Skip to main content

Synthesis of biologically active sulfonamide-based indole analogs: a review

Abstract

The class of heterocyclic compounds exhibits a variety of pharmacological actions, long employed as an active ingredient in drug design and production. Indole and its derivatives are crucial in medicinal chemistry. Due to its physiological action, it has been gaining a lot of interest by exhibiting antibacterial, anticonvulsant, antifungal, antimalarial, antidiabetic, antidepressant, anticancer, and antimicrobial properties. The indole moiety constitutes a benzene ring fused with a pyrrole ring to form a bicyclic structure with a nitrogen atom as the major component and is produced by a variety of techniques. The sulfonamide analogs of indole usually referred to as sulfa medicines have recently been produced and exhibit strong antimicrobial actions. The goal of this work is to present the most recent methods for synthesizing indole-sulfonamide derivatives, together with data on their reported activities and synthetic scheme from 2010 to 2023. We anticipate that this review will help medicinal chemists rationally develop pharmacologically active derivatives of indole with sulfonamide scaffolds.

Graphical Abstract

Background

A large variety of heterocyclic molecules, including “Indole”, is clinically active, making heterocyclic chemistry crucial in medication design [1]. In 1866, while researching the indigo dye, Baeyar and knop created indole for the first time [2]. Baeyar used reduced oxindole [3] to make indole. Indole, with the chemical formula “C8H7N”, is often referred to as 1H-benzo[b] pyrrole [4]. The temperature at which indole melts is noted to be around 52 °C, while its boiling happens around 253 °C giving a solid crystalline structure of this compound, a bicyclic structure [5].

Some naturally occurring alkaloids have an indole ring. There have been claims that indole derivatives have biological effects and possess anti-inflammatory [6, 7], antitubercular [8,9,10], antidiabetic properties [11, 12], antioxidant [13, 14], anticancer [15,16,17,18], anticholinesterases [19, 20], and antiviral [21,22,23] properties. Indole-sulfonamide is thought to be a suitable pharmacophore equivalent for substituting active sites in drug design since it undergoes substitution, primarily at the C-3 position, and has hydrophilic properties similar to the sulfonyl group[24]. Indole is a relatively weak basic chemical. This results from the delocalization of the nitrogen lone pair into the free-to-move-electronic system within the indole ring. As a result, the lone pair of electrons on nitrogen is not available for protonation and it becomes protonated at the C-3 position instead since this configuration is thermodynamically more stable due to the retention of aromaticity. As a result, it participates in a variety of chemical processes, including cycloaddition [25], carbon lithiation [26], oxidation [27], electrophilic substitution [28], and organometallic indole anion complexes. These processes are especially prevalent at the C-3 position. Indole is a solid at room temperature, and it naturally arises in human feces and gives out a fecal odor. However, it emits a floral smell at lesser concentrations and is a component of many perfumes, colognes, and coal tars [3]. In addition, indole has a role in several metabolic processes that take place within the body [29]. It controls spore formation, plasmid stability, drug resistance, biofilm development, and virulence, among other aspects of bacterial physiology [30].

Sulfonamides play a crucial role in pharmaceuticals and are physiologically active [31, 32]. Sulpha or sulfa medications are produced by the sulfonamide moiety's fundamental structure with corresponding reactants [33]. The greater potency demonstrated for developing novel characteristics of sulfonamides can also produce active derivatives. The chemistry of indole-carboxylic acid moiety [34] with that of sulfonamides can produce a pharmacologically active scaffold via different methods as reported in the literature.

Main text

Synthesis and biological evaluation of indolenyl sulfonamides

In 2010, by condensation, reacting indole-3-carboxaldehyde with various sulfonamides including sulfamethazine, sulfaguanidine, sulfadiazine, sulfisoxazole, sulfathiazole, sulfamethoxazole, and sulphanilamide seven novel indolenyl sulfonamides have been created [34]. The possible ligands for complexation with some specific divalent transition metal ions have been utilized as these manufactured compounds (copper, zinc, and cobalt). The findings of this inquiry were consistent with the indolenyl sulfonamides’ proposed structures. It has been proposed that certain functional groups, including azomethine or hetero-aromatics [35], present in these compounds showed biological activity, which may be the cause of the molecules’ increased hydrophobicity and liposolubility. Hence, all produced sulfonamides have potent antibacterial [36] and antifungal activities. This in turn increased the activity of the compounds and biological absorbance. This study played a pivotal role in synthesizing sulfonamide derivatives of indole with biological activities in a new way. The synthetic scheme, which was adopted for this study, is described below (Scheme 1).

