Next Article in Journal
Functional Polymer Hybrid Nanocomposites Based on Polyolefins: A Review
Next Article in Special Issue
Evolution of Nitrogen-Based Alkylating Anticancer Agents
Previous Article in Journal
Effect of Strain Hardening Laws on Solution Behavior Near Frictional Interfaces in Metal Forming Processes: A Simple Analytical Example
Previous Article in Special Issue
Functionalized N-Pyridinylmethyl Engrafted Bisarylmethylidenepyridinones as Anticancer Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Molecular Biology Studies of Spirooxindole Heterocyclic Hybrids

by
Dhaifallah M. Al-thamili
,
Abdulrahman I. Almansour
,
Natarajan Arumugam
,
Faruq Mohammad
and
Raju Suresh Kumar
*
Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
*
Author to whom correspondence should be addressed.
Processes 2020, 8(11), 1473; https://doi.org/10.3390/pr8111473
Submission received: 5 October 2020 / Revised: 10 November 2020 / Accepted: 16 November 2020 / Published: 17 November 2020
(This article belongs to the Special Issue Advances in Anticancer Agents)

Abstract

:
In the present report, we provide the results of the molecular biology studies of spiroheterocyclic hybrids, where the derivatives are found to possess potential anticancer activity towards cancer cells. A series of spiroxindole–pyrrolidine heterocyclic hybrids were evaluated for cell viability and proliferation against HepG2 cancer cells at concentrations in the range of 12.5–200 µg/mL over two different time periods of 24 and 48 h. In addition, the highly active compounds were also verified for their behavior towards noncancer cells (L929 cells), and it was found that the tested derivatives were not aggressive due to the observation of only limited cell loss, as compared to the cancer cells. Further analysis of the observed toxicity mechanism showed the apoptotic pathway was mediated by oxidative stress, with the involvement of caspases.

1. Introduction

Cancer, an uncontrolled growth of abnormal cells, is the second foremost origin of death worldwide after heart disease [1,2,3]. Cancer is the cause of nearly 14 million demises every year and this number is predicted to double by the year 2030. Cancerous diseases are mostly complicated in humans and can originate in various organs like breast, colon, lung, stomach, and liver, to mention a few [4]. In order to treat such cancerous diseases, various treatment modalities are available, but each method has its own limitations. Among many different treatment modalities, cancer chemotherapy has proven to be easy and proficient for the eradication of aggressive cancer cells without causing trouble to normal cells at a higher level [5]. For the successful implementation of this chemotherapeutic treatment, the selection and recognition of highly active chemical moieties are of the highest significance because of the determined ability of the tumor cell’s immune response towards traditional chemotherapeutic agents. Since multidrug resistance development is one of the major challenges associated with the current chemotherapeutics, continuous research is highly demanded in the oncology sector for the identification and testing of novel chemo-moieties that can offer lesser toxicities and uplifted efficiencies [6,7].
Apoptosis, also called programmed cell death, is a highly preferred mechanism for the control of tumor cells, which generally occurs through oxidative stress and DNA damage when the cells lose their capacity to repair the damage caused by the therapeutic/anticancer drugs. In addition, the repair mechanism associated with the apoptosis process helps to control the aggressive growth and proliferation of unwanted cells [8]. Hence, any defects involved in the apoptotic signaling process may lead to the propagation of various diseases in humans, which mainly includes cancer. The