Skip to main content
3 Biotech logoLink to 3 Biotech
. 2021 Apr 24;11(5):235. doi: 10.1007/s13205-021-02768-x

Exploring the endogenous potential of Hemidesmus indicus against breast cancer using in silico studies and quantification of 2-hydroxy-4-methoxy benzaldehyde through RP-HPLC

Akash Anand Bansod 1, Gnanam Ramasamy 1,, Bharathi Nathan 1, Rajamani Kandhasamy 1, Meenakshisundaram Palaniappan 1, Santhanakrishnan Vichangal Pridiuldi 1
PMCID: PMC8068754  PMID: 33968579

Abstract

Being a woman and getting older are the main risk factors for breast cancer. While admitting the increasing prevalence of breast cancer among females globally, there is an increasing urge for widening the range of chemical compounds that can act as potential inhibitors for certain cancer target receptors. Current investigation involves virtually screening of 19 protein receptors having major role in signal transduction pathway of breast cancer development against 47 compounds present in Hemidesmus indicus. Virtual screening and supplementary analysis were performed using freely available softwares, tools and online servers. To obtain meaningful results, a comparative scenario was created by screening FDA-approved drugs/drug analogues against the same 19 receptors by keeping all the parameters same as to that of ligands. Two ligands namely Taraxasteryl acetate and Rutin were found to be the best ligands with high binding affinity towards six protein receptors establishing strong receptor ligand interactions. Furthermore, the major volatile compound, a high demand flavouring agent and an isomer of vanillin, namely 2-hydroxy-4-methoxy benzaldehyde (MBALD) specifically found in the roots of Hemidesmus, was quantified by RP-HPLC using a reverse phase C-18 column. The methanolic extract of fresh roots was found to contain 0.221 mg of MBALD/gram of tissue. From the current investigation, it could be surmised that Hemidesmus indicus had demonstrated its potential in both pharmaceuticals and the food industry.

Keywords: Breast cancer, HPLC, MBALD, Virtual screening

Introduction

Breast cancer is the second-leading cause of cancer death in women, aside from skin cancer, and is the highest cause of mortality among female cancer patients. Breast cancer (BC) accelerates an uncontrolled growth of particular breast cells and part of them can metastasize to other body parts eventually leading to death. Out of all, breast cancer accounts 25% of all types of newly diagnosed cancers in women (Hwang et al. 2019). Due to the heavy occurrence of this type of cancer, around 40,000 women die annually in the US alone, therefore, imposes serious threats among women globally. Among the various reasons involved in causing breast cancer, the most associated reason is the genetics besides chances of acquiring it with an increase in age in women, by virtue of this, it is considered as the disease of aging. This disease is characterized by uncontrolled cell proliferation, aberrant cell apoptosis, and tumor formation in breast tissues (Gam 2012). The most common yardstick for classifying breast cancer into four subtypes (Table 1) is based on the status of cell surface receptors or biomarkers viz., the estrogen receptor (ER), the progesterone receptor (PR), and the human epidermal growth factor 2 (HER2) receptor (Eroles et al. 2012). Among all these subtypes of breast cancer, triple-negative breast cancer (TNBC), an aggressive form of breast cancer exhibits heterogeneity at multiple levels hence constituting significant diagnostic and therapeutic challenges. It is one of the most serious threats as it has to be cured only by conventional chemotherapy and radiation therapy and no FDA-approved drugs are available for this condition. (Hwang et al. 2019). Though Trodelvy (drug) was granted accelerated approval by FDA, yet due to the risk of severe neutropenia, severe diarrhoea, hypersensitivity reactions including severe anaphylactic (allergic) reactions, it demands continuous patient monitoring and further clinical trials are required (FDA.gov 2020).

Table 1.

Subtypes of Breast Cancer based on the status of biomarkers

Subtype of cancer Biomarker status Prevalence %
ER* PR* HER2*
Luminal A Positive Positive Negative most common of all, 50–60% of all breast cancers
Luminal B Positive and/or Positive Positive 10 and 20% of all breast cancers
HER2 over-expressing Negative Negative Positive 15 to 20% percent of all breast cancers
Triple negative breast cancer (TNBC) Negative Negative Negative 15–20% percent of all breast cancers

*ER Estrogen Receptor; PR Progesterone Receptor; HER2 Human Epidermal Growth Factor 2

In developing countries, a silent crisis persists in cancer treatment and at least 50–60% of cancer victims use radiotherapy for the destruction of cancerous tumors, but the search for an inexpensive alternative therapy with minimal side effects persists. Though different conventional and non-conventional medicines have been prescribed, the adverse effects and dissatisfaction among users could not give enough relief to patients (Baskar et al. 2012).

The ethnobotanical knowledge has helped us to cure many fatal diseases with ease and the process continues. In this context, a rich diversity of chemical compounds found in plants, based on their intrinsic complexity, could represent a novel and promising approach as they can interact with different molecular receptors (Ferruzzi et al. 2013). From ancient times, mankind is exploiting mother nature particularly herbs to obtain natural products with varied chemical compositions which provides wide spectra of applications in various fields for finding novel lead compounds against a disease (Cragg and Newman 2013).

Hemidesmus indicus (HI) is one of the important medicinal plants used from ancient times for healing many ailments including cancer and holds significance in Ayurveda (Kawlni et al. 2017). Simple aqueous extract of HI roots consists of a variety of phytochemicals including phenols (1.1%), flavonoids (1.12%), saponins (12.55%), terpenoids (0.79%), coumarins (0.91%), alkaloids (1.23%), and tannins (3.06%) (Ananthi et al. 2010) and a series of novel compounds like coumarino-lignans (hemisdesmins) and steroidal glycosides (hemidesmosides A–C) (Manjulatha et al. 2014). Amidst all these compounds, an expensive and high-value flavor product, 2-hydroxy-4-methoxy benzaldehyde (MBALD) (Fig. 1) C8H8O3 is the major compound of interest which displays a typical aroma to the roots and constitutes 91% of the total steam distillation product from the root (Darekar et al. 2009). The molecular weight is 152.147 g/mol, same as vanillin; the difference being in the positions of the hydroxyl and methoxy groups and thus often used as a substitute for vanilla in ice creams.

Fig. 1.

Fig. 1

2-Hydroxy-4-methoxy benzaldehyde (MBALD)

Many reports are available on the anti-cancer activity of H. indicus. Thabrew et al. (2005) documented the cytotoxic activity of roots decoction on HepG2 cells. Pal et al. (2014) observed H. indicus decoction to be cytotoxic to MCF7 breast cancer cell line. Similar results could be seen in other publications on cancer prevention by H. indicus (Banerji et al. 2017; More and Mali 2018; Swathi et al. 2019).

In the present work, an attempt has been made to investigate and reveal the anti-breast cancerous property of H. indicus through in silico studies. The structure-based virtual screening (SBVS) was utilized to screen the compounds present in H. indicus against the potential drug targets for breast cancer and a comparison was also made with already available FDA-approved drugs in respect of binding interactions with receptor proteins using freely available softwares, tools, and online servers. Proteins that were found to be potential drug targets were chosen and used for the construction of a protein library. Based on earlier reports on GC/MS studies of H. indicus, the compounds discovered during the research were thoroughly analysed and compounds that were common from each study were chosen as ligands. A freely available software, Pyrex 0.8 was used for performing SBVS that utilized the Autodock Vina tool for virtual screening. Absorption, distribution, metabolism, and excretion (ADME) analysis was carried out for determining the drug likeness nature of the ligands using SwissADME (online free server). Potential hits obtained after in silico studies presumably be further used for drug development against breast cancer after carrying out in vitro experiments, in vivo studies and clinical trials as well. Further efforts were made using aqueous methanolic extract to quantify 2-hydroxy-4-methoxy benzaldehyde, an astounding food flavouring metabolite and a major compound from fresh roots of Hemidesmus indicus.

Materials and methods

In silico virtual screening

Protein library preparation: Current investigation includes 19 different proteins that behave as target receptors having a role in different cell regulation pathways and are involved in causing breast cancer.

These 19 protein receptors were found to be actively participating in inducing breast cancer which were identified from breast cancer pathway map05224 of the Kyoto Encyclopedia of Genes and Genomes (KEGG) and were further analysed for their role using available literature. The list of 19 proteins selected as target for ligands for virtual screening is given in Table 2. X-ray crystallographic structures of these proteins already complexed with their respective ligands were retrieved from the RCSB Protein Data Bank (Table 3).

Table 2.

