Skip to main content

Antiproliferative effect of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues on IL-6 mediated STAT3 and role of the apoptotic pathway in albino Wistar rats of ethyl carbamate-induced lung carcinoma: In-silico, In-vitro, and In-vivo study

Abstract

Lung cancer (LC) ranks second most prevalent cancer in females after breast cancer and second in males after prostate cancer. Based on the GLOBOCAN 2020 report, India represented 5.9% of LC cases and 8.1% of deaths caused by the disease. Several clinical studies have shown that LC occurs because of biological and morphological abnormalities and the involvement of altered level of antioxidants, cytokines, and apoptotic markers. In the present study, we explored the antiproliferative activity of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues against LC using in-vitro, in-silico, and in-vivo models. In-vitro screening against A549 cells revealed compounds 9B (8-methoxy-5-(3,4,5-trimethoxyphenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine) and 12B (5-(4-chlorophenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine) as potential pyrimidine analogues against LC. Compounds 9B and 12B were docked with different molecular targets IL-6, Cyt-C, Caspase9, and Caspase3 using AutoDock Vina 4.1 to evaluate the binding affinity. Subsequently, in-vivo studies were conducted in albino Wistar rats through ethyl-carbamate (EC)- induced LC. 9B and 12B imparted significant effects on physiological (weight variation), and biochemical (anti-oxidant [TBAR’s, SOD, ProC, and GSH), lipid (TC, TG, LDL, VLDL, and HDL)], and cytokine (IL-2, IL-6, IL-10, and IL-1β) markers in EC-induced LC in albino Wistar rats. Morphological examination (SEM and H&E) and western blotting (IL-6, STAT3, Cyt-C, BAX, Bcl-2, Caspase3, and caspase9) showed that compounds 9B and 12B had antiproliferative effects. Accordingly, from the in-vitro, in-silico, and in-vivo experimental findings, we concluded that 9B and 12B have significant antiproliferative potential and are potential candidates for further evaluation to meet the requirements of investigation of new drug application.

Introduction

Lung cancer (LC) is the major cause of cancer incidence and mortality [1] and is the second leading cancer in males and females [2]. In India, LC accounts for  5.9% (in females) and  8.5% (in males) of all cancer-related deaths [3, 4]. Males have a higher incidence and mortality than females globally [5, 6]. Therefore, identifying or synthesizing new chemical moieties that target molecular pathways is a key step in the development of new treatment approaches. Researchers have reported that thiazolo[3,2-a]pyrimidine is a group of heterocyclic compounds known to have various significant biological properties such as antimicrobial, antipsychotic, anti-inflammatory, anti-parkinson’s, antidepressant, and anti-HIV [7, 8].

These molecules have increased the considerable scope of the anticancer activity of thiazolo[3,2-a]pyrimidine-related compounds. Since the last decade, broad-spectrum standard anti-cancerous drugs such as camptothecin, doxorubicin, paclitaxel, and 5-fluorouracil (5-FU) have been extensively used for multiple treatments. 5-FU or 5-fluro-2, 4-pyrimidinedione is a chemotherapeutic drug frequently delivered either by a single medication or in combination with other therapeutic regimens [9,10,11,12]. 5-FU is an anticancer antimetabolite drug primarily derived from thiazolo-pyrimidines that comprise a thiazole and a pyrimidine ring, which inhibit the proliferation of cancer cells [13, 14]. This has led to considerable hope for the development of new chemical moieties with pyrimidine ring.

Thiazolopyrimidine ring systems possess antitumor activity. The previously reported significance of such synthons has generated interest in exploiting this valuable structure in the design and synthesis of new thiazolo-pyrimidines analogues as antitumor agents [15].

Moreover, a series of these molecules had already been synthesized in our laboratory [16]. The structures of the synthesized products were confirmed using spectroscopic techniques such as mass and NMR spectrometry [16].

Therefore, in continuation of our previous work, this study has been designed to explore the anticancer potential of the synthesized thiazolo[3,2-a]pyrimidine analogues (9B (8-methoxy-5-(3,4,5-trimethoxyphenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine), Mol. wt. 422.50 and 12B (5-(4-chlorophenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine), Mol. wt. 336.84) against LC.

Materials and methods

Drugs and reagents

EC was procured from the Tokyo chemical industry, Japan. IL-2 (RAB0288) and IL-6 (RAB0311) ELISA kits were purchased from Sigma-Aldrich, Bengaluru, India. IL-1β (GX3930E1) and IL-10 (GX8140E1) experimental ELISA kits were obtained from Genetix Biotech Asia Pvt. Ltd, India. Caspase-9 (ITER0804) and Caspase-3 (KHO1091) ELISA kits were purchased from Invitrogen and Thermo Fisher, India, respectively. All primary antibodies, including IL-6 (SC-32,296), STAT-3 (SC8019), BAX (SC23959), Bcl-2 (SC7382), Cyt-C (SC13561), Caspase-9 (SC73548), Caspase-3 (SC56052), β-actin (SC517582), were procured from Santa Cruz Biotechnology, USA. All other remaining chemicals and solvents of molecular grade were purchased from Hi-Media laboratories, Sigma-Aldrich, India.

In-vitro cell cytotoxicity assay

To determine the cytotoxicity potential of the synthesized compounds. All samples were sent to the ACTREC, Tata Memorial Centre, Mumbai, India. They followed the standard protocol for sample analysis of the synthesized compounds. Adriamycin (ADR) was selected as a standard at 10, 20, 40, and 80 µg/mL to screen for indeno [1, 2-d] thiazolo [3,2-a] pyrimidine analogues. GI50 growth inhibition of 50% was calculated using the [(Ti − Tz)/(C − Tz)] ×100% formula, where C, controlled growth; Tz, time zero growth; and Ti, test growth at four concentration levels in the presence of the drug [17]. The GI50 value of the compounds was considered to demonstrate antiproliferative activity against A549 cells [18,19,20].