cancer cells that develop in such a way contribute to the growth of unwanted masses, called “tumors”, by surviving over their intended life spans, subverting them from the need for exogenous survival factors and protecting them from oxidative stress and hypoxia. Thus, the developed tumors maintain the genetically altered cells, which promote angiogenesis, deregulate the normal cell proliferation process, interfere with cell differentiation, and increase the possibilities of invasiveness during the progression stage. Therefore, restoring the natural capacity of diseased cells to easily participate in the normal apoptotic process can be a reliable strategy in potential cancer treatments [9].
Spiropyrrolidine-oxindole molecular scaffolds hold interesting structural features and robust bioactivity profiles, including antibacterial [10], antiviral [11], anticancer [12,13,14], and local anesthetic activities [15], and have ensued as promising synthetic targets. The activity of such central ring systems for the exploration and exploitation of pharmacophore space via diversity-oriented synthesis (DOS) has led to the discovery of new drug candidates [16,17]. For instance, natural spirooxindoles viz. horsfiline [18], and elacomine [19] display important biological activities [20,21]. Notably, even non-natural spirooxindoles, namely, MI-773 and MI-888 (Figure 1), maintain the capacity to control the cell-cycle and protein–protein interactions associated with the nonpeptidic inhibitors of p53–MDM2, which, in general, is considered as dangerous for the p53 protein’s activity of tumor-suppression. Further, inhibiting the MDM2–p53 protein interactions can have potential therapeutic implications in cancer treatment [22].
In this context, we have been engaged in the exploration of the biological potential of spiropyrrolidine–oxindole heterocyclic hybrids. For instance, the spirooxindole–pyrrolidine heterocyclic hybrids (A) with unsubstituted piperidone moiety have displayed promising anticancer activity in leukemia lymphoblastic (CCRF-CEM), human breast carcinoma (MDA-MB-231), and ovarian carcinoma (SK-OV-3) cells [23]. Additionally, the N-styryl-substituted spirooxindole–pyrrolidine heterocyclic hybrids (B) have displayed potent anticancer activities as well [24,25]. Moreover, the N-acrolyl derivatives (C) displayed promising cholinesterase inhibitory activities [26]. Furthermore, compounds embedded with N-pyridyl structural subunits have been shown to exhibit good-to-excellent anticancer activities against HepG2 cells [27]. It is pertinent to note that the inherent ability of spiroheterocyclic hybrids to project functionality in all three dimensions facilitates easy interactions of ligands with the three-dimensional binding site of spiroheterocyclic hybrids than with planar systems [28,29]. Prompted by this characteristic feature and the abovementioned biological precedents of the spiroheterocyclic hybrids, herein, we present the behavior of spiropyrrolidine–oxindole heterocyclic hybrids derived from N-pyridyl-4-piperidone-α,β-unsaturated ketones towards cancer and noncancer cells. We presume the molecular hybridization of spirooxindole, pyrrolidine, piperidone, and N-pyridy units embedded with an α,β-unsaturated ketone moiety into new single hybrid architectures, with preselected activities of the original individual subunits that would enhance the biological profiles of new constructs. In one of our recent studies, we investigated the biological potential of N-pyridyl 4-piperidones embedded with an α,β-unsaturated ketone moiety [30]. As a continuation, we present our findings of N-pyridyl-substituted spirooxindole–pyrrolidine heterocyclic hybrids constructed through the aforementioned N-pyridyl 4-piperidone α,β-unsaturated ketones and their molecular biology activities towards cancer and noncancer cells.

