ENMD-2076 is highly effective in glioblastoma-bearing rats. Aging (Albany NY). 2019; 11:9738–66. https://doi.org/10.18632/aging.102422 [PubMed]