In-silico molecular docking analysis

Docking study of the synthesized compounds were performed using IL-6, Cytochrome-C (Cyt-C), Caspase 9, and Caspase 3. The molecular structure of the ligands 9B and 12B was generated using ChemDraw Profession 16.0. Additionally, we utilized the NCBI (https://www.ncbi.nlm.nih.gov/protein) and RCSB (https://www.rcsb.org) databases as sources to obtain the desired protein targets: IL-6 (1ALU), Cyt-C (5TY3), caspase 9 (2AR9), and caspase 3 (2XYG). 3D structures of different target proteins were prepared in PDB format [21] using AutoDock Vina 4.1. DS visualizing software (Discovery Studio Visualizer, 2021) was used to remove specific protein targets, water molecules [22], hetatms (hetero atoms) from the co-crystal structure, and active pocket sites of protein by utilizing the “Active site determination from PDB site record” function. To dock the ligand with desire proteins (kcal/mol), we used AutoDock Vina 4.1 [22,23,24,25]. AutoDock Vina 4.1 [25] calculated the predicted binding energy and expressed it in kcal/mol [26]. The interaction between these complexes was analyzed using the DS visualizer, and the potential hydrogen bonds, hydrophobic amino acid interactions in close proximity were also examined.

Acute toxicity study

It is necessary to investigate the safety and efficacy of the synthesized compounds in vivo, which was performed as per the guidelines of the Organization for Economic Cooperation and Development guidelines 423 (OECD 423). The synthesized compounds were orally administered at doses of 5, 10, and 15 mg/kg body weight to albino Wistar rats for 15 days (n = 5), and the animals were observed every day for any toxic signs at various doses. Morphological and oxidative stress parameters were used to determine dose-related toxicity.

Pharmacokinetic study using high-performance liquid chromatography (HPLC)

9B and 12B compounds were orally administered at 10 mg/kg (p.o. dose obtained from acute toxicity studies) to rats, and blood was collected from the latetal tail vein at 0.083, 0.25, 0.5, 1, 2, 4, 8, 12, 16, 24, and 48 h [27]. After collection, blood was centrifuged and plasma was separated, which was kept at -20 °C for further HPLC determination. Chromatographic separations were performed using a Shimadzu Prominence-I LC-2030 Plus equipped with HPLC and a photodiode array (PDA) detector. The mobile phase consisted of a gradient elution of 2:8, water: methanol, and a continuous flow rate of 1 mL/min in an RP-C18 column (5.0 µ particle sizes, 4.6 mm internal diameter, and 250 mm length) with different λmax of 310 and 320 nm, respectively, at 40 °C throughout the experiment. The column was washed using an elution solvent (water: methanol = 50:50) after each run, and the final output was analyzed using WinNonlin version 1.5.3 software.

Animal and study design

Six-week-old experimental male albino Wistar rats (100–120 g) were procured from the animal house facility of Babu Banarasi Das Northern India Institute of Technology Lucknow and approved by the Institutional Animal Ethical Committee (CPCSEA No. 809/PO/Re/S/03/CPCSEA) with approval no. BBDNIIT/IAEC/APR/2022/06. The animal welfare protocol and experiments were performed as per the CPCSEA guidelines for laboratory animals and ethics, Department of Animal Welfare, Government of India. They were randomly distributed into five groups of eight animals each. The experimental rats were acclimated under standard research laboratory conditions with free access to standard rat chow and water ad libitum. Group 1 (NC, normal control group): 0.25% CMC (2 mL/kg, p. o.), Group 2 (CC, carcinogen control/EC-exposed group): EC (0.375 mg/kg, four i.p. injections within a gap of three weeks between each dose over twelve weeks period), Group 3 (PC, positive control group): EC + 5-FU (10 mg/kg, i.p. for 15 days after EC induced LC), Group 4 (9B): EC + Compound 9B (10 mg/kg, p.o. for 15 days after EC induced LC), and Group 5 (12B): EC + Compound 12B (10 mg/kg, p.o. for 15 days after the induction of LC). At the end of the treatment, the animals were euthanized by cervical decapitation after anesthesia using a combination of diazepam and ketamine hydrochloride (5 mg/kg, and 100 mg/kg, i.m. respectively). The lung was excised immediately, rinsed in ice-cold saline, and stored at -80 °C for further study. Serum was collected and stored for further analysis.

In-vivo antineoplastic studies of 9B and 12B

Estimation of the various physiological parameters

Weight variation

Change in body weight (gm) was measured on the initial and final day of the experiment, and percent (%) weight gain/loss was calculated.

Measurement of the serum lipid profile

Serum total cholesterol (TC), triglycerides (TG), and high-density lipoprotein (HDL) levels were measured by using lipid profile kits (Agappe Diagnostic Ltd., Kerala, India). In brief, the assay was performed according to the manufacturer’s instructions. Low-density lipoprotein (LDL) and very-low-density lipoprotein (VLDL) levels were estimated and calculated using Friedewald’s formula [24].

Measurement of the oxidative changes

Biochemical parameters such as protein carbonyl (PC), superoxide dismutase (SOD), glutathione (GSH), and thiobarbituric acid reactive substances (TBARs) were estimated in 10% lung tissue homogenate using our previously standardized laboratory protocols [17, 28, 29].

Cytokine estimation

The pro-inflammatory cytokines IL-2, IL-6, IL-10, and IL-1β were assayed using commercially available kits according to the manufacturer’s instructions.

Histopathological analysis

Histopathological studies were conducted to determine intracellular changes in lung tissue. Tissue samples were stained with H&E (haemotoxyline and eosin) using a methodology established previously at our laboratory for histological analysis and observed under an optical microscope (Steindroff N-120, 40X).

Scanning electron microscopy analysis

Lung tissue samples of 2–4 mm thickness were sectioned, and the samples were prepared by fixation, washing, post-fixation, and dehydration according to the standardized procedures of our laboratory. Finally, the samples were mounted on aluminium stubs with adhesive tape and examined for morphological changes using SEM (JEOL JSM-6490LV).