2. Materials and Methods

The selection of cell lines and all the assay protocols for testing the anticancer activity and intracellular mechanisms of spirooxindole–pyrrolidine heterocyclic hybrids were in accordance with our recently published article [30]. The derivatives 1(ah) with a range of concentrations (12.5–200 µg/mL) were first tested against HepG2 cancer cells over an incubation period of 24–48 h. From those studies, the two most active compounds (1b and 1f) were carried over to test for cell viability and proliferation against the L929 noncancer cell line. Furthermore, the intracellular mechanistic pathways of treatment with highly active compound 1b were studied against the cancer cell line by means of apoptosis, oxidative stress, and caspase activity assays. For the analysis, melphalan (Mel; 15 µg/mL) was used as the positive control, and cells without any treatment were used as the negative control. Additionally, based on statistical analysis, * corresponds to significant values (p < 0.05) and ** to highly significant (p < 0.01) values against the control measurements. The detailed procedure for the maintenance of cell cultures and assay protocols is provided in the supporting information file.

3. Results and Discussion

The required N-pyridyl–spirooxindole–pyrrolidine heterocyclic hybrids 1(ah) were synthesized in accordance with our recent study [31]. The molecular structure of these heterocyclic hybrids 1(ah) is shown in Scheme 1. Initially, these heterocyclic hybrids 1(ah) were tested for their anticancer potential against the HepG2cancer cell line. The in-vitro cell viability studies, following the incubation of HepG2 cancer cells with that of the test compounds 1(ah) at concentrations in the range of 12.5–200 µg/mL over 24 and 48 h, are shown in Figure 2 and Figure 3, respectively.
From the comparative analysis of the obtained results (Figure 2), all the tested compounds exhibited some level of activity against the cancer cells as we observed a significant number of cell losses starting from 50 µg/mL concentration (48% cell loss) over a 24-h period (as compared against positive control of Mel and negative control of no treatment). Additionally, with an increase of the incubation period to 48 h (Figure 3), this activity increased (72% cell loss at the same concentration of 50 µg/mL) due to increased interaction of the testing compounds with the cells. The IC50 values, following the incubation of synthesized compounds 1(ah) with that of HepG2 cancer cells for 24 and 48 h, are tabulated in Table 1. From the comparison of IC50 values, the least activity was observed for compound 1g at 24 h and compound 1a at 48 h incubation periods. However, the highest activity was noted for compound 1b, followed by compound 1f, at both the incubation periods of 24 and 48 h. Based on the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay results, the anticancer activity and the associated IC50 values order during the 24 h incubation period is 1b > 1f > 1d > 1h > 1a > 1e > 1c > 1g, while for the 48 h period, it is 1b > 1f > 1c > 1g > 1h > 1d > 1e > 1a.
The MTT studies (HepG2 cancer cells) for the synthesized derivatives indicated that compounds 1b and 1f are highly active at the highest incubation period of 48 h, and hence, we selected these two compounds and assay conditions for the following analysis. Therefore, to test the behavior of highly active compounds 1b and 1f on healthy normal cells, the noncancer L929 cells were studied by incubating them at 12.5–200 µg/mL concentration over a 48-h period (Figure 4). From the comparative analysis of the results with that of positive and negative controls, both compounds 1b and 1f were not found to induce significant cell viability losses, as we observed only 18% loss for compound 1b and 22% loss for compound 1f at 50 µg/mL concentration. The cells at the highest tested concentration of 200 µg/mL provided approximately 40% loss, indicating that the compounds were behaving in a safe manner towards the noncancer cells while offering anticancer activity against the cancer cells. In addition, the information provided in Table 1 indicates that compound 1b has the IC50 value of 301 µg/mL, while compound 1f has the IC50 value of 257 µg/mL; these values are quite high compared to the cancer cell IC50 values. Thus, taking into consideration such variation in the activity of our synthesized compounds against cancer and noncancer cells, it can be inferred that compounds 1b and 1f potentially serve as safe therapeutic agents.