Proteins selected as target receptors and their role in cancer development

Sr. no Receptor Role in cancer References
1 Akt, also known as Protein kinase B

PI3K/AKT or Kinase signalling pathway plays major role in cellular processes

The abnormal overexpression or activation of AKT has been found to be associated with risk of many cancers, increased cancer cell proliferation and their survival

Song et al. (2019)
2 CDK2

Cyclin-dependent kinases (CDKs) are mainly involved in regulation of cell-cycle

The characteristics of “S” and ‘G2/M-phase” regulated by CDK1/CDK2 are significant events that may cause abruption in the process of cell division resulting in cancer

Chohan et al. (2018)
3 CDK6

Tumor growth can be seen as a result of aggravated levels of CDK4/CDK6 during

G1-phase of cell cycle

Chohan et al. (2018)
4 EGFR: epidermal growth factor receptor In cancer condition, EGFR is often constantly stimulated for the continuous production of EGFR ligands in the tumor microenvironment or due to mutation in EGFR which locks this protein receptor in a state of continual activation Akhtar and Benter (2013)
5 ERK2 Certain processes in mammalian cells including cell cycle progression, differentiation, protein synthesis, metabolism, survival, migration and senescence are carried out by the RAS/RAF/MEK/ERK pathway. Mutations in gene components of these pathway like RAS or b-RAF leads to tumorigenesis Ward et al. (2015)
6 Estrogen receptor (ER1) Estrogen receptor viz. it’s receptor-mediated hormonal activity causing cellular proliferation, increased mutation rates causing direct genotoxic effects through a cytochrome P450-mediated metabolic activation, and induction of aneuploidy Russo and Russo (2006)
7 FGFR: fibroblast growth factor receptors FGF/FGFR signalling network under certain conditions plays a critical role in cancer cell proliferation, survival, differentiation, migration, and apoptosis Tucker et al. (2014)
8 Grb2 Grb2 upregulation in association with EGFR/HER2 signalling results in breast cancer development Ijaz et al. (2017)
9 HER2: human epidermal growth factor receptor 2 HER2-positive type patients show amplification or overexpression of the gene, i.e., upto 25–50 copies of gene, and up to 40–100-fold increase in HER2 protein resulting in 2 million receptors expressed at the tumor cell surface Iqbal and Iqbal (2014)
10 IGFR1: insulin like growth factor receptor 1 In some breast cancer subtypes, activation and over-expression of IGF-1R has been found, additionally disease progression, increased resistance to radiotherapy and poor prognosis are linked with the molecules involved in downstream signalling of IGF-1R Jackson and Lee (1997)
11 MAP2K1 also known as MEK1 Mitogen-activated protein kinase (MAPK) pathway is complex pathway which involves Ras/Raf/MEK/ERK receptor signaling cascade. As this pathway majorly deals with cell cycle progression and maintenance, any aberrant change may lead to tumorigenesis Isshiki et al. (2011)
12 PARP1: poly ADP-ribose Polymerase1 One of the hallmarks of cancer is genomic instability caused by defect in DNA repair mechanism of a cells which is normally carried out by PARP proteins and subsequent mutations in that cells that promote the development of genotypes are found to be favourable for tumorigenesis Dulaney et al. (2017)
13 PARP2: poly ADP-ribose Polymerase 2 Same as PARP1 Dulaney et al. (2017)
14 PI3K: phosphoinositide 3-kinase In triple-negative breast cancer, aberrant activation of the PI3K pathway is mostly observed as the main cause Papadimitriou et al. (2018)
15 PIM1: proto-oncogene 1 Overexpression of PIM1 has been observed in many cancer subtypes and acts as potential biomarker which contributes to the growth of cancer cells under abnormal conditions Gao et al. (2019)
16 PTK6: protein tyrosine kinase 6 PTK6 overexpression contributes to anchorage-independent survival, proliferation, and migration of breast cancer cells Park et al. (2015)
17 SOS1: son of sevenless1 Overexpression of SOS1, a component of EGF-dependent pathways facilitates cell growth and survival of cancer cells De et al. (2014)
18

VEGFR: vascular endothelial growth factor A (VEGF-A)

VEGFR1

VEGF-A primarily mediates tumor angiogenesis via two receptors VEGFR-1 and VEGFR-2. To date, the role of VEGFR-1 in angiogenic signal delivery for VEGF in tumor angiogenesis is poorly examined and still not entirely clear. Targeting the VEGF receptors VEGFR-1 & VEGFR-2 will have a potential impact on the motility of tumor cells Srabovic et al. (2013)
19 VEGFR2 Same as VEGFR1 Srabovic et al. (2013)

Table 3.

Various protein receptors with PDB ID with Amino acid involved in active site complexed with respective drugs/inhibitors

Sr Protein PDB ID Amino acid residues of active sites Inhibitor/drug (from Pubchem) References
1 Akt 4GV1 Glu228, Ala230, Glu234, Glu278, Asp292 Capivasertib Addie et al. (2013)
2 CDK2 1PYE Leu83, Phe82, Lys33, Gln131, Glu81, Ile10, Asp145, Leu134 Aminoimidazo[1,2-a] pyridines Hamdouchi et al. (2004)
3 CDK6 5L2I His100, Thr107, Phe98, Lys43, Tyr24, Asp104 Palbociclib Chen et al. (2016)
4 EGFR 1M17 Gly696, Gly1022 Afatinib Stamos et al. (2002)
5 ERK2 4ZZN Asp106, Lys54, Leu107, Gln105, Cys166, Glu71, Asp167, Met108 CQ8 Ward et al. (2015)
6 ER1 2OUZ Asp 351, Leu540, His524, Leu525 Lasofoxifene Vajdos et al. (2007)
7 FGR1 4V01 Tyr563, Val561, Glu531, Ala564, Phe642, Asp641, Ile620 Ponatinib Tucker et al. (2014)
8 Grb2 1X0N Arg67, Arg86, Lys109, His107, Trp121, Leu11 BDBM50102025 Ogura et al. (2008)
9 HER2 3RCD Met801, Lys753, Asp863, Glu770, Gly865, Phe864, Thr862 TAK-285 Ishikawa et al. (2011)
10 IGFR1 3D94 Gly1122, Gly1125, Met1052, Met1049, Met1112, Lys1003, Asp1056, Asp1123, Phe1124, Phe980, Leu975 PQIP Wu et al. (2008)
11 MEK1 3OS3 Lys97, Val127, Val211, Ser212, CH4858061 Isshiki et al. (2011)
12 PARP1 4UND Glu988, Tyr896, Tyr907, Asp766, Gly863, Arg878, Ser904, Glu763 Talazoparib Thorsell et al. (2017)
13 PARP2 4TVJ Arg444, Tyr462, Tyr473, Ser470, Gly429, Glu335, Asp339 Olaparib Thorsell et al. (2017)
14 PI3K 3L08 Val882, Lys833, Tyr867 Omipalisib Knight et al. (2010)
15 PIM1 2O64 Arg122, Ser54, Leu120, Glu89, Glu121, Glu124, Asp186, Lys67, Val126,Pro123 Quercetagetin Holder et al. (2007)
16 PTK6 5H2U Arg195, Ala217, Glu235, Glu274, Ile262, Phe331, Asp330, Gly329, Leu248, Leu319, Ser271, Met267, Thr264 Dasatinib Thakur et al. (2017)
17 SOS1 5OVE Asn879, His905, Leu901, Met878 AXE Hillig et al. (2019)
18 VEGFR1 3HNG Glu878, Asp1040, Cys912, Ile1038, Val907, Glu910, Tyr 911, Ala 859 CHEMBL101683 Tresaugues et al. (2013)
19 VEGFR2 3WZD Ala866,Phe918,Val848,Val898,Val899, Cys919, Gly841,922, Asn923, Leu840, Leu1035,Leu1049, Ile888, Asp146 Levatinib Okamoto et al. (2015)

Binding-site analysis: The receptors were chosen from the available literature and appropriate data were collected regarding the binding sites for each receptor which allowed site-specific docking of ligands for more precise binding.

Preparation of chemical library: For identifying the compounds present in Hemidesmus indicus many researchers have contributed to the literature. During GC/MS analysis of Hemidesmus indicus, Nagarajan et al. (2001), Murugan et al. (2018), and Sharma et al. (2017) have extracted many compounds with organic solvents from the root powder. A total of 47 such compounds that were found to be prominent and common from these three articles were pooled and selected to serve as ligands (Table 4). All the compounds were retrieved from the PubChem database in SDF format. Compounds were converted to mol2 format using Open babel 3.0.0. (Boyle et al. 2011). For examining the significance of binding affinity of ligands with the protein receptors, currently available FDA-approved drugs for each protein receptor were also included in the study to get a meaningful comparison between the binding affinity of ligands and drugs towards the receptors. FDA-approved drugs for TNBC subtype were not available mainly since all the three particular protein receptors were not getting expressed simultaneously and this put together drive the disease difficult to cure. Under the above-mentioned circumstances, during this investigation, docking against any particular subtype of breast cancer was not focused. A separate drug library was maintained after downloading their structures from Pubchem.