Western blot analysis

Protein expression levels of IL-6 (pro-inflammatory), STAT-3 (pro-inflammatory), Bax (pro-apoptotic), Bcl-2 (anti-apoptotic), Cyt-C (pro-apoptotic), Caspase-9, and Caspase-3 (pro-apoptotic/executor caspase) were assessed by immunoblotting. Proteins were electrophoresed on sodium dodecyl sulphate polyacrylamide gel (SDS-PAGE) and probed to detect molecules of interest in the mixture. The membrane was developed with enhanced chemiluminescence ECL, and images were obtained using Chemidoc XRS+ (Bio-rad).

Result

In-vitro cell cytotoxicity assay

Thiazolo [3, 2-a] pyrimidine-containing compound series (1B-15B) were analzsed against adriamycin (ADR) using the A549 cell line for cytotoxic estimation, as depicted in Fig. 1. Based on the GI50 value of the series, 9B (< 20) and 12B (< 10) were found to be the most potential compounds (Fig. 1 (III)). Consequently, 9B and 12B were considered for further evaluation.

Fig. 1
figure 1

(I) Structure of 9B (8-methoxy-5-(3,4,5-tri-methoxyphenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine), (II) 12B (5-(4-chlorophenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine), and (III) SRB assay of (1B-15B and ADR) on human lung cancer cell line A549, graph plot between drug concentration (µg/ml) vs. % control growth

In-silico molecular docking analysis

We performed docking studies of the synthesized compounds with the molecular targets IL-6 (PDB: 1ALU), Cyt-C (PDB: 5TY3), Caspase 9 (PDB: 2AR9), and Caspase 3 (PDB: 2XYG) using AutoDock Vina 4.1. The binding affinities (kcal/mole) and the number of probable hydrogen bonds was evaluated. The binding affinities for 9B (-6.1, -6.3, -7.4, and − 7.2), and 12B (-7.0, -6.2, -7.5, and − 7.3) were found to be with IL-6, Cyt-C, Caspase 9, and Caspase 3, respectively (Fig. 2 (I and II)) which indicates the amino-acid interactions with both ligands. Hydrongen bonds involvements with 9B and 12B were showed in Table 1.

Fig. 2
figure 2

Molecular docking of 9B and 12B. (I) 3D and 2D molecular docking images of 9B with (A and A’) IL-6 (PDB: 1ALU), (B and B’) Cyt-C (PDB: 5TY3), (C and C’) Caspase 9 (PDB: 2AR9) and (D and D’) Caspase 3 (PDB: 2XYG) and (II) 3D and 2D docking images of 12B with (E and E’) IL-6 (PDB: 1ALU), (F and F’) Cyt-C (PDB: 5TY3), (G and G’) Caspase 9 (PDB: 2AR9) and (H and H’) Caspase 3 (PDB: 2XYG). Comparative studies were performed using AutoDock Vina 4.1

Table 1 Binding interaction of compound 9B and 12B with IL-6 (PDB: 1ALU), Cyt-C (PDB: 5TY3), Caspase 9 (PDB: 2AR9), and Caspase 3 (PDB: 2XYG). Comparative studies were performed using AutoDock Vina 4.1

Acute toxicity study

Both compounds were orally administrated (9B and 12B) for 15 days at different doses, such as 5, 10, and 15 mg/kg, and showed no toxic effect. No mortality was observed in the animals during this study. Histological examination of lung, liver, and kidney tissue, we could not find any toxic effect. Moreover, antioxidant marker estimation showed no change in SOD, CAT, GSH, and TBARs values. Based on our above findings, we proceeded with a 10 mg/kg dose, which is similar to 5-FU for in-vivo anti-proliferative activity evaluation (Supplementary Figs. 1 and 2).

Pharmacokinetic study

Pharmacokinetic profiling of 9B and 12B at 10 mg/kg was evaluated using HPLC. 1 µg/ml − 100 µg/ml gave a 0.9889 correlation coefficient for 9B and 0.987 for 12B. The retention time of 9B was 14.931 and that of 12B was 7.394. The plasma concentration-time profiles of 9B and 12B were analyzed by using GraphPad Prism 5.02. 12B had a higher plasma distribution compared with 9B at the same time (Fig. 3 (I) 9B (A, and B) and (II) 12B (A, and B)). The T1/2- 7.95, 11.49; Tmax (h): 4.00, 4.00; Cmax (µg/ mL)- 30.84 ± 1.59, 40.15 ± 2.06; AUMC (µg.h2/ mL)- 3028.82, 4557.60; MRT (h)- 10.59 ± 0.99, 10.44 ± 1.08; CL (h)- 0.003, 0.006; AUC0−∞ (µg.h/mL)- 287.60, 436.49 for 9B and 12B, respectively. 12B showed a good biological availability as compared to 9B (Table 2).

Fig. 3
figure 3

A pharmacokinetic study of 9B and 12B using HPLC; (I) Pharmacokinetic profiling of 9B (A) Chromatogram of 9B (RT 14.931 min) through HPLC, (B) Plasma drug concentrations at various time points after oral administration of 9B in albino Wistar rats (II) Pharmacokinetic profiling of 12B (A) Chromatogram of 12B (RT 7.394 min) through HPLC, (B) Plasma drug concentrations at various time points after oral administration of 12B in albino Wistar rats

Table 2 Pharmacokinetic study of 9B and 12B in rat plasma

In-vivo antineoplastic studies of 9B and 12B

Estimation of the various physiological parameters

Weight variation

Weight variation in rats against EC showed significant weight loss (-13.31 ± 3.15 gm). NC, PC, 9B, and 12B rats exhibited increased weight compared with the CC group (10.42 ± 1.72, 7.21 ± 1.75, 6.01 ± 1.08, and 17.52 ± 1.33gm, respectively) (Fig. 4 (I)).

Measurement of the serum lipid profile

Figure 4 (II) indicates the cancer risk associated with the administration of EC, which further increased the levels of TC, TG, and LDL as well as the decreased levels of HDL and VLDL in the CC group. The EC-exposed group was more associated with a significant increase in TC and LDL levels, as well as a decrease in HDL levels (Fig. 4 (II)). 9B and 12B restored the alteration of the lipid profile in the treatment groups.