Figure 5 shows the comparison of the flow cytometry of the apoptotic assay results for the highly active compound 1b at its IC50 concentration (24 µg/mL; 37 µM) with that of positive control (Mel; 15 µg/mL) and negative control (cells without any treatment). From Figure 5, A-1, B-1, and C-1 correspond to the total population of cells used for the fluorescence analysis of the negative control, the positive control (Mel), and compound 1b, respectively. Similarly, A-2 (untreated control), B-2 (Mel), and C-2 (compound 1b) correspond to the combined fluorescence of PI and Annexin-V-FITC, while A-3 (untreated control), B-3 (Mel), and C-3 (compound 1b) correspond to the Annexin-V-FITC fluorescence intensity. From the analysis, it was found that the compound-1b-treated cells (Figure 5, C-2) exhibited about 48% live, 6% early-apoptotic, 37% late-apoptotic, and 7% necrotic cells. However, the positive control (Mel)-treated cells (Figure 5, B-2) had 2% early-apoptotic, 46% late-apoptotic, and 34% necrotic cells, while the negative control cells had 97% live cells. The comparison of our compound 1b result (Figure 5, C-2) with that of positive (Figure 5, B-2) and negative controls (Figure 5, A-2) can primarily indicate the occurrence of a late-apoptotic mechanism. Furthermore, the compound-1b-treated cells (Figure 5, C-3) indicated the M1 and M2 values of 37% (normal cells) and 63% (apoptotic cells), similar to the positive control (Figure 5, B-3; 35% normal and 65% apoptotic), and thereby confirm an exclusive apoptotic pathway in the cancer cells.
Figure 6 shows the comparison of ROS assay results provided by flow cytometry for the highly active compound 1b at its IC50 concentration (24 µg/mL) with that of positive control (Mel; 15 µg/mL) and negative control (cells without any treatment). From the figure, the population of cells tested for the flow cytometric analysis of the ROS assay is represented in Figure 6 of A-1 (negative control), B-1 (positive control), and C-1 (compound 1b), while the corresponding sample’s H2DCFDA fluorescence population are in A-2, B-2, and C-2, respectively. The H2DCFDA fluorescence intensities from the negative control, the positive control, and compound 1b are compared in Figure 6 (A-3, B-3, and C-3, respectively). From the comparison of fluorescence intensities for compound-1b-treated cells (Figure 6, C-3), about 10% of cells (M1) were normal and 89% (M2) were experiencing ROS-mediated oxidative stress. This is quite high when compared to the M1 and M2 values indicated by positive-control-treated cells (Figure 6, B-3), i.e., 86% normal and 13% ROS-producing cells (respectively), and, thereby, confirms the oxidative ability of our synthesized compound 1b.
Figure 7 shows the comparison of the caspase-3 activity of compound 1b (24 µg/mL concentration) with that of positive control (Mel; 15 µg/mL) and negative control (cells without any treatment). From Figure 7d, the compound-1b-treated cells exhibited higher relative fluorescence intensity than of positive-control-treated cells, and the same can also be visualized from the cells in Figure 7b,c. Green fluorescence is observed in the cells that are activated through the release of caspase-3 proteins, and, at the same time, the cells are alive and undergoing the apoptotic pathway because of the effect of compound 1b. The observation of high fluorescence levels for the compound-1b-treated cancer cells confirms the active role played by the caspases (in support of caspase-3 activity). However, no significant fluorescence is observed in the untreated controls (Figure 7a) because of the absence of caspase activity, while slight fluorescence in Mel (Figure 7b) and significant fluorescence in compound-1b-treated cells (Figure 7c) confirm the caspase-3 activity, along with the apoptosis mechanism in live cells.
In the present study, the anticancer activity of spirooxindole–pyrrolidine heterocyclic hybrids has been investigated against cancer cells, where the results have proven that all the derivatives exhibit some level of activity. This activity seems to increase with that of concentration and incubation period, and the highly active compound was found to be 1b, with the lowest IC50 value of ~24 µg/mL over a treatment time of 48 h. Further testing of the intracellular pathways followed by compound 1b treatment proved that the cancer cells are experiencing the apoptotic mechanism, with the release of oxidative stress mediated by ROS and the involvement of caspases. Comparing the present study results with that of our recently published work [31], it primarily indicates that the spirooxindole–pyrrolidine heterocycles are a little less active than the N-pyridinylmethyl-engrafted bisarylmethylidenepyridinones. The lower activity is reflected in the form of IC50 values, i.e., we observed lower activity at about 12 µg/mL (24 h) for the N-pyridinylmethyl-engrafted bisarylmethylidenepyridinones and at 24 µg/mL (48 h) for the spirooxindole–pyrrolidine heterocycles. However, the other assay results, such as apoptosis, oxidative stress, and caspase activity, were almost the same for both of the chemical moieties under similar experimental conditions. In addition, the response of the spirooxindole–pyrrolidine heterocycles towards noncancer cells is the same as the N-pyridinylmethyl-engrafted bisarylmethylidenepyridinones, revealing that the chemical moiety is able to differentiate between the aggressively growing cancer and nonaggressive healthy normal cells. This shows that the spirooxindole–pyrrolidine heterocycles can be employed for the prolonged therapeutic actions against cancer cells; for a similar but quicker effect, the N-pyridinylmethyl-engrafted bisarylmethylidenepyridinones are more suitable.