Table 4.

Various H. indicus compounds selected for virtual screening with receptors

Sr. no Compound Sr. No Compound
1 1,8-Cineole 25 Ferulic acid
2 2-Hydroxy-4- methoxy benzoic acid 26 Guaiacol
3 2-Hydroxy-4-methoxy-benzaldehyde 27 Hemidescine
4 3-Hydroxy-4-methoxy-benzaldehyde 28 Hemidine
5 4-Hydroxy-3-methoxy-benzaldehyde 29 Heminine
6 4-Isobutylaniline 30 Hemisine
7 16-Dehydropregnenolone 31 Indicine
8 Alpha-Amyrin 32 Isocaryophyllene
9 Alpha-Amyrin acetate 33 Isoquercitin
10 Alpha-Terpinyl acetate 34 Ledol
11 Anisaldehyde 35 Linalyl acetate
12 Beta-Amyrin 36 Lupanone
13 Beta-Amyrin acetate 37 Lupeol acetate
14 Beta-Amyrin palmitate 38 Lupeol
15 Beta-Sitosterol 39 Medidesmine
16 Borneol 40 Nerolidol
17 Campesterol 41 Nonadienal
18 Camphor 42 Octanoic acid
19 Cholesterol 43 Palmitic acid
20 Decanoic acid 44 Rutin
21 Desmisine 45 Salicylaldehyde
22 Dihydrocarvyl acetate 46 Taraxasteryl acetate
23 Dodecanoic acid 47 Thymol
24 Emidine

Protein preparation: Nineteen protein receptors maintained in the protein library were prepared for further analysis and were subjected to water removal and the removal of inhibitors/drugs complexed with protein receptor using UCSF Chimera 1.14 and were saved in PDB format. For preparation of proteins for virtual screening, Autodock 1.5.6 was utilized where polar hydrogens and Gasteiger charges were added (Morris et al. 2014). Proteins were finally saved in PDBQT format for further processing.

Virtual screening: in silico analysis of screening the 47 ligands against 19 receptor proteins were done by Pyrex 0.8 which has a built-in Autodock Vina tool for doing virtual screening (Dallakyan and Olson 2015). Energy minimization of ligands was done before doing screening to generate alternate conformations with rotatable bonds. After selecting ligands and macromolecules, the grid box (Table 5) was set to the required dimensions to achieve site-specific binding. The exhaustiveness of the run was kept at 8 (1–8) by default which sets the number of runs in parallel producing several results and captures the promising intermediate results and merges them in final results. The docking strategy followed involved screening of 47 ligands against one protein receptor at a time. All 19 receptors were screened in the same manner. The comparative scenario was created by performing virtual screening of FDA-approved drugs/drug analogues (Table 6) against receptors while keeping all the parameters same as that for ligands. Flowchart of virtual screening strategy is shown in Fig. 2.

Table 5.

Grid box dimensions used for target receptors for site-specific binding

Sr. no Protein receptor PDB ID Center grid box Grid dimensions
1 Akt 4GV1

center_x = − 26.4433962881

center_y = 5.87999413858

center_z = 11.481468964

size_x = 17.5491814942

size_y = 15.1645658997

size_z = 21.8659356895

2 CDK2 1PYE

center_x = 12.1595741137

center_y = − 7.70462760237

center_z = 23.0591549663

size_x = 17.7521482273

size_y = 19.2645153251

size_z = 19.3582063978

3 CDK6 5L2I

center_x = 13.6859667843

center_y = 27.3511289596

center_z = 6.22084657539

size_x = 16.7923335687

size_y = 20.0245340571

size_z = 17.5056366158

4 EGFR 1M17

center_x = 23.5645

center_y = 1.03071634208

center_z = 59.3942

size_x = 25.0

size_y = 16.0328635225

size_z = 25.0

5 ERK2 4ZZN

center_x = − 12.2250832319

center_y = 8.05511487232

center_z = 40.667428966

size_x = 29.0012970612

size_y = 21.8798196449

size_z = 25.4618379814

6 ER1 2OUZ

center_x = 35.710540759

center_y = − 1.11024654188

center_z = 19.8339786257

size_x = 21.971205293

size_y = 11.4629069162

size_z = 21.0837572515

7 FGR1 4V01

center_x = 91.6988892157

center_y = 1.02538503952

center_z = 14.3191711067

size_x = 14.6200017781

size_y = 14.214699205

size_z = 28.5126577866

8 Grb2 1X0N

center_x = 10.4649827631

center_y = − 5.06671193968

center_z = − 11.4349550251

size_x = 19.3516503493

size_y = 16.0383761206

size_z = 16.9432965669

9 HER2 3RCD

center_x = 9.57195981331

center_y = 0.245084953123

center_z = 30.4012441869

size_x = 22.2285879054

size_y = 14.2196042874

size_z = 29.4330189124

10 IGFR1 3D94

center_x = 24.701693415

center_y = 20.141

center_z = − 5.69169550793

size_x = 20.4824446255

size_y = 25.0

size_z = 21.3766089841

11 MEK1 3OS3

center_x = 6.50648876111

center_y = 40.8855898132

center_z = − 7.97359571258

size_x = 12.4307617464

size_y = 18.6773087119

size_z = 11.4541015458

12 PARP1 4UND

center_x = 2.52013090416

center_y = 64.7952621889

center_z = 189.002653312

size_x = 22.1182356719

size_y = 33.4007987591

size_z = 26.1022239335

13 PARP2 4TVJ

center_x = 17.1830806036

center_y = − 1.36290358543

center_z = 15.2093271218

size_x = 18.5955056509

size_y = 23.4819401532

size_z = 19.9685457564

14 PI3K 3L08

center_x = 22.642825206

center_y = 9.16699071362

center_z = 25.655091069

size_x = 20.0860669009

size_y = 22.7169166893

size_z = 29.8341638301

15 PIM1 2O64

center_x = 75.3772686654

center_y = 36.5030541821

center_z = − 2.32919493626

size_x = 18.6933550319

size_y = 29.1592228792

size_z = 18.0199945636

16 PTK6 5H2U

center_x = 30.939769054

center_y = − 0.972784774999

center_z = 40.7047208664

size_x = 26.4239844981

size_y = 33.6100567727

size_z = 34.7953371444

17 SOS1 5OVE

center_x = − 1.13570940655

center_y = − 32.3718926619

center_z = 42.5591887498

size_x = 18.1005232395

size_y = 19.2871979035

size_z = 13.389975125

18 VEGFR1 3HNG

center_x = 5.3838

center_y = 18.4599

center_z = 25.3055

size_x = 25

size_y = 25

size_z = 25

19 VEGFR2 3WZD

center_x = 3.2326708109

center_y = − 4.76562803382

center_z =  z = 14.3045293041

size_x = 23.9182816046

size_y = 22.0987087958

size_z = 30.4377413918

Table 6.

Target protein receptors and their corresponding FDA-approved drugs/drug analogues selected for virtual screening

Sr. no Target protein FDA-approved drugs/drug analogues
1 CDK2 Aminoimidazo [1,2a] pyridines
TG-02
AT7519
AC1NCSZQ
Dinaciclib
Milciclib
2 EGFR Erlotinib
Neratinib
Gefitinib
Lapatinib
Osimertinib
3 ERK2 CQ8
AC1M8GZK
Ravoxertinib
DEL-22379
LY3214996
4 HER2 TAK-285
Afatinib
Lapatinib
Neratinib
Aplaviroc
5 PARP1 Talazoparib
Olaparib
Niraparib
Rucaparib
6 PARP2 Olaparib
Niraparib
Rucaparib
7 PIM1 Quercetagetin
Lapatinib
Dabrafenib
Idelalisib
Vemurafenib
Nilotinib
8 PTK6 Dasatinib
Pazopanib
Dasatinib
Vandetanib
Sunitinib

Fig. 2.

Fig. 2

Strategy followed for virtual screening

In silico ADME assessment: Usually, ADME assessment is the first step after selecting ligands but in this investigation, all 47 ligands were first screened against target receptors followed by their selection based on the performance of ADME parameters. In the first round of selection, ligands coupled with receptors were checked for ADME properties, the protein receptors with complexed ligand (that gave satisfactory results) were forwarded to the next round of screening against FDA-approved drugs. The rationale behind delaying ADME profiling was to avoid rejection of potent and possible drug-like ligands in the initial steps, moreover, in case, if a well-performing ligand fails to stand ADME parameters, it may be modified based on the need. For ADME analysis, ligands originally in SDF format were converted into smiles format using Open babel 3.0.0. and smiles were submitted onto the Swiss ADME online server for analysis that tested compounds for 46 various parameters. The obtained results were checked for few main parameters such as molecular weight, gastrointestinal (GI) absorption, blood brain barrier (BBB), P-glycoprotein substrates and inhibitors, cytochrome P (CYP) inhibitory promiscuity, Lipinski's rule of 5 (LRo5), druglikeness violations, and synthetic accessibility, otherwise the selection would have been difficult with all 46 parameters. Scoring was done for all 47 compounds using the above-mentioned parameters.