Measurement of the oxidative changes

The antioxidant potential of both 9B and 12B was estimated using the findings of SOD, GSH, ProC, and TBARS. Decreased levels of GSH and SOD and increased levels of ProC and TBARS exposed the carcinogenic effect of EC on experimental animals. From the therapeutic point of view, both compounds support the antiproliferative potential by restoring the altered levels of SOD, GSH, ProC, and TBARs (Fig. 4 (III)).

Cytokine estimation

Increased levels of cytokine IL-1β, IL-2, IL-6, and IL-10 were significantly decreased after treatment with 9B and 12B, as depicted in Fig. 4 (IV).

Fig. 4
figure 4

Physiological and biochemical changes in EC-induced lung carcinogenic rats after oral administration of 9B and 12B at 10 mg/kg for 15 days. (I) Changes in body weight of experimental animals, (II) Effects of 9B and 12B, on lipid profile including TG, TC, HDL, LDL, and VLDL. (III) Effects of 9B and 12B, on oxidative stress estimation of SOD (superoxide dismutase), PC (Protein carbonyls), TBARS (Thiobarbituric acid reactive substances), and GSH (Glutathione), and (IV) Effects of 9B and 12B, on pro-inflammatory cytokines such as IL-2, IL-6, IL-10, and IL-1β. Each value is represented here as mean ± SD (n = 6/group), statistically significant differences were observed between lung carcinogenic (CC) and test groups (9B and 12B) [one-way ANOVA followed by Bonferroni multiple comparison tests for biochemical estimation, statistical significance differences were considered concerning control (***p < 0.001, **p < 0.01 and *p < 0.05)]. Where, NC: Normal Control, CC: Carcinogen Control, PC: Positive Control, 9B (10 mg/kg), 12B (10 mg/kg)

Histopathological analysis

In histological analysis, the number of carcinogenic nodule formations in the lungs along with structural differences, blockage of alveoli, and reduced size of alveoli confirmed the carcinogenic effect of EC. The EC effect was found to be more prominent in the CC group, which was further reduced in different groups after the treatments. The histopathological recovery of lung tissue architectures explored the therapeutic effects of 9B and 12B along with 5-FU, as shown in Fig. 5 [I].

Fig. 5
figure 5

Morphological changes evaluation in EC-induced LC. [I] Histopathological studies of lung tissue sections of albino Wistar rats, each group analyzed under a light microscope (40X, scale bar 50 μm). Abbreviation: AC- Alveolar cell, Ad- Alveolar duct, IC- Inflammatory cell, Am- Alveolar macrophages, MA- Mucinous adenocarcinoma, CC- Carcinoma cells where (A) Normal Control [NC]; (B) Carcinogen Control [CC]; (C) Positive Control [PC]; (D) 9B; (E) 12B [II] Scanning electron microscopic photomicrography of lung tissue (1000X) in albino Wistar rats of each group, where (A) Normal Control [NC]; (B) Carcinogen Control [CC]; (C) Positive Control [PC]; (D) 9B; (E) 12B

SEM analysis

In SEM, NC animals represent uniform tissue structure with the presence of alveolar sacks, which were perforated through alveolar ducts to provide air supply to the lungs. Pentagonal-shaped annuli containing alveolar sacs were reduced after EC administration in CC animals, and recovered after treatment with thiazolo[3,2-a]pyrimidine-containing compounds (9B and 12B), as shown in Fig. 5 [II].

Western blot analysis

Administration of EC enhanced the expression of markers that promote inflammation and prevent apoptosis (IL-6, STAT-3, and Bcl-2). In addition, EC decreased the expression of pro-apoptotic markers (Bax and Cyt-c) and caspases (Caspase-3 and Caspase-9) in the CC group. Remarkably, following treatment with 9B and 12B, the expression of IL-6, STAT-3, and Bcl-2 decreased, whereas the expression of caspases increased. Bax and Cyt-C also showed the higher expression with 12B treatment than with 9B treatment in the CC group (Fig. 6).

Fig. 6
figure 6

Immunoblotting assay for protein expression in lung samples; Protein expression levels of IL-6, STAT-3, Bax, Bcl-2, Cyt-C, Caspase-9, and Caspase-3 after treatment with 9B and 12B and relative band density of NC, CC, PC, 9B, and 12B with different interests of protein. Data are represented as mean ± SD. Statistically significant differences were observed between CC and treatment groups (PC, 9B, and 12B) using one-way-ANOVA followed by Bonferroni multiple comparison test (***p < 0.001, **p < 0.01, *p < 0.05)]

Discussion

The most prevalent and deadly solid cancer is LC due to uncontrolled respiratory exponentiation [30]. 5-FU is a well-known anti-cancer drug which consisting a thiazole, and a pyrimidine ring, and is considered as standard for the treatment of different solid tumors. Therefore, we synthesized a new series of synthetic molecules with thiazolo[3,2-a]pyrimidine ring by adopting rational drug design strategies [16]. All synthesized thiazolo[3,2-a]pyrimidine analogues were screened against A549 cell line for their cytotoxic potential. In the series of 15 compounds, 9B and 12B exhibited the best cytotoxic effect in the SRB assay among the entire synthesized compounds against the A549 cell line. Similar study conduted by keshari et al., showed cytotoxic potential of the thiazolo[3,2-a]pyrimidine analogues against Hep-G2 cell line. In addition, a molecular docking study showed a good binding affinity and better stability with IL-6, Cyt-C, caspase-9 and caspase-3 [27,28,29,30,31] as well as hydrogen bondings with both compounds. Moreover, 9B and 12B compounds have the strong binding affinity with the caspase 9 (-7.5 kcal/mol and − 7.3 kcal/mol) and caspase 3 (-7.4 kcal/mol and − 7.2 kcal/mol) respectively. Hydrogen bonds involvement with caspases suggesting strong binding activity with them. Caspases play an important role in initiating and executing apoptosis [32, 33]. Accordingly, it might be a promising antiproliferative activity containing compounds.

The acute toxicity studies of thiazolo[3,2-a]pyrimidines containing 9B and 12B compounds showed no toxic effect in the histological and oxidative scrutinization at 5, 10, and 15 mg/kg dose. Therefore, we selected a dose of 10 mg/kg (p.o.) for further assessments similar to 5-FU [10, 11, 13].