4. Conclusions

The biological evaluation of our synthesized compounds 1(ah) revealed that all the derivatives exhibit anticancer activity against HepG2 cancer cells, and this activity is quite high at the 48 h incubation period. Additionally, the mechanistic investigation of the highly active compound 1b provided the information that the cells are experiencing an apoptotic pathway, with the induction of oxidative stress carried by ROS and the involvement of caspases. The observed activity of compound 1b at a very low IC50 value and, at the same time, its not-very-aggressive nature against noncancer cells indicate the potential role that can be played by spiropyrrolidine derivatives as chemotherapeutic agents.

Supplementary Materials

The following are available online at https://www.mdpi.com/2227-9717/8/11/1473/s1.

Author Contributions

R.S.K.: conceptualization, writing original draft preparation, and supervision; D.M.A.-t.: investigation; A.I.A.: investigation, supervision, and funding acquisition; N.A.: investigation; F.M.: investigation, writing, and original draft preparation. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Deanship of Scientific Research, King Saud University, Riyadh, Saudi Arabia, through the grant number RG-1438-052.

Acknowledgments

The authors thank Stellixir Biotech Pvt. Ltd, Bangalore, India for performing the biological assays. The authors also thank the Deanship of Scientific Research, King Saud University, Riyadh, Saudi Arabia for funding this work through the grant number RG-1438-052.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  2. Ames, B.N.; Gold, L.S.; Willett, W.C. The causes and prevention of cancer. Proc. Natl. Acad. Sci. USA 1995, 92, 5258–5265. [Google Scholar] [CrossRef] [Green Version]
  3. Seyfried, T.N.; Huysentruyt, L.C. On the origin of cancer metastasis. Crit. Rev. Oncog. 2013, 18, 43–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Blackadar, C.B. Historical review of the causes of cancer. World J. Clin. Oncol. 2016, 7, 54–86. [Google Scholar] [CrossRef] [PubMed]
  5. Schirrmacher, V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review). J. Oncol. 2019, 54, 407–419. [Google Scholar]
  6. Mansoori, B.; Mohammadi, A.; Davudian, S.; Shirjang, S.; Baradaran, B. The different mechanisms of cancer drug resistance: A brief review. Adv. Pharm. Bull. 2017, 7, 339–348. [Google Scholar] [CrossRef]
  7. Roy, S.; Kumar, A.; Islam, M.S.; Rabbi, F.A.; Paul, P.; Mia, M.M.; Islam, A.K.M.K.; Ray, A.K. Drug resistance and its future perspectives in cancer treatment. Asian Oncol. Res. J. 2020, 3, 26–46. [Google Scholar]
  8. Cotter, T.G. Apoptosis and cancer: The genesis of a research field. Nat. Rev. Cancer. 2009, 9, 501–507. [Google Scholar] [CrossRef]
  9. Plati, J.; Bucur, O.; Khosravi-Far, R. Dysregulation of apoptotic signaling in cancer: Molecular mechanisms and therapeutic opportunities. J. Cell. Biochem. 2008, 104, 1124–1149. [Google Scholar] [CrossRef] [Green Version]
  10. Chande, M.S.; Verma, R.S.; Barve, P.A.; Khanwelkar, R.R.; Vaidya, R.B.; Ajaikumar, K.B. Facile synthesis of active antitubercular, cytotoxic and antibacterial agents: A Michael addition approach. Eur. J. Med. Chem. 2005, 40, 1143–1148. [Google Scholar] [CrossRef]
  11. Lundahl, K.; Schut, J.; Schlatmann, J.L.; Paerels, G.B.; Peters, A. Synthesis and antiviral activities of adamantane spiro compounds. 1. Adamantane and analogous spiro-3’-pyrrolidines. J. Med. Chem. 1972, 15, 129–132. [Google Scholar] [CrossRef] [PubMed]
  12. Yang, J.; Liu, X.; Wang, D.; Tian, M.; Han, S.; Feng, T.; Liu, X.; Mei, R.; Zhou, Y. Diversity-oriented one-pot multicomponent synthesis of spirooxindole derivatives and their biological evaluation for anticancer activities. Tetrahedron 2016, 72, 8523–8536. [Google Scholar] [CrossRef]
  13. Lotfy, G.; Said, M.M.; Ashry, E.E.; Tamany, E.S.; Al-Dhfyan, A.; Aziz, Y.M.; Barakat, A. Synthesis of new spirooxindole-pyrrolothiazole derivatives: Anti-cancer activity and molecular docking. Bioorg. Med. Chem. 2017, 254, 1514–1523. [Google Scholar] [CrossRef] [PubMed]