High-performance liquid chromatography

Plant material: Hemidesmus indicus plants were collected from the Forestry College and Research Institute, Mettupalayam (11.19′ N, 77.56′ E), Coimbatore, Tamil Nadu, India, and were maintained at the greenhouse facility available at the Department of Plant Biotechnology, Tamil Nadu Agricultural University, Coimbatore. Fresh roots were used for the study.

Reagents and chemicals: Solvents including methanol and TFA used were of HPLC grade; mobile phase, and other reagents were prepared using Milli Q water.

HPLC condition: HPLC analysis was performed on the Shimadzu HPLC system equipped UV–Vis detector. Separation of compounds was achieved by the C18 reversed-phase column (INNO column, 5 µm, 120 Å, 4.6 × 250 mm). Shimadzu CLASS-VPTMsoftware was used for data acquisition, processing, and reporting on the Windows XP platform. Solvent preparation was carried out by following the protocol given by Sircar et al. (2007). Isocratic solvent mixture (mobile phase) was prepared by adding 1 mM aqueous TFA and methanol in a 70:30 ratio with a flow rate of 1 ml min−1. The wavelength was set to 280 nm for monitoring chromatograms. The sample was identified based on the comparison of retention time with those of the standard with keeping the same conditions.

Standard solution and sample preparation: HPLC grade (98%) 2-hydroxy-4-methoxy benzaldehyde standard was purchased from Sigma–Aldrich (Catalogue No. 160695, molecular weight 152.15 g/mol and PubChem Substance ID 24849887). A 1000 ppm standard stock solution was prepared by adding 1 mg of MBALD in 1 mL of aqueous methanol (50:50, v/v). The working standard solution was prepared with a concentration of 10 ppm by dilution of standard stock solution with aqueous methanol (50:50, v/v). All stock solutions were stored at 4 °C.

Sample plants maintained at the greenhouse were uprooted and roots were washed thoroughly under tap water to remove excess particles. 1 gm of fresh roots were macerated into a fine powder using liquid nitrogen and extracted with 5 mL of aqueous methanol (50:50, v/v). The extract was incubated for 2 days with continuous shaking and was subjected to centrifugation at 10,000 rpm for 10 min. The supernatant taken was first filtered with Whatmann filter paper no. 1 and further filtered through a 0.22 µm filter. 20 µl of the sample was injected into the HPLC system.

For the quantification purpose of 2-hydroxy-4-methoxy benzaldehyde, the retention time and peak area were obtained from the chromatogram. The formula for calculating the percentage of 2-hydroxy-4-methoxy benzaldehyde in the methanolic extract used is as follows:

Peak area of samplePeak area of standard×Concentration of standardConcentration of sample×Purity of standard

Results

In silico analysis

The current study aimed to reveal the anti-breast cancerous property of H. indicus phytochemicals by using in silico tools. Forty-seven compounds from H. indicus were chosen as ligands and in silico virtual screening (VS) was performed against the target proteins (Table 7). Complete SBVS analysis suggested that out of 19 target receptors, 6 protein receptors showed high binding affinity towards the ligands as compared to that of FDA-approved drugs (Table 8). Two ligands namely Taraxasteryl acetate and Rutin were majorly involved in building these strong interactions with minimum binding energy with the target receptors. Whereas, ERK2 and PIM1 were not considered for final results as they showed more binding affinity towards FDA-approved drugs as compared to that of their respective ligands.

Table 7.

Virtual screening of ligands with protein receptors and their binding energies

Sr. no. Receptor Ligand Ligand ID and energy Binding energy (kCal/mole) rmsd/ub rmsd/lb
1 Akt 2,Hydroxy-4-methoxy benzoic acid 21292822_uff_E = 167.22 − 7.5 0 0
2 *CDK2 Taraxasteryl acetate 13970053_uff_E = 798.31 − 11 0 0
3 CDK6 Heminine 102014181_uff_E = 909.83 − 9.6 0 0
4 *EGFR Taraxasteryl acetate 13970053_uff_E = 1772.22 − 11 0 0
5 *ERK2 Rutin 5280805_uff_E = 751.59 − 9 0 0
6 Estrogen/ ER1 16-Dehydro-pregnenolone 92871_uff_E = 438.85 − 9.3 0 0
7 FGFR Hemidine 101594607_uff_E = 718.42 − 8.6 0 0
8 Grb2 Emidine 101664026_uff_E = 1502.28 − 7.5 0 0
9 *HER2 Rutin 5280805_uff_E = 751.59 − 10.8 0 0
10 IGFRK1 Emidine 101664026_uff_E = 1502.28 − 8.6 0 0
11 MEK1 2,Hydroxy-4-methoxy benzoic acid 21292822_uff_E = 167.22 − 7.3 0 0
12 *PARP1 Rutin 5280805_uff_E = 751.59 − 12.4 0 0
13 *PARP2

Taraxasteryl

acetate

13970053_uff_E = 798.31 − 12.4 0 0
14 PI3K Desmisine 102446079_uff_E = 1499.09 − 10.2 0 0
15 *PIM1 Hemidescine 101664025_uff_E = 922.51 − 9.7 0 0
16 *PTK6 Rutin 5280805_uff_E = 751.59 − 10.4 0 0
17 SOS1 Cholesterol 5997_uff_E = 549.32 − 7.6 0 0
18 VEGFR1 Lupanone 129730785_uff_E = 857.53 − 9.9 0 0
19 VEGFR2 Campesterol 173183_uff_E = 573.30 − 9.8 0 0

*Indicates the receptor with respective ligand selected on the basis of ADME properties of ligands to be forwarded for next round of screening against FDA-approved drugs

Table 8.

Final comparison for minimum binding energy between FDA-approved drugs and H. indicus compounds selected for virtual screening with receptors

Sr. no. Target protein FDA-approved drugs/drug analogues ID & energy Binding energy (kCal/mole) Ligand ID & energy Binding energy of ligands
(kCal/mole)
1 CDK2 Aminoimidazo [1,2a] pyridines 11275025_uff_E = 290.74 − 5.3 Taraxasteryl acetate 13970053_uff_E = 798.31 − 11
TG-02 16739650_uff_E = 341.49 − 8
AT7519 11338033_uff_E = 609.37 − 6.9
AC1NCSZQ 45380979_uff_E = 1851.79 − 6.1
Dinaciclib 46926350_uff_E = 514.91 − 7.1
Milciclib 16718576_uff_E = 766.77 − 6.8
2 EGFR Erlotinib 176870_uff_E = 599.78 − 7.8 Taraxasteryl acetate 13970053_uff_E = 1772.22 − 11
Neratinib 9915743_uff_E = 818.89 − 9.4
Gefitinib 123631_uff_E = 518.86 − 7.9
Lapatinib 208908_uff_E = 1028.85 − 9.1
Osimertinib 71496458_uff_E = 865.16 − 8.1
3 *ERK2 CQ8 91758407_uff_E = 399.97 − 7.3 Rutin 5280805_uff_E = 751.59 − 9
AC1M8GZK 2486631_uff_E = 961.91 − 9.6
Ravoxertinib 71727581_uff_E = 578.46 − 8.2
DEL− 22379 11224574_uff_E = 891.85 − 8.2
LY3214996 121408882_uff_E = 938.70 − 7.6
4 HER2 TAK-285 11620908_uff_E = 669.77 − 6.2 Rutin 5280805_uff_E = 751.59 − 10.8
Afatinib 10184653_uff_E = 543.13 − 6.7
Lapatinib 208908_uff_E = 1028.85 − 8
Neratinib 9915743_uff_E = 818.89 − 6.6
Aplaviroc 3001322_uff_E = 576.31 − 7.9
5 PARP1 Talazoparib 135565082_uff_E = 560.9 − 9.5 Rutin 5280805_uff_E = 751.59 − 12.4
Olaparib 23725625_uff_E = 1707.03 − 9.8
Niraparib 24958200_uff_E = 461.77 − 8.1
Rucaparib 9931954_uff_E = 653.31 − 8.1
6 PARP2 Olaparib 23725625_uff_E = 1707.03 − 9.7 Taraxasteryl acetate 13970053_uff_E = 798.31 − 12.4
Niraparib 24958200_uff_E = 461.77 − 9.2
Rucaparib 9931954_uff_E = 653.31 − 8.6
7 *PIM1 Quercetagetin 5281680_uff_E = 392.35 − 9.0 Hemidescine 101664025_uff_E = 922.51 9.7− 
Lapatinib 208908_uff_E = 1028.85 − 9.8
Dabrafenib 44462760_uff_E = 1053.48 − 10.2
Idelalisib 11625818_uff_E = 669.97 − 8.7
Vemurafenib 42611257_uff_E = 1132.51 − 9.8
Nilotinib 644241_uff_E = 626.59 − 10.8
8 PTK6 Dasatinib 3062316_uff_E = 744.65 − 6.4 Rutin 5280805_uff_E = 751.59 − 10.4
Pazopanib 10113978_uff_E = 1068.53 − 6.9
Dasatinib 3062316_uff_E = 744.65 − 6.4
Vandetanib 3081361_uff_E = 464.09 − 6.1
Sunitinib 5329102_uff_E = 836.64 − 6.2