The pharmacokinetic studies demonstrated a greater plasma volume distribution of 9B and 12B, with 12B demonstrating better oral bioavailability than 9B. From the findings of hydrogen bond donors, the molecular weight and binding affinity of both 9B and 12B compounds can be further used as good orally absorbed compounds [16].

The significant cytotoxic potential, binding affinity, safety profile, and oral bioavailability of 9B and 12B paved the way for further in-vivo estimations against EC-induced LC in albino Wistar rats. Higher weight loss in the CC group was observed because of cancer cachexia and loss of adipose tissue [34, 35]. At a dose of 10 mg/kg, 12B demonstrated better efficacy in weight restoration than 9B.

Various empirical research has indicated that changes in lipid metabolism are considered to be a characteristic feature of carcinogenesis [36]. Cancer cells required more energy for their proliferation by denovo reactivated lipid synthesis. Additionally, overexpression of multifunctional enzyme fatty acid synthases, play key role in poor prognosis of LC due to significantly increased cancer aggressiveness [37]. Several studies proven that LDL concentration is a prognostic factor in small cell lung cancer. High TG concentration and lower HDL created a predisposition to higher LC incidence [36, 38]. Additionally, Increased levels of TC and LDL are connected with the cancer growth [39] and potentially associated with increased cellular proliferation [29, 40]. On the other hand, HDL involvement in the reverse transport process of cholesterol facilitates the removal of excess cholesterol levels from peripheral tissues [41]. Additionally, an inverse relationship between cancer and HDL was demonstrated in LC. Several studies have shown that increased HDL levels may be associated with anti-inflammatory effects through cytokine protection and leukocyte adhesion; therefore decreased HDL may reduce inflammation and antioxidant activity as well as cancer development [42, 43]. Our results showing TC, TG, and LDL level in EC-treated animals are consistent with the aforementioned research.

Previous studies have reported that EC is metabolized to vinyl carbamate, followed by the formation of epoxide, which interacts with nucleic acids to trigger ROS formation and DNA damage [44]. The resultant ROS generation and DNA damage lay the foundation for the development and progression of cancer [44], which was evident with increased levels of ProC and TBARs in the CC group. Concomitant administration of 9B and 12B resulted in decreased levels of ProC and TBARs, which could be attributed to the presence of electronegative substitution available on the thiazolo[3,2-a]pyrimidine ring [45]. The antioxidant potential of 9B and 12B was further affirmed through the restoration of the enzymatic activity of SOD and GSH. The free radical scavenging potential of 9B and 12B can be attributed to the thiazolo[3,2-a]pyrimidine ring, which is consistent with previous reports [8].

The efficacy of 9B and 12B was further scrutinized through pro-inflammatory markers (IL-2, IL-6, IL-10, and IL-1β). EC administration was evident in the upsurge in the levels of inflammatory cytokines [35, 46, 47]. These findings suggest that EC-induces inflammatory signaling associated with anti-apoptotic mechanisms, which is corroborated by previous reports [34].

Histopathological analysis of CC revealed the loss of tissue architecture, alveolar damage, elevated mucinous adenocarcinoma cells, and inflammatory cells to support the proliferation of the lung alveolar epithelium. However, treatment with 9B and 12B showed a healthy architecture of the bronchioles, and alveoli, reduced lung proliferation and restoration of tissue architecture.

In morphological analysis of intact organs, the existence of tumor formation was higher in the EC-induce model, which was further reduce after treatment with 9B and 12B. SEM analysis revealed the presence of a capillary network interlining the fibrous structure of the alveolar wall in NC [34]. However, the CC group accounted for the parenchymal compaction with reducing the size of annuli and loss of preferred region [48]. The 12B treated group demonstrated better efficacy than 9B for restoration of tissue architectures.

The administration of 9B and 12B curbed inflammatory markers and restored the apoptotic machinery, evident through the upregulation of caspase-3 and caspase-9 levels. The findings reported above clearly demonstrate the anticancer potential of 9B and 12B against EC-induced LC in albino Wistar rats. However, to investigate the possible mechanism underlying the biological activity of 9B and 12B, we scrutinized the expression of various apoptotic markers using immunoblotting.

Pro-inflammatory IL-6 regulates STAT3 to promote Bcl-2 activation and prevent Cyt-C release [49, 50]. The release of Cyt-C is an important step in the intrinsic apoptotic pathway, where caspase 3 and 9 are key regulators [51]. In contrast, caspase-9 is a key step in intrinsic apoptosis [51]. In view of the above, we examined the expression of IL-6, STAT3, Bcl-2, Bax, Cyt-C, caspase-3, and caspase-9 in lung tissue samples.

In the CC group, we observed increased expression of IL-6, STAT3, and Bcl-2 with decreased expression of Cyt-C, caspase-9, and caspase-3. IL-6 is a potent cytokine that regulates the immune defense process of the body. IL-6 is a crucial link between inflammation and cancer. Higher IL-6 concentrations are indicated by secretion through tumor-associated macrophages or tumor cells due to malignancy-induced chronic stress. IL-6 release involves different tyrosine kinase pathways or transcription 3 activation. STAT3 deficiency indicates tumor growth, and IL-6/STAT3 has a crucial role in carcinogenesis [52]. Oral administration of 9B and 12B significantly downregulates IL-6, STAT3, and Bcl-2 expression, indicating the initiation of the apoptotic mechanism. Previous studies have implying a family of cysteine proteases, formally known as caspases, is associated with the process of apoptosis induction [53]. In the continuation of this study, 9B and 12B upregulate the expression of Cyt-C, caspase-9, and caspase-3, which affirms that both the analogues exert anticancer activity against LC by regulating the intrinsic apoptotic mechanism. This could serve as potential lead molecules for the development of LC drugs in the future for clinical and therapeutic research.