  14. Barakat, A.; Islam, M.S.; Ghawas, H.M.; Al-Majid, A.; El-Senduny, F.F.; Badria, F.; Elshaier, Y.A.; Ghabbour, H. Substituted spirooxindole derivatives as potent anticancer agents through inhibition of phosphodiesterase 1. RSC Adv. 2018, 8, 14335–14346. [Google Scholar] [CrossRef] [Green Version]
  15. Kornet, M.J.; Thio, A.P. Oxindole-3-spiropyrrolidines and -piperidines. Synthesis and local anesthetic activity. J. Med. Chem. 1976, 19, 892–898. [Google Scholar] [CrossRef] [PubMed]
  16. Maclean, D.; Schullek, J.R.; Murphy, M.M.; Ni, Z.J.; Gordon, E.M.; Gallop, M.A. Encoded combinatorial chemistry: Synthesis and screening of a library of highly functionalized pyrrolidines. Proc. Natl. Acad. Sci. USA 1997, 94, 2805–2810. [Google Scholar] [CrossRef] [Green Version]
  17. Lo, M.M.-C.; Neumann, C.S.; Nagayama, S.; Perlstein, E.O.; Schreiber, S.L. A Library of Spirooxindoles Based on a Stereoselective Three-Component Coupling Reaction. J. Am. Chem. Soc. 2004, 126, 16077–16086. [Google Scholar] [CrossRef]
  18. Jossang, A.; Jossang, P.; Hadi, H.A.; Sevenet, T.; Bodo, B. Horsfiline, an oxindole alkaloid from Horsfieldia superba. J. Org. Chem. 1991, 56, 6527–6530. [Google Scholar] [CrossRef]
  19. Pellegrini, C.; Weber, M.; Borschberg, H.-J. Total Synthesis of (+)-Elacomine and (−)-Isoelacomine, Two Hitherto Unnamed Oxindole Alkaloids from Elaeagnus commutate. Helvitica 1996, 79, 151–168. [Google Scholar]
  20. Galliford, C.V.; Scheidt, K.A. Pyrrolidinyl-spirooxindole natural products as inspirations for the development of potential therapeutic agents. Angew. Chem. Int. Ed. 2007, 46, 8748–8758. [Google Scholar] [CrossRef]
  21. Roeder, E.; Wiedenfeld, H. Pyrrolizidine alkaloids in plants used in the traditional medicine of Madagascar and the Mascarene islands. Pharmazie 2011, 66, 637–647. [Google Scholar] [PubMed]
  22. Wang, S.; Zhao, Y.; Aguilar, A.; Bernard, D.; Yang, C.-Y. Targeting the MDM2–p53 Protein–Protein Interaction for New Cancer Therapy: Progress and Challenges. Cold Spring Harb. Perspect. Med. 2017, 7, a026245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Almansour, A.I.; Kumar, R.S.; Beevi, F.; Shirazi, A.N.; Osman, H.; Ismail, R.; Choon, T.S.; Sullivan, B.; McCaffrey, K.; Nahhas, A.; et al. Facile, Regio- and Diastereoselective Synthesis of Spiro-Pyrrolidine and Pyrrolizine Derivatives and Evaluation of Their Antiproliferative Activities. Molecules 2014, 19, 10033–10055. [Google Scholar] [CrossRef] [Green Version]
  24. Almansour, A.I.; Arumugam, N.; Kumar, R.S.; Subbarayan, P.V.; Alshatwi, A.A.; Ghabbour, H.A. Anticancer Agents. U.S. Patent 9,486,444 B1, 8 November 2016. [Google Scholar]
  25. Almansour, A.I.; Arumugam, N.; Kumar, R.S.; Subbarayan, P.V.; Alshatwi, A.A.; Athinarayanan, J. Anticancer Agents. U.S. Patent 9,873,699B1, 23 January 2018. [Google Scholar]
  26. Kia, Y.; Osman, H.; Kumar, R.S.; Basiri, A.; Murugaiyah, V. Synthesis and discovery of highly functionalized mono- and bis-spiro-pyrrolidines as potent cholinesterase enzyme inhibitors. Bioorg. Med. Chem. Lett. 2014, 24, 1815–1819. [Google Scholar] [CrossRef] [PubMed]
  27. Filosa, R.; Peduto, A.; de Caprariis, P.; Saturnino, C.; Festa, M.; Petrella, A.; Pau, A.; Pinna, G.A.; Colla, P.L.; Busonera, B.; et al. Synthesis and antiproliferative properties ofN3/8-disubstituted3,8-diazabicyclo [3.2.1]octane analogues of 3,8-bis[2-(3,4,5-trimethoxyphenyl)pyridin-4-yl]methyl-piperazine. Eur. J. Med. Chem. 2007, 42, 293–306. [Google Scholar] [CrossRef] [PubMed]
  28. Carreira, E.M.; Fessard, T.C. Four-membered ring-containing spirocycles: Synthetic strategies and opportunities. Chem. Rev. 2014, 114, 8257–8322. [Google Scholar] [CrossRef]
  29. Zheng, Y.-J.; Tice, C.M. The utilization of spirocyclic scaffolds in novel drug discovery. Expert Opin. Drug Discov. 2016, 11, 831–834. [Google Scholar] [CrossRef] [Green Version]