*Shows the receptors which were excluded from final results as they failed to perform better than FDA-approved drugs/drug analogues

To describe briefly, six crystallized protein receptors were strongly bonded with two ligands in their active sites inhibiting them by affecting their interactions with other possible ligands. The maximum number of conventional hydrogen bonds (5) were formed between PTK6 residues namely ASP330, LYS219, ASN317, ARG316, GLU274, and Rutin that provided the stability to the complex. Whereas four conventional hydrogen bonds each were formed in the case of ERK2, HER2, and PARP1 when complexed with Rutin. EGFR formed three hydrogen bonds while CDK2 failed to form any hydrogen bonds when interacted with Taraxasteryl acetate. In case of virtual screening of FDA-approved drugs, two drug compounds viz., (1) AC1M8GZK (docked with ERK2 with the binding energy − 9.6) and (2) Dabrafenib (complexed with PIM1 with the binding energy − 10.2) bonded strongly with the receptors than the corresponding ligands. A detailed description of receptor–ligand interactions is given in the Table 9. Interactions of best receptor-FDA-approved drug/drug analogue complexes were also studied and results are summarized in Table 10.

Table 9.

Residues involved in the interactions between target receptors and ligand

Interactions CDK2-
Ligand
EGFR-
Ligand
ERK2-
Ligand
HER2-
Ligand
PARP1-
Ligand
PARP2-
Ligand
PIM1-
Ligand
PTK6-
Ligand
van der WAALS GLU8, ILE10, GLY11, ALA31, LYS33, PHE80, PHE82, LEU83, HIS84, GLN85, ASP86, LYS89, ASN132, LEU134 ALA719, MET769, LEU768, PRO770, GLY772, LEU820, CYS773, VAL702, ASP831, LYS721, MET742, LEU764 ASP165, ALA33, TYR34, GLU31, GLY32, LYS112, ILE82, ASP104, SER151, CYS164, THR108, MET106, LEU105, LYS52 VAL782, ILE752, THR798, ALA751, CYS805, LEU852, VAL734, GLY729, PHE731, ILE767, GLU770, GLY865, ASP863, PHE864, ILE861, ARG784 ASN868, ARG865, HIS909, GLN759, GLY888, LEU769, ILE872, LEU877, GLY876, ILE895, TRP861, ASN987, TYR9989, GLU988, SER904, GLY863, LYS903 ASN434, ILE438, LEU443, HIS428, TYR473, GLN324, ILE461, ASP339, ARG444, GLU335, ILE445, ALA446, TYR455, GLY454

PRO42, GLU124, PRO125, GLU171, VAL126, LEU44, VAL52, ILE185, LYS67, ASP186, GLU89, LEU120, ILE104, LEU174,

ASP128, GLN127

LEU266, MET267, ALA268, GLY270, SER271, LEU273, GLY200, PHE202, GLY329, LEU248, THR264, ILE262, GLY198
CONVEN-TIONAL HB (no.)

(3)

THR766, CYS751, THR830

(4) ASP109, GLN103, GLU107, ASN152 (4) ARG849, THR862, LEU785, SER783 (4) TYR896, PHE897, GLU763, SER864

(1)

MET456

(2)

SER54, ARG122

(5)ASP330,

LYS219,

ASN317, ARG316, GLU274

CARBON HB ARG878 SER199
PI CATION LYS219
PI DONOR HB TYR889
PI SIGMA ILE29 LEU785 TYR462 PHE49

LEU197

LEU319

PI SULFUR MET774
PI–PI STACKED TYR907
PI–PI T SHAPED TYR896 -
ALKYL VAL18, ALA144 LEU694 ALA880
PI AKYL ALA50, LEU154, VAL37 LEU796, LYS753, LEU755 ALA898 VAL205, ALA217

UNFAV

ACCEPTOR-

ACCEPTOR

ASN850

UNFAV

DONOR-DONOR

–– - ARG878 ASN317

HB stands for hydrogen bond

UNFAV stands for unfavourable

Table 10.

Residues involved in the interactions between target receptors and FDA-approved drugs

Interactions CDK2-FDA EGFR-
FDA
ERK2-
FDA
HER2-
FDA
PARP1-
FDA
PARP2-
FDA
PIM1-
FDA
PTK6-
FDA
van der Waals GLU12, LYS129, GLN131, GLY11, ASP86, LEU134, VAL18

ASP813,

PHE699, LEU834, GLY833,

CYS773,

GLU738,

GLY772

TRP190, TYR191, SER151, GLU31, ILE82, ARG189, THR188, LYS112, GLY32, GLY30, TYR34, ALA33, ASP165

SER728, GLY727, PHE1004, ASP808,

ALA730, LYS753

GLU688, LYS684, GLY913

ASP339,

GLU335, ALA446, ILE445, TYR279, GLY338, ILE342, ILE275

SER46, GLY45, PHE49, ASP186, LEU120, ILE104, ALA65, ARG122, LEU174, PRO123, VAL126, ASP131 ARG431, PRO432, LEU437, CYS423, TRP371, LEU376, GLU298, GLN295, CYS326, HIS244, PHE331, LEU248, LEU303, TYR302
CONVEN-TIONAL HB (no.)

(1)

THR766

(1)

GLN103

(3)

ARG849,

LEU726,

CYS805

(2)

SER911, ASP914

(1)

ARG444

(1)

GLU121

(2)

ASP369

MET300

CARBON HB

THR830,

ASN818

ASP109 GLY729 ASP128, LEU44 ALA297
HALOGEN ASP863, ASP845, ASN850 GLU171
UNFAV BUMP VAL370, GLU304, THR366, PHE434, MET300, CYS301, LEU313, VAL296, GLY299, HIS246, ILE247
UNFAV DONOR-DONOR LYS33
UNFAV +  + 
PI CATION LYS52, L–YS149
PI ANION ASP831 ASP128
PI DONOR HB
PI SIGMA VAL272

ILE185

VAL52

AMIDE PI STACKED
PI SULFUR CYS164 MET300
PI-PI STACKED
PI-PI T SHAPED TYR111 PHE731 PHE373
ALKYL

ARG817,

LEU820,

VAL702,

MET742

ILE29, VAL37, ALA50 LYS276 LYS327, LYS367
PI AKYL

ALA719,

LEU694, LEU764, LYS721

LEU154 VAL734 LYS276 LYS67 LYS327, LYS367
SALT BRIDGE ASP145

HB stands for hydrogen bond

UNFAV stands for unfavourable

 +  + stands for positive–positive

Docked structures of ligands and FDA-approved drugs/drug analogues with target receptors are shown in Figs. 3, 4, 5, 6, 7, 8, 9, 10, whereas, comparative figures of interactions between ligands/receptor FDA-approved drugs with target receptors are shown in Figs. 11, 12, 13, 14, 15, 16, 17, 18. The virtual hits identified in this study could be used as an alternative targeting agent for breast cancer after being tested through in vitro experiments, animal lab studies, and clinical trials.

Fig. 3.

Fig. 3

CDK2 in complex with Taraxasteryl acetate (red) and TG-02 (blue)

Fig. 4.

Fig. 4

EGFR in complex with Taraxasteryl acetate (red) and Neratinib (blue)

Fig. 5.

Fig. 5

ERK2 in complex with Rutin (red) and AC1M8GZK (blue)

Fig. 6.

Fig. 6

HER2 in complex with Rutin (red) and Lapatinib (blue)

Fig. 7.

Fig. 7

PARP1 in complex with Rutin (red) and Olaparib (blue)

Fig. 8.

Fig. 8

PARP2 in complex with Taraxasteryl acetate (red) and Olaparib (blue)

Fig. 9.