Conclusion

The present study sheds light on the ameliorative effect of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues. In-vitro, in-silico, and in-vivo studies showed the antiproliferative effects of both synthesized compounds. 9B and 12B showed significant antiproliferative activity against A549 LC cells. Molecular docking, morphological, biochemical, anti-inflammatory, and antioxidant profiling, as well as the expression of apoptotic proteins, all supported that 9B and 12B could control the growth of EC-induced LC. Based on the overall findings, we can speculate that both synthetic compounds may be considered novel therapeutic compounds for LC treatments.

Data availability

The datasets generated or analyzed in this study are accessible upon reasonable request..

Abbreviations

5-FU:

5-Fluorouracil

A549:

Adenocarcinomic human alveolar basal epithelial cells

AD:

Adenocarcinoma

ADR:

Adriamycin

AUC:

Area under the curve

BAX:

Bcl-2-associated X protein

Bcl-2:

B-cell lymphoma 2

CMC:

Carboxymethyl cellulose

CYP2E1:

Cytochrome P450 2E1

CYP450:

Cytochrome P450

Cyt-C:

Cytochrome complex

DNA:

Deoxyribonucleic acid

EC:

Ethyl Carbamate or Urethane

ECL:

Enhanced chemiluminescent

ELISA:

Enzyme-linked immunosorbent assay

GSH:

Glutathione

H&E:

Hematoxylin and eosin

HepG-2:

Hepatoblastoma cell line

HPLC:

High-performance liquid chromatography

HSV-1:

Anti-herpes simplex virus type-1

IARC:

International Agency for Research on Cancer

LC:

Lung cancer

LCC:

Large cell carcinoma

Mcl-1:

Myeloid cell leukemia sequence 1

NSCLC:

Non-small-cell lung cancer

ProC:

Protein-Carbonyl

PVDF:

Polyvinylidene fluoride

RIPA:

Radio Immunoprecipitation Assay

SCC:

Squamous cell carcinoma

SCLC:

Small cell lung cancer

SOD:

Superoxide Dismutase

SRB assay:

Sulforhodamine B assay

STAT3:

Signal transducer and activator of transcription 3

TBARS:

Thiobarbituric acid reactive substances

TBS-T:

Tris-buffered Tween-20

WB:

Western Blot

References

  1. Li Y, Wang X, Blau DM, Caballero MT, Feikin DR, Gill CJ, et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in children younger than 5 years in 2019: a systematic analysis. Lancet. 2022;399(10340):2047–64.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin. 2021;71(3):209–49.

    Article  Google Scholar 

  3. Singh N, Agrawal S, Jiwnani S, Khosla D, Malik PS, Mohan A et al. Lung cancer in India. Vol. 16, Journal of Thoracic Oncology. Elsevier; 2021. pp. 1250–66.

  4. Xu K, Zhang C, Du T, Gabriel ANA, Wang X, Li X, et al. Progress of exosomes in the diagnosis and treatment of lung cancer. Biomed Pharmacother. 2021;134:111111.

    Article  CAS  PubMed  Google Scholar 

  5. May L, Shows K, Nana-Sinkam P, Li H, Landry JW. Sex differences in Lung Cancer. Cancers. 2023;15(12):3111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Siegel RL, Miller KD, Fuchs HE, Jemal A, Cancer statistics. 2022. CA: a cancer journal for clinicians. 2022;72(1):7–33.

  7. Sekhar T, Thriveni P, Venkateswarlu A, Daveedu T, Peddanna K, Sainath SB. One-pot synthesis of thiazolo [3, 2-a] pyrimidine derivatives, their cytotoxic evaluation and molecular docking studies. Spectrochim Acta Part A Mol Biomol Spectrosc. 2020;231:118056.

    Article  CAS  Google Scholar 

  8. Youssef MM, Amin MA. Microwave assisted synthesis of some new thiazolopyrimidine, thiazolodipyrimidine and thiazolopyrimidothiazolopyrimidine derivatives with potential antioxidant and antimicrobial activity. Molecules. 2012;17(8):9652–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Denise C, Paoli P, Calvani M, Taddei ML, Giannoni E, Kopetz S, et al. 5-fluorouracil resistant colon cancer cells are addicted to OXPHOS to survive and enhance stem-like traits. Oncotarget. 2015;6(39):41706.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Elbanan ME, Amer ME, El-Missiry MA, Othman AI, Shabana SM. Melatonin protects mice from 5-FU hepatotoxicity and improves 5-FU antitumor effects by an apoptotic pathway dependent on cell cycle arrest in A549 human lung cancer cells. RPS Pharm Pharmacol Rep. 2023;2(4):rqad034.

    Article  Google Scholar 

  11. Ghosh S, Pal A, Ray M. Methylglyoxal in combination with 5-Fluorouracil elicits improved chemosensitivity in breast cancer through apoptosis and cell cycle inhibition. Biomed Pharmacother. 2019;114:108855.

    Article  CAS  PubMed  Google Scholar 

  12. Yu Z, Chan S, Wang X, Sun R, Wang M, Wang Z, et al. 5-Fluorouracil Combined with Rutaecarpine synergistically suppresses the growth of Colon cancer cells by inhibiting STAT3. Development and Therapy: Drug Design; 2023. pp. 993–1006.

    Google Scholar 

  13. Wang J, Yang M, Yagi S, Hoffman RM. Oral 5-FU is a more effective antimetastatic agent than UFT. Anticancer Res. 2004;24(3A):1353–60.

    CAS  PubMed  Google Scholar 

  14. YILDIRIM AB, Mutlu E. Cytotoxic effects of Thiazolo [3, 2-C] pyrimidines against Mcf-7 and Hepg2/C3a carcinoma cell lines. Hacettepe J Biology Chem. 2018;46(2):237–46.

    Article  Google Scholar 

  15. Al-Omary FA, Hassan GS, El-Messery SM, El-Subbagh HI. Substituted thiazoles V. Synthesis and antitumor activity of novel thiazolo [2, 3-b] quinazoline and pyrido [4, 3-d] thiazolo [3, 2-a] pyrimidine analogues. Eur J Med Chem. 2012;47:65–72.