  30. Al-thamili, D.M.; Almansour, A.I.; Arumugam, N.; Mohammad, F.; Kumar, R.S. Functionalized N-pyridinylmethyl engrafted bisarylmethylidenepyridinones as anticancer agents. Processes 2020, 8, 1154. [Google Scholar] [CrossRef]
  31. Kumar, R.S.; Al-thamili, D.M.; Almansour, A.I.; Arumugam, N.; Dege, N. [Bmim]Br accelerated one-pot three-component cascade protocol for the construction of spirooxindole–pyrrolidine heterocyclic hybrids. Molecules 2020, 25, 4779. [Google Scholar] [CrossRef]
Figure 1. Representative natural and unnatural spirooxindole–pyrrolidine heterocyclic hybrids. (A) The spirooxindole–pyrrolidine heterocyclic hybrids, (B) The N-styryl-substituted spirooxindole–pyrrolidine heterocyclic hybrids, (C) The N-acrolyl derivatives.
Figure 1. Representative natural and unnatural spirooxindole–pyrrolidine heterocyclic hybrids. (A) The spirooxindole–pyrrolidine heterocyclic hybrids, (B) The N-styryl-substituted spirooxindole–pyrrolidine heterocyclic hybrids, (C) The N-acrolyl derivatives.
Processes 08 01473 g001
Scheme 1. Schematic representation of spirooxindole–pyrrolidine heterocyclic hybrids of the present study.
Scheme 1. Schematic representation of spirooxindole–pyrrolidine heterocyclic hybrids of the present study.
Processes 08 01473 sch001
Figure 2. The percentage viability of HepG2 cancer cells when treated with the synthesized derivatives 1(ah) in the concentration range of 12.5–200 µg/mL and compared with that of positive (melphalan (Mel); 15 µg/mL) and negative controls (no treatment) over a 24 h period. * and ** correspond to significant (p < 0.05) and highly significant (p < 0.01) values against the untreated control.
Figure 2. The percentage viability of HepG2 cancer cells when treated with the synthesized derivatives 1(ah) in the concentration range of 12.5–200 µg/mL and compared with that of positive (melphalan (Mel); 15 µg/mL) and negative controls (no treatment) over a 24 h period. * and ** correspond to significant (p < 0.05) and highly significant (p < 0.01) values against the untreated control.
Processes 08 01473 g002
Figure 3. The percentage viability of HepG2 cancer cells, followed by the treatment of synthesized derivatives 1(ah) in the concentration range of 12.5–200 µg/mL and compared against the positive control (Mel; 15 µg/mL) and negative control (no treatment) over a 48 h period. * and ** correspond to significant (p < 0.05) and highly significant (p < 0.01) values against the untreated control.
Figure 3. The percentage viability of HepG2 cancer cells, followed by the treatment of synthesized derivatives 1(ah) in the concentration range of 12.5–200 µg/mL and compared against the positive control (Mel; 15 µg/mL) and negative control (no treatment) over a 48 h period. * and ** correspond to significant (p < 0.05) and highly significant (p < 0.01) values against the untreated control.
Processes 08 01473 g003
Figure 4. The percentage viability of L929 noncancer cells, followed by the treatment of compounds 1b and 1f at concentrations in the range of 12.5–200 µg/mL (48 h period). Positive control of Mel (15 µg/mL) and negative control of cells without any treatment were applied. * corresponds to significant (p < 0.05) values against the untreated control.
Figure 4. The percentage viability of L929 noncancer cells, followed by the treatment of compounds 1b and 1f at concentrations in the range of 12.5–200 µg/mL (48 h period). Positive control of Mel (15 µg/mL) and negative control of cells without any treatment were applied. * corresponds to significant (p < 0.05) values against the untreated control.
Processes 08 01473 g004
Figure 5. Comparison of apoptosis assay results for HepG2 cancer cells at 48 h incubation period. From the figure, A, B, and C correspond to the negative control, the positive control (Mel; 15 µg/mL), and compound 1b, respectively, while 1, 2, and 3 stand for the population of cells selected for the analysis (FSC-H: forward scatter histogram; SSC-H: side scatter histogram), combined fluorescence from PI and Annexin-V-FITC, and pure Annexin-V-FITC fluorescence, respectively.