Fig. 9

PIM1 in complex with Hemidescine (red) and Nilotinib (blue)

Fig. 10.

Fig. 10

PTK6 in complex with Rutin (red) and Pazopanib (blue)

Fig. 11.

Fig. 11

a Receptor-ligand interaction between CDK2 and Taraxasteryl acetate. b Receptor-ligand interaction between CDK2 and TG-02

Fig. 12.

Fig. 12

a Receptor-ligand interaction between EGFR and Taraxasteryl acetate. b Receptor-ligand interaction between EGFR and Neratinib

Fig. 13.

Fig. 13

a Receptor-ligand interaction between ERK2 and Rutin. b Receptor-ligand interaction between ERK2 and AC1M8GZK

Fig. 14.

Fig. 14

a Receptor-ligand interaction between HER2 and Rutin. b Receptor-ligand interaction between HER2 and Lapatinib

Fig. 15.

Fig. 15

a Receptor-ligand interaction between PARP1 and Rutin. b Receptor-ligand interaction between PARP1 and Olaparib

Fig. 16.

Fig. 16

a: Receptor-ligand interaction between PARP2 and Taraxasteryl acetate. b Receptor-ligand interaction between PARP2 and Olaparib

Fig. 17.

Fig. 17

a Receptor-ligand interaction between PIM1 and Hemidescine. b Receptor-ligand interaction between PIM1 and Nilotinib

Fig. 18.

Fig. 18

a Receptor-ligand interaction between PTK6 and Rutin. b Receptor-ligand interaction between PTK6 and Pazopanib

HPLC analysis

Quantification of 2-hydroxy-4-methoxy benzaldehyde was carried out using fresh roots extracted with aqueous methanolic extract and using an isocratic mixture of 1 mM TFA and methanol in 70:30 ratio as mobile phase with the flow rate of 1 ml min−1 and was monitored at 280 nm with 25–30 °C temperature. The retention time for the standard 2-hydroxy-4-methoxy benzaldehyde at 10 ppm concentration was recorded in HPLC and was observed as 54.400 min at 280 nm (Fig. 19). Besides this, the peak for the sample was achieved at 55.499 min. (Fig. 20). The identity of the sample was confirmed by comparing the chromatograms of both standard and sample. Using the formula for calculating the percentage of compounds present in the sample, it was found that the methanolic root extract of fresh Hemidesmus indicus roots contains 0.221 mg of 2-hydroxy-4-methoxy benzaldehyde/gram of tissue.

Fig. 19.

Fig. 19

HPLC-chromatogram of Standard sample (MBALD) at concentration of 10 ppm

Fig. 20.

Fig. 20

HPLC-chromatogram of fresh root sample of Hemidesmus indicus

Discussion

Breast cancer derails societal stability as the mortality rate of different breast cancer subtypes is alarming and for one subtype, i.e., TNBC, pharmaceutical outputs/drugs are yet to hit the market and conventional therapies are not much effective but the only source of hope.

Earlier works on breast cancer have profoundly shown the anti-breast cancer activity of some aromatic compounds against potential drug targets. Taraxasteryl acetate has shown antiproliferative activity in vitro against TNBC MDA-MB-231 cells which are commonly used to model late-stage breast cancer (Ramos et al. 2017). Rutin has been seen as an emerging inhibitor of c-Met which can control TNBC (Elsayed et al. 2017). Cancer cell growth inhibition has been observed by benzoic acid derivatives which act on histone deacetylases (Anantharaju et al. 2017). Targeting some of these kinds of known drug targets with the existing available inputs could be seen as the bombarding strategy to tackle this disease.

The protein receptors performed well with the ligands, viz. CDK2, EGFR, and PARP2 interacted with Taraxasteryl acetate with the minimum binding energies of − 11, − 11, and − 12.4 kcal/mole respectively and HER2, PARP1, and PTK6 complexed with Rutin with minimum binding energies of − 10.8, − 12.4, and − 10.4 kcal/mole respectively. On the other hand, ERK2 and PIM1 made strong interactions with FDA-approved drugs/drug analogues. As CDKs plays critical role in governing cell cycle transitions, cell division, and cell cycle control through CDK inhibition has been re-established as an attractive option in the development of targeted cancer therapy (Ding et al. 2020), use of Taraxasteryl acetate as an CDK inhibitor could be seen as alternative in future.

In case of EGFR, it enhances cell proliferation and survival and regulates a multitude of cell signaling pathways towards ontogenesis but, in cancer conditions, EGFR either gets mutated or becomes activated without any stimulus or due to continuous production of EGFR ligand, gets stimulated constantly. In both the conditions, inhibiting EGFR may contribute to alleviate the diseased condition. EGFR-targeting therapeutics have yielded modest, unpredictable, and variable (1.7–38.7%) responses (Baselga et al. 2005; Savage et al. 2017) in breast cancer.

During HER2-positive subtype, breast cancer cells express higher than normal levels of HER2 and about one out of five breast cancers are HER2-positive. These cancers tend to grow and spread faster, more aggressive than other breast cancers but are much more likely to respond to treatment with drugs that target the HER2 protein (Weiss 2020) which calls for the opportunity to inhibit this receptor with the help of Rutin as a good alternative.

In some cases, breast cancer occurred might also be due to poly [ADP-ribose] polymerase 1 (PARP1) gene dysregulation. PARP1 and PARP2 both regulate DNA repair and transcription and PARP inhibitors cause synthetic lethality in BRCA-mutated cancer cells from defective DNA damage repair (Dziadkowiec et al. 2016). Moreover, during the current investigation, these two receptors had bound strongly (with the minimum binding energy of − 12.4) with Taraxasteryl acetate and Rutin suggesting use of these two ligands in future pharmaceuticals with desired modifications.

Breast tumor kinase (BRK, also known as PTK6) abundant in several tumor types, including prostate, ovarian, and breast cancers, overexpressed in about 85% of all breast carcinomas, is one of the important targets. It displays low or no expression in the normal mammary gland. The expression of PTK6 is directly correlated with histological tumor grade (Tsui and Miller 2015) which makes it a desirable target to be inhibited by particular ligand compound.

In today’s fast-growing industrial world, consumer demand for natural products free of synthetic chemical adulterants is increasing. This paradigm shift of going synthetic to natural has brought pressure on the available natural resources. The vast majority of these essential plant products with varied therapeutic and other potentials have merited special interest (Wang et al. 2010). These compounds are mostly secondary metabolites in nature and are opt for commercial production. The compound, 2-hydroxy-4-methoxy benzaldehyde usually found in the roots of Hemidesmus indicus is one of the major aromatic volatile constituents among other phytochemicals present in the species. HMBA gives a typical aroma to the roots and is one of the under-utilized flavouring agents (Nagarajan et al. 2001). The compound confers many properties to the plant such as anti-microbial, anti-aflatoxigenic potency, anti-oxidant, etc. It has good water solubility and is a potent tyrosinase inhibitor (Murthy et al. 2006). MBALD, an aromatic compound found in roots of HI is an isomer of vanillin and has high demand in the food and flavouring industry (Rathi et al. 2017). Therefore, quantifying the compound holds great importance from both the commercial and the therapeutical point of view. An earlier report on the quantification of MBALD from dried roots was found to be 0.2638 mg/g of tissue (Prathibha Devi et al. 2016). Whereas, the amount of MBALD in fresh roots observed is slightly different and comes around 0.221 mg/g of tissue.

Conclusion

Finding a new drug for a particular disease is now becoming a little bit easy because of computational aids like virtual screening and molecular docking as the time required for screening the ligand compounds has reduced considerably. From the current investigation, it is clear that earlier reports on the anti-cancer activity of H. indicus hold significance as the results of the current investigation strongly support the inhibitory activity of the H. indicus compounds for six potential drug targets. Moreover, the software and web servers utilized during the study were freely available which makes the investigation a cost-cutting one. Further investigation on pharmacokinetics/pharmacodynamics properties, molecular simulation, in vitro experiments, and medical trials of the inhibitory compounds can lead to a potential drug in the future for tackling this serious problem.

HPLC performed for the current study is simple and cost-effective as it utilizes only two HPLC grade chemicals namely trifluoroacetic acid and methanol. The quantification assay of the sample is easy to perform and analyse. This technique can be used routinely to serve various purposes like identifying and quantifying samples and their quality control. The retention time to achieve the peak for the sample is observed to be higher than the earlier established reports maybe because of the variations in column dimensions and HPLC conditions. Variation in the length and diameter of the existing reverse phase column could be seen as the way to reduce the retention time which will facilitate more accuracy by repeating the number of runs.