    Article  CAS  PubMed  Google Scholar 

  16. Keshari AK, Singh AK, Raj V, Rai A, Trivedi P, Ghosh B et al. p-TSA-promoted syntheses of 5H-benzo [h] thiazolo [2, 3-b] quinazoline and indeno [1, 2-d] thiazolo [3, 2-a] pyrimidine analogs: molecular modeling and in vitro antitumor activity against hepatocellular carcinoma. Drug design, development and therapy. 2017;1623–42.

  17. Keshari AK, Singh AK, Kumar U, Raj V, Rai A, Kumar P et al. 5H-benzo [h] thiazolo [2, 3-b] quinazolines ameliorate NDEA-induced hepatocellular carcinogenesis in rats through IL-6 downregulation along with oxidative and metabolic stress reduction. Drug design, development and therapy. 2017;11:2981.

  18. Skehan P, Storeng R, Scudiero D, Monks A, McMahon J, Vistica D, et al. New colorimetric cytotoxicity assay for anticancer-drug screening. JNCI: J Natl Cancer Inst. 1990;82(13):1107–12.

    Article  CAS  PubMed  Google Scholar 

  19. Vichai V, Kirtikara K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc. 2006;1(3):1112–6.

    Article  CAS  PubMed  Google Scholar 

  20. Kumar P, Faruqui T, Yadav AK, Chandra D, Verma S, Saha S et al. Targeting caspase pathway by novel N-Me aziridine derivatives for hepatocellular carcinoma drug discovery. J Biomol Struct Dynamics. 2023;1–12.

  21. Singh AK, Sreelakshmi P, Pathak P, Kumar A, Singh H, Yadav JP, et al. Design, virtual screening, Molecular Docking, ADME and cytotoxicity studies of 1, 3, 5-Triazine containing heterocyclic scaffolds as selective BRAF Monomeric, Homo and heterodimeric inhibitors. Combinatorial Chemistry & High Throughput Screening; 2024.

  22. Pathak P, Shukla PK, Kumar V, Kumar A, Verma A. Quinazoline clubbed 1, 3, 5-triazine derivatives as VEGFR2 kinase inhibitors: design, synthesis, docking, in vitro cytotoxicity and in ovo antiangiogenic activity. Inflammopharmacology. 2018;26:1441–53.

    Article  CAS  PubMed  Google Scholar 

  23. Dong N, Liu X, Zhao T, Wang L, Li H, Zhang S, et al. Apoptosis-inducing effects and growth inhibitory of a novel chalcone, in human hepatic cancer cells and lung cancer cells. Biomed Pharmacother. 2018;105:195–203.

    Article  CAS  PubMed  Google Scholar 

  24. Kushwaha PS, Raj V, Singh AK, Keshari AK, Saraf SA, Mandal SC, et al. Antidiabetic effects of isolated sterols from Ficus racemosa leaves. RSC Adv. 2015;5(44):35230–7.

    Article  CAS  Google Scholar 

  25. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010;31(2):455–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Pathak P, Naumovich V, Grishina M, Shukla PK, Verma A, Potemkin V. Quinazoline based 1, 3, 5-triazine derivatives as cancer inhibitors by impeding the phosphorylated RET tyrosine kinase pathway: design, synthesis, docking, and QSAR study. Arch Pharm. 2019;352(9):1900053.

    Article  Google Scholar 

  27. Kumar P, Singh AK, Raj V, Rai A, Keshari AK, Kumar D et al. Poly (lactic-co-glycolic acid)-loaded nanoparticles of betulinic acid for improved treatment of hepatic cancer: characterization, in vitro and in vivo evaluations. Int J Nanomed. 2018;975–90.

  28. Kumar P, Kumar M, Gautam AK, Sonkar AB, Verma A, Singh A, et al. Ameliorative effect of fluvoxamine against colon carcinogenesis via COX-2 blockade with oxidative and metabolic stress reduction at the cellular, molecular and metabolic levels. BBA Adv. 2022;2:100046.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sonkar AB, Kumar P, Kumar A, Gautam AK, Verma A, Singh A, et al. Vinpocetine mitigates DMH-induce pre-neoplastic colon damage in rats through inhibition of pro-inflammatory cytokines. Int Immunopharmacol. 2023;119:110236.

    Article  Google Scholar 

  30. Abdel-Fattah MM, Mohamed WR, Hassanein EH, Arab HA, El-shaimaa AA. Role of NF-κB/ICAM-1, JAK/STAT-3, and apoptosis signaling in the anticancer effect of tangeretin against urethane-induced lung cancer in BALB/c mice. Life Sci. 2023;325:121749.

    Article  CAS  PubMed  Google Scholar 

  31. Harvey NL, Kumar S. The role of caspases in apoptosis. Apoptosis. 1998;107–28.

  32. Porter AG, Jänicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 1999;6(2):99–104.

    Article  CAS  PubMed  Google Scholar 

  33. Boice A, Bouchier-Hayes L. Targeting apoptotic caspases in cancer. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2020;1867(6):118688.

  34. Bodduluru LN, Kasala ER, Barua CC, Karnam KC, Dahiya V, Ellutla M. Antiproliferative and antioxidant potential of hesperetin against benzo (a) pyrene-induced lung carcinogenesis in Swiss albino mice. Chemico-Biol Interact. 2015;242:345–52.

    Article  CAS  Google Scholar 

  35. Radwan E, Ali M, Faied SM, Omar HM, Mohamed WS, Abd-Elghaffar SK, et al. Novel therapeutic regimens for urethane-induced early lung cancer in rats: combined cisplatin nanoparticles with vitamin-D3. IUBMB Life. 2021;73(2):362–74.

    Article  CAS  PubMed  Google Scholar 

  36. Kucharska-Newton AM, Rosamond WD, Schroeder JC, McNeill AM, Coresh J, Folsom AR. HDL-cholesterol and the incidence of lung cancer in the atherosclerosis risk in communities (ARIC) study. Lung Cancer. 2008;61(3):292–300.

    Article  PubMed  Google Scholar 

  37. Siemianowicz K, Gminski J, Stajszczyk M, Wojakowski W, Goss M, Machalski M, et al. Serum total cholesterol and triglycerides levels in patients with lung cancer. Int J Mol Med. 2000;5(2):201–6.