Figure 5. Comparison of apoptosis assay results for HepG2 cancer cells at 48 h incubation period. From the figure, A, B, and C correspond to the negative control, the positive control (Mel; 15 µg/mL), and compound 1b, respectively, while 1, 2, and 3 stand for the population of cells selected for the analysis (FSC-H: forward scatter histogram; SSC-H: side scatter histogram), combined fluorescence from PI and Annexin-V-FITC, and pure Annexin-V-FITC fluorescence, respectively.
Processes 08 01473 g005
Figure 6. Comparison of flow cytometric analysis results for ROS generation in HepG2 cancer cells at 48 h incubation. From the figure, A, B, and C correspond to the negative control (without any treatment), the positive control (Mel; 15 µg/mL), and compound 1b (at IC50), respectively, while 1, 2, and 3 stand for the cell population tested, number of cells exhibiting H2DCFDA fluorescence, and % of fluorescent intensity, respectively.
Figure 6. Comparison of flow cytometric analysis results for ROS generation in HepG2 cancer cells at 48 h incubation. From the figure, A, B, and C correspond to the negative control (without any treatment), the positive control (Mel; 15 µg/mL), and compound 1b (at IC50), respectively, while 1, 2, and 3 stand for the cell population tested, number of cells exhibiting H2DCFDA fluorescence, and % of fluorescent intensity, respectively.
Processes 08 01473 g006
Figure 7. Caspase 3 activity comparison of HepG2 cancer cells, followed by the treatment of IC50 concentration of compound 1b (c) with that of positive control of Mel (15 µg/mL) (b) and negative control (a) over a 48 h period. Relative fluorescence intensity of all the samples is compared in (d).
Figure 7. Caspase 3 activity comparison of HepG2 cancer cells, followed by the treatment of IC50 concentration of compound 1b (c) with that of positive control of Mel (15 µg/mL) (b) and negative control (a) over a 48 h period. Relative fluorescence intensity of all the samples is compared in (d).
Processes 08 01473 g007
Table 1. Comparison of the IC50 values of the tested compounds 1(a-h) against HepG2 cells over 24 and 48 h incubation periods.
Table 1. Comparison of the IC50 values of the tested compounds 1(a-h) against HepG2 cells over 24 and 48 h incubation periods.
EntryCompoundIC50 Values in µg/mL (µM); 24 hIC50 Values in µg/mL (µM); 48 h
HepG2 CellsHepG2 CellsL929 Cells
1 Processes 08 01473 i001237.59 ± 12.5
(385.23 ± 20.27)
126.81 ± 10.4
(205.61 ± 16.86)
-
2 Processes 08 01473 i00272.24 ± 8.7
(112.03 ± 13.49)
23.81 ± 2.3
(36.92 ± 3.57)
301.72 ± 12.51
(467.93 ± 19.40)
3 Processes 08 01473 i003265.24 ± 9.4
(391.90 ± 13.89)
29.83 ± 2.1
(44.07 ± 3.10)
-
4 Processes 08 01473 i004107.74 ± 7.5
(152.44 ± 10.62)
44.1 ± 2.6
(62.40 ± 3.68)
-
5 Processes 08 01473 i005237.64 ± 8.9
(368.55 ± 13.80)
45.86 ± 2.8
(71.12 ± 4.34)
-
6 Processes 08 01473 i00690.01 ± 5.6
(131.28 ± 8.17)
25.01 ± 3.2
(36.48 ± 4.67)
257.16 ± 9.6
(375.06 ± 14.00)
7 Processes 08 01473 i007509.28 ± 14.3
(780.23 ± 21.91)
40.51 ± 3.6
(62.06 ± 5.52)
-
8 Processes 08 01473 i008157.27 ± 11.3
(208.43 ± 14.98)
43.17 ± 4.5
(57.21 ± 5.96)
-
9 Processes 08 01473 i009---
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

M. Al-thamili, D.; Almansour, A.I.; Arumugam, N.; Mohammad, F.; Suresh Kumar, R. In Vitro Molecular Biology Studies of Spirooxindole Heterocyclic Hybrids. Processes 2020, 8, 1473. https://doi.org/10.3390/pr8111473

AMA Style

M. Al-thamili D, Almansour AI, Arumugam N, Mohammad F, Suresh Kumar R. In Vitro Molecular Biology Studies of Spirooxindole Heterocyclic Hybrids. Processes. 2020; 8(11):1473. https://doi.org/10.3390/pr8111473

Chicago/Turabian Style

M. Al-thamili, Dhaifallah, Abdulrahman I. Almansour, Natarajan Arumugam, Faruq Mohammad, and Raju Suresh Kumar. 2020. "In Vitro Molecular Biology Studies of Spirooxindole Heterocyclic Hybrids" Processes 8, no. 11: 1473. https://doi.org/10.3390/pr8111473

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

Article Metrics

Back to TopTop