References

  1. Addie M, Ballard P, Buttar D, et al. Discovery of 4-amino-N-[(1S)-1-(4-chlorophenyl)-3-hydroxypropyl]-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide (AZD5363), an orally bioavailable, potent inhibitor of Akt kinases. J Med Chem. 2013;56:2059–2073. doi: 10.1021/jm301762v. [DOI] [PubMed] [Google Scholar]
  2. Akhtar S, Benter IF. The role of epidermal growth factor receptor in diabetes-induced cardiac dysfunction. BioImpacts. 2013;3:5–9. doi: 10.5681/bi.2013.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Anantharaju P, Reddy B, Padukudru M, et al. Naturally occurring benzoic acid derivatives retard cancer cell growth by inhibiting histone deacetylases (HDAC) Cancer Biol Ther. 2017;18:492–504. doi: 10.1080/15384047.2017.1324374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ananthi R, Chandra N, Santhiya ST. Protective effect of Hemidesmus indicus R.Br. root extract against cisplatin-induced cytogenetic damage in mouse bone marrow cells. Genet Mol Biol. 2010;33:182–185. doi: 10.1590/S1415-47572010005000011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Banerji A, Banerji J, Das M, et al (2017) Some aspects of investigation of the indian medicinal plant Hemidesmus indicus R. Br.: chemical constituents and anti-diabetic activity. Available online www.jocpr.com. J Chem Pharm Res 2017:50–64
  6. Baselga J, Albanell J, Ruiz A, et al. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J Clin Oncol. 2005;23:5323–5333. doi: 10.1200/JCO.2005.08.326. [DOI] [PubMed] [Google Scholar]
  7. Baskar R, Lee KA, Yeo R, Yeoh K-W. Cancer and radiation therapy: current advances and future directions. Int J Med Sci. 2012;9:193–199. doi: 10.7150/ijms.3635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Boyle N, Banck M, James C, et al (2011) Open Babel: an open chemical toolbox - 1758–2946–3–33.pdf. 1–14 [DOI] [PMC free article] [PubMed]
  9. Chen P, Lee N, Hu W, et al. Spectrum and Degree of CDK Drug interactions predicts clinical performance. Mol Cancer Ther. 2016;15:2273–2281. doi: 10.1158/1535-7163.MCT-16-0300. [DOI] [PubMed] [Google Scholar]
  10. Chidambara Murthy KN, Rajasekaran T, Giridhar P, Ravishankar GA. Antioxidant property of Decalepis hamiltonii wight and arn. Indian J Exp Biol. 2006;44:832–837. [PubMed] [Google Scholar]
  11. Chohan T, Qayyum A, Rehman K, et al. An insight into the emerging role of cyclin-dependent kinase inhibitors as potential therapeutic agents for the treatment of advanced cancers. Biomed Pharmacother. 2018;107:1326–1341. doi: 10.1016/j.biopha.2018.08.116. [DOI] [PubMed] [Google Scholar]
  12. Cragg G, Newman D. Natural products: a continuing source of novel drug leads. Biochim Biophys Acta Gen Subj. 2013;1830:3670–3695. doi: 10.1016/j.bbagen.2013.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Dallakyan S, Olson A (2015) Small-molecule library screening by docking with PyRx. Methods Mol Biol:243–250 [DOI] [PubMed]
  14. Darekar R, Khetre A, Singh S, Damle M. HPTLC quantitation of 2-hydroxy-4-methoxybenzaldehyde in Hemidesmus indicus R.Br. root powder and extract. J Planar Chromatogr Mod TLC. 2009;22:453–456. doi: 10.1556/JPC.22.2009.6.13. [DOI] [Google Scholar]
  15. De S, Dermawan T, Stark R. EGF receptor uses SOS1 to drive constitutive activation of NFκB in cancer cells. Proc Natl Acad Sci USA. 2014;111:11721–11726. doi: 10.1073/pnas.1412390111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Ding L, Cao J, Lin W, et al. The roles of cyclin-dependent kinases in cell-cycle progression and therapeutic strategies in human breast cancer. Int J Mol Sci. 2020;21:1–28. doi: 10.3390/ijms21061960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Dulaney C, Marcrom S, Stanley J, Yang S. Poly(ADP-ribose) polymerase activity and inhibition in cancer. Semin Cell Dev Biol. 2017;63:144–153. doi: 10.1016/j.semcdb.2017.01.007. [DOI] [PubMed] [Google Scholar]
  18. Dziadkowiec K, Gasiorowska E, Nowak-Markwitz E, Jankowska A. PARP inhibitors: review of mechanisms of action and BRCA1/2 mutation targeting. Prz Menopauzalny. 2016;15:215–219. doi: 10.5114/pm.2016.65667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Elsayed H, Ebrahim H, Mohyeldin M, et al. Rutin as a novel c-Met Inhibitory Lead For The Control Of Triple Negative Breast Malignancies. Nutr Cancer. 2017;69:1256–1271. doi: 10.1080/01635581.2017.1367936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Eroles P, Bosch A, Alejandro Pérez-Fidalgo J, Lluch A. Molecular biology in breast cancer: Intrinsic subtypes and signaling pathways. Cancer Treat Rev. 2012;38:698–707. doi: 10.1016/j.ctrv.2011.11.005. [DOI] [PubMed] [Google Scholar]
  21. FDA.gov (2020) FDA grants accelerated approval to pembrolizumab forlocally recurrent unresectable or metastatic triple negativebreast cancer. https://www.fda.gov/news-events/press-announcements/fda-approves-new-therapy-triple-negative-breast-cancer-has-spread-not-responded-other-treatments. Accessed 27 Mar 2021
  22. Ferruzzi L, Turrini E, Burattini S, et al. Hemidesmus indicus induces apoptosis as well as differentiation in a human promyelocytic leukemic cell line. J Ethnopharmacol. 2013;147:84–91. doi: 10.1016/j.jep.2013.02.009. [DOI] [PubMed] [Google Scholar]
  23. Gam L-H. Breast cancer and protein biomarkers. World J Exp Med. 2012 doi: 10.5493/wjem.v2.i5.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Gao X, Liu X, Lu Y, et al. PIM1 is responsible for IL-6-induced breast cancer cell EMT and stemness via c-myc activation. Breast Cancer. 2019;26:663–671. doi: 10.1007/s12282-019-00966-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Hamdouchi C, Keyser H, Collins E, et al. The discovery of a new structural class of cyclin-dependent kinase inhibitors, aminoimidazo[1,2-a]pyridines. Mol Cancer Ther. 2004;3:1–9. doi: 10.1186/1476-4598-3-1. [DOI] [PubMed] [Google Scholar]
  26. Hillig RC, Sautier B, Schroeder J, et al. Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. Proc Natl Acad Sci USA. 2019;116:2551–2560. doi: 10.1073/pnas.1812963116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Holder S, Zemskova M, Zhang C, et al. Characterization of a potent and selective small-molecule inhibitor of the PIM1 kinase. Mol Cancer Ther. 2007;6:163–172. doi: 10.1158/1535-7163.MCT-06-0397. [DOI] [PubMed] [Google Scholar]
  28. Hwang SY, Park S, Kwon Y. Recent therapeutic trends and promising targets in triple negative breast cancer. Pharmacol Ther. 2019;199:30–57. doi: 10.1016/j.pharmthera.2019.02.006. [DOI] [PubMed] [Google Scholar]
  29. Ijaz M, Wang F, Shahbaz M, et al. The Role of Grb2 in cancer and peptides as Grb2 antagonists. Protein Pept Lett. 2017 doi: 10.2174/0929866525666171123213148. [DOI] [PubMed] [Google Scholar]
  30. Iqbal N, Iqbal N. Human epidermal growth factor receptor 2 (HER2) in cancers: overexpression and therapeutic implications. Mol Biol Int. 2014;2014:1–9. doi: 10.1155/2014/852748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Ishikawa T, Seto M, Banno H, et al. Design and synthesis of novel human epidermal growth factor receptor 2 (HER2)/epidermal growth factor receptor (EGFR) dual inhibitors bearing a pyrrolo[3,2-d]pyrimidine scaffold. J Med Chem. 2011;54:8030–8050. doi: 10.1021/jm2008634. [DOI] [PubMed] [Google Scholar]
  32. Isshiki Y, Kohchi Y, Iikura H, et al. Design and synthesis of novel allosteric MEK inhibitor CH4987655 as an orally available anticancer agent. Bioorg Med Chem Lett. 2011;21:1795–1801. doi: 10.1016/j.bmcl.2011.01.062. [DOI] [PubMed] [Google Scholar]