    CAS  PubMed  Google Scholar 

  38. Zhou T, Zhan J, Fang W, Zhao Y, Yang Y, Hou X, et al. Serum low-density lipoprotein and low-density lipoprotein expression level at diagnosis are favorable prognostic factors in patients with small-cell lung cancer (SCLC). BMC Cancer. 2017;17:1–9.

    Article  CAS  Google Scholar 

  39. Pathak P, Rimac H, Grishina M, Verma A, Potemkin V. Hybrid quinazoline 1, 3, 5-triazines as epidermal growth factor receptor (EGFR) inhibitors with anticancer activity: design, synthesis, and computational study. ChemMedChem. 2021;16(5):822–38.

    Article  CAS  PubMed  Google Scholar 

  40. Hadisaputri YE, Andika R, Sopyan I, Zuhrotun A, Maharani R, Rachmat R et al. Caspase cascade activation during apoptotic cell death of human lung carcinoma cells A549 induced by marine sponge Callyspongia aerizusa. Drug Design, Development and Therapy. 2021;1357–68.

  41. Simons K, Ikonen E. How cells handle cholesterol. Science. 2000;290(5497):1721–6.

    Article  CAS  PubMed  Google Scholar 

  42. Zab\locka-S\lowińska K, P\laczkowska S, Skórska K, Prescha A, Pawe\lczyk K, Porębska I, et al. Oxidative stress in lung cancer patients is associated with altered serum markers of lipid metabolism. PLoS ONE. 2019;14(4):e0215246.

    Article  Google Scholar 

  43. Zhao C, Zhao H, Li S, Tian H. Independent associations between blood lipid profiles and Lung Cancer Risk. 2021.

  44. Colombo NBR, Rangel MP, Martins V, Hage M, Gelain DP, Barbeiro DF, et al. Caryocar brasiliense camb protects against genomic and oxidative damage in urethane-induced lung carcinogenesis. Braz J Med Biol Res. 2015;48:852–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Khalilpour A, Asghari S, Pourshab M. Synthesis and characterization of Novel Thiazolo [3, 2-a] pyrimidine derivatives and evaluation of antioxidant and cytotoxic activities. Chem Biodivers. 2019;16(5):e1800563.

    Article  PubMed  Google Scholar 

  46. Gomes M, Teixeira AL, Coelho A, Araujo A, Medeiros R. The role of inflammation in lung cancer. Inflamm cancer. 2014;1–23.

  47. Xu C, Zhou L, Lu L, Chen T, Wei S, Lin X, et al. Inflammation has a role in urethane-induced lung cancer in C57BL/6J mice. Mol Med Rep. 2016;14(4):3323–8.

    Article  CAS  PubMed  Google Scholar 

  48. Freed AD, Einstein DR, Carson JP, Jacob RE. Viscoelastic model for lung parenchyma for multi-scale modeling of respiratory system, phase II: Dodecahedral micro-model. Pacific Northwest National Lab.(PNNL), ;; 2012. Richland, WA (United States).

  49. Catlett-Falcone R, Landowski TH, Oshiro MM, Turkson J, Levitzki A, Savino R, et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 1999;10(1):105–15.

    Article  CAS  PubMed  Google Scholar 

  50. Dutta P, Sabri N, Li J, Li WX. Role of STAT3 in lung cancer. Jak-stat. 2014;3(4):e999503.

    Article  PubMed  Google Scholar 

  51. Brentnall M, Rodriguez-Menocal L, De Guevara RL, Cepero E, Boise LH. Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 2013;14:1–9.

    Article  Google Scholar 

  52. Vinocha A, Grover RK, Deepak R. Clinical significance of interleukin-6 in diagnosis of lung, oral, esophageal, and gall bladder carcinomas. J Cancer Res Ther. 2018;14(Suppl 3):S758–60.

    Article  CAS  PubMed  Google Scholar 

  53. young Yoo J, Kim CH, Song SH, Shim BY, Jeong YJ, Im Ahn M, et al. Expression of caspase-3 and c-myc in non-small cell lung cancer. Cancer Res Treatment: Official J Korean Cancer Association. 2004;36(5):303–7.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work is dedicated to the late Dr. Sudipta Saha, who tragically passed away in November 2021. We gladly acknowledge the Department of Science and Technology (DST) Government of India sanctioned the grant (Ref. No. EEQ/2020/000017) to the late Dr. Sudipta Saha and the Indian Council of Medical Research (ICMR), New Delhi, India for the grant of SRF to Archana Bharti Sonkar (ICMR-SRF award no. -3/2/2/52/2022-NCD-III).

Author information

Authors and Affiliations

Authors

Contributions

A.B.S.: Writing – original draft, A.V.: Software, S.Y.: Methodology and formal analysis, R.K.: Methodology, J.S.: Data curation. A.K.K.: Conceptualization. S.R: Methodology. A.K.: Investigation. D.K.: Investigation, N.K.S.: Investigation. S.R.: Investigation. M.K.A.: Visualization. M.N.A.: Validation. A.S.S.: Validation. S.S.: Conceptualization and Supervision, and G.K.: Project administration and supervision.

Corresponding author

Correspondence to Gaurav Kaithwas.

Ethics declarations

Ethical approval and consent to participate

The experiment was conducted in accordance with the CPCSEA guidelines for laboratory animals and ethics. The study was approved by the Institutional Animal Ethical Committee (CPCSEA No. 809/PO/Re/S/03/CPCSEA) of Babu Banarasi Das Northern India Institute of Technology Lucknow (approval no. BBDNIIT/IAEC/APR/2022/06).

Consent for publication

Not applicable.

Conflict of interest

The authors affirm that they do not possess any obvious conflicting financial interests that could have potentially influenced the findings presented in this paper.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sonkar, A.B., Verma, A., Yadav, S. et al. Antiproliferative effect of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues on IL-6 mediated STAT3 and role of the apoptotic pathway in albino Wistar rats of ethyl carbamate-induced lung carcinoma: In-silico, In-vitro, and In-vivo study. Cancer Cell Int 24, 219 (2024). https://doi.org/10.1186/s12935-024-03390-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-024-03390-6

Keywords