  33. Jackson G, Lee V. Insulin-like parameters growth in breast and factor cancer : binding with and insulin receptor correlation clinical. Clin Cancer Res. 1997;3:103–109. [PubMed] [Google Scholar]
  34. Kawlni L, Bora M, Upadhyay SN, et al. Pharmacological and therapeutic profile of anantamula (hemidesmus indicus (l.) R. Br.): a comprehensive review. Int J Ayurveda Pharma Res. 2017;5:49–57. [Google Scholar]
  35. Knight S, Adams N, Burgess J, et al. Discovery of GSK2126458, a highly potent inhibitor of PI3K and the mammalian target of rapamycin. ACS Med Chem Lett. 2010;1:39–43. doi: 10.1021/ml900028r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Manjulatha K, Saritha K, Setty H (2014) Phytochemistry, pharmacology and therapeutics of Hemidesmus indicus (L.) R. Br. Med Plants Phytochem Pharmacol Ther. 10.1017/CBO9781107415324.004
  37. More D, Mali P. A review article on species used as sarivain different regions of India: hemidesmus indicus, Ichnocrpus frutescens, Decalepis hamiltoni and Cryptolepis buchanani. Ayurlog Natl J Res Ayurved Sci. 2018;6:1–13. [Google Scholar]
  38. Murugan N, Mishra B, Paul B. Antioxidant, antibacterial and GC-MS analysis of methanol root extract of Hemidesmus indicus (L.) R. Br J Pharmacogn Phytochem. 2018;7:1669–1674. [Google Scholar]
  39. Nagarajan S, Jagan Mohan Rao L, Gurudutt N. Chemical composition of the volatiles of Hemidesmus indicus R. Br Flavour Fragr J. 2001;16:212–214. doi: 10.1002/ffj.985. [DOI] [Google Scholar]
  40. Ogura K, Shiga T, Yokochi M, et al. Solution structure of the Grb2 SH2 domain complexed with a high-affinity inhibitor. J Biomol NMR. 2008;42:197–207. doi: 10.1007/s10858-008-9272-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Okamoto K, Ikemori-Kawada M, Jestel A, et al. Distinct binding mode of multikinase inhibitor lenvatinib revealed by biochemical characterization. ACS Med Chem Lett. 2015;6:89–94. doi: 10.1021/ml500394m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Pal S, Moulik S, Dutta A, Chatterjee A. Extracellular matrix protein laminin induces matrix metalloproteinase-9 in human breast cancer cell line MCF-7. Cancer Microenviron. 2014;7:71–78. doi: 10.1007/s12307-014-0146-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Papadimitriou M, Mountzios G, Papadimitriou CA. The role of PARP inhibition in triple-negative breast cancer: unraveling the wide spectrum of synthetic lethality. Cancer Treat Rev. 2018;67:34–44. doi: 10.1016/j.ctrv.2018.04.010. [DOI] [PubMed] [Google Scholar]
  44. Park H, Ito K, Olcott W, et al. PTK6 inhibition promotes apoptosis of Lapatinib-resistant Her2+ breast cancer cells by inducing Bim. Breast Cancer Res. 2015;17:1–13. doi: 10.1186/s13058-015-0594-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Prathibha Devi P, Devi Cherku P, Rama Devi B, et al. Estimation of 2-hydroxy-4-methoxybenzaldehyde, lupeol and other unreported compounds in an elite ecotype of Hemidesmus indicus (L.) R. BR An Int J Ann Phytomed. 2016;5:51–58. [Google Scholar]
  46. Ramos PAB, Guerra ÂR, Guerreiro O, et al (2017) Antiproliferative effects of Cynara cardunculus L. Var. altilis (DC) lipophilic extracts. Int J Mol Sci. 10.3390/ijms18010063 [DOI] [PMC free article] [PubMed]
  47. Rathi N, Harwalkar K, Jayashree V, et al (2017) 2-hydroxy-4-methoxybenzaldehyde, an astounding food flavoring metabolite: a review. Asian J Pharm Clin Res 10:105–110. 10.22159/ajpcr.2017.v10i10.19729
  48. Russo J, Russo H. The role of estrogen in the initiation of breast cancer. J Steroid Biochem Mol Biol. 2006;102:89–96. doi: 10.1016/j.jsbmb.2006.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Savage P, Blanchet-Cohen A, Revil T, et al. A targetable EGFR-dependent tumor-initiating program in breast cancer. Cell Rep. 2017;21:1140–1149. doi: 10.1016/j.celrep.2017.10.015. [DOI] [PubMed] [Google Scholar]
  50. Sharma HP, Plants M, Analysis G (2017) GC-MS analysis of a medicinal plant: Hemidesmus indicus. Bot Reena Keywords : 53–55
  51. Song M, Bode AM, Dong Z, Lee MH. AKt as a therapeutic target for cancer. Cancer Res. 2019;79:1019–1031. doi: 10.1158/0008-5472.CAN-18-2738. [DOI] [PubMed] [Google Scholar]
  52. Srabovic N, Mujagic Z, Mujanovic-Mustedanagic J, et al. Vascular endothelial growth factor receptor-1 expression in breast cancer and its correlation to vascular endothelial growth factor A. Int J Breast Cancer. 2013;2013:1–6. doi: 10.1155/2013/746749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Stamos J, Sliwkowski MX, Eigenbrot C. Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J Biol Chem. 2002;277:46265–46272. doi: 10.1074/jbc.M207135200. [DOI] [PubMed] [Google Scholar]
  54. Swathi S, Rani R, Amareshwari P, Venkatesh K. Phytochemical and pharmacological benefits of Hemidesmus indicus: an updated review. J Pharmacogn Phytochem. 2019;8:256–262. [Google Scholar]
  55. Thabrew M, Mitry R, Morsy A, Hughes D. Cytotoxic effects of a decoction of Nigella sativa, Hemidesmus indicus and Smilax glabra on human hepatoma HepG2 cells. Life Sci. 2005;77:1319–1330. doi: 10.1016/j.lfs.2005.01.022. [DOI] [PubMed] [Google Scholar]
  56. Thakur M, Birudukota S, Swaminathan S, et al. Co-crystal structures of PTK6: With Dasatinib at 2.24 Å, with novel imidazo[1,2-a]pyrazin-8-amine derivative inhibitor at 1.70 Å resolution. Biochem Biophys Res Commun. 2017;482:1289–1295. doi: 10.1016/j.bbrc.2016.12.030. [DOI] [PubMed] [Google Scholar]
  57. Thorsell A-G, Ekblad T, Karlberg T, et al. Structural basis for potency and promiscuity in poly(ADP-ribose) polymerase (PARP) and tankyrase inhibitors. J Med Chem. 2017;60:1262–1271. doi: 10.1021/acs.jmedchem.6b00990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Tresaugues L, Roos A, Arrowsmith C, et al (2013) Crystal structure of VEGFR1 in complex with N-(4-Chlorophenyl)-2-((pyridin-4-ylmethyl) amino) benzamide. RCSB PDB
  59. Tsui T, Miller W. Cancer-associated mutations in breast tumor kinase/PTK6 differentially affect enzyme activity and substrate recognition. Biochemistry. 2015;54:3173–3182. doi: 10.1021/acs.biochem.5b00303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Tucker J, Klein T, Breed J, et al. Structural insights into FGFR kinase isoform selectivity: diverse binding modes of AZD4547 and ponatinib in complex with FGFR1 and FGFR4. Structure. 2014;22:1764–1774. doi: 10.1016/j.str.2014.09.019. [DOI] [PubMed] [Google Scholar]
  61. Vajdos F, Hoth L, Geoghegan K, et al. The 2.0 A crystal structure of the ERalpha ligand-binding domain complexed with lasofoxifene. Protein Sci. 2007;16:897–905. doi: 10.1110/ps.062729207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Version A, Morris GM, Goodsell DS, et al (2014) AutoDock Version 4.2. 1–69
  63. Wang J, Liu H, Zhao J, et al. Antimicrobial and antioxidant activities of the root bark essential oil of Periploca sepium and its main component 2-hydroxy-4-methoxybenzaldehyde. Molecules. 2010;15:5807–5817. doi: 10.3390/molecules15085807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Ward R, Colclough N, Challinor M, et al. Structure-guided design of highly selective and potent covalent inhibitors of ERK1/2. J Med Chem. 2015;58:4790–4801. doi: 10.1021/acs.jmedchem.5b00466. [DOI] [PubMed] [Google Scholar]
  65. Weiss MC (2020) Molecular subtypes of breast cancer. https://www.breastcancer.org/. Accessed 10 Jul 2019
  66. Wu J, Li W, Craddock B, et al. Small-molecule inhibition and activation-loop trans-phosphorylation of the IGF1 receptor. EMBO J. 2008;27:1985–1994. doi: 10.1038/emboj.2008.116. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from 3 Biotech are provided here courtesy of Springer

RESOURCES