Scheme 1
scheme 1

Synthesis of sulfonamide derivatives of indole

Synthesis of N 1-arylsulfonyl-3-piperazinyl indole derivatives

To create equivalent substituted N1-acetyl-3-piperazinyl indole derivatives, sodium triacetoxyborohydride, N-alkyl piperazines, and titanium isopropoxide were reacted with the derivatives of substituted N-alkyl acetyl indoxyl. Further, the deacetylated product was produced by deacetylating these indole derivatives with simple under reflux such as NaOH and KOH. The N1 sulfonylation of indole was accomplished using several distinct techniques. Dimethylformamide (DMF) and sodium hydride were determined to be the most effective combination. As an alternative, it was discovered that potassium hydroxide and tetrahydrofuran (THF) were equally useful for producing target chemicals in high quantities. The synthetic mechanism followed a multistep route as all the reactants were treated in alternative ways and a good yield was determined at the end of the reaction. The conditions were closely monitored by various techniques and finally, a successful characterization yielded the product. The final product was further checked for any biological activity and it showed a positive result [37] (Scheme 2).

Scheme 2
scheme 2

Compounds (7a-7t) synthesis

Synthesis of C2 acyl sulfonamide analogs of indole

In a study done by Anilkumar et al. [38], the majority of the analogs were successfully synthesized at the C2 location. Under simple conditions, hydrolysis of the C2 ester produced approximately quantifiable yields of the advanced intermediate. C2 acyl sulfonamide intermediates were produced after carbonyl di imidazole activation and then reacting alkylated sulfonamides. The last synthetic modification to generate analogs by demethylation in dioxane reflux was performed. A C2 acyl sulfamide analog was produced after treatment with sulfonylurea and a consecutive demethylation process. The formed sulfonamide-indole scaffold is highly potent in terms of biological significance. The schematic synthesis of the above-mentioned product is illustrated (Scheme 3).

Scheme 3
scheme 3

C-2 analogs general synthesis

Significance of Friedel–Craft reaction for the synthesis of indole sulfonamides

As possible anticancer medicines, Ratchanok and team created indole core structures using the Friedel–Craft reaction catalyzed by tungstosilicic acid hydrate in this study. Due to the catalyst's favorable environmental characteristics and simplicity, it has drawn a lot of attention. Here, the structure–activity relationship (SAR) of the synthesized indole analogs and their antiproliferative effect on cancer cell lines are reported [39]. The significance of this method was that the used Friedel–Craft reaction is utilized precisely to obtain a good yield. The mechanism of its synthesis was specific to producing a cytotoxic product with much wider applications as an agent. Its mechanism is environment-friendly and specific in action. Various methods including spectroscopic techniques [40] confirmed the formed product (Scheme 4).

Scheme 4
scheme 4

Synthesis of indole benzenesulfonamide derivatives (10–15)

Chiral squaramide-catalyzed cascade sulfa Michael addition reactions

Through an organocatalyzed addition process (Cascade sulfa Michael/Michael addition), Li et al. [41] discovered an enantioselective compound named (E)-3-aryl-2-(indol-3-ylcarbonyl) acrylonitriles. The catalysts used called chiral squaramide [42] helped to speed up this process and produced 3-substituted chiral indole scaffolds having 3 stereo centers of tetrahydrothiophene [43] in good to exceptional yields. With the best catalyst in place, further research was done to determine the best reaction conditions by examining the number of reactants, solvent interactions, and given temperature. The reaction was observed under optimal circumstances, and a noticeable impact was observed by the electronic interactions that the substituent had on the indole ring that resulted in an enantioselective product. The asymmetric reaction provided a highly specific environment for the reaction to grow in optimal conditions given the fact that the chiral indole derivatives were the ideal products (Scheme 5).

Scheme 5
scheme 5

Screening of different chiral organocatalysts

Two-step enantioselective synthesis of sulfonamide-indole derivatives via cascade cyclization reaction

Dixon and coworkers reported the synthetic route for the generation of highly enantioselective sulfonamide-indole derivatives by a cyclization cascade reaction involving two steps in the synthesis, i.e. hydroamination and catalyzed cyclization [44]. Observed yield for this product by using toluene as a solvent ranged between 85 and 96%, and the whole reaction mixture was refluxed for about 20 h at the temperature of 20 °C. This reaction gave pure enantioselective [45] products which were obtained in pure form. The importance of this reaction was the selectivity of the product, and the formation of a cyclized product with a higher yield was reported (Scheme 6).

Scheme 6
scheme 6

Indole-derived sulfonamide enantioselective synthesis

Sulfonamide-indole derivatives as antiproliferative agents: structure–activity relationship

Ruo-Jun Man outlined a synthetic route for many new derivatives of indole with sulfonamide moiety, assessed their anticancer [46] and anti-tubulin capabilities, investigated their effects on cell division, cell death, and microtubule dynamics, and examined the structure–activity link [47]. He looked at the links between structure and activity by adding different substituents to the benzene and indole rings. Electron-donating groups inside the benzene ring of the compounds showed stronger antiproliferative [48] along with tubulin depolymerization effects [49]. Molecular docking and the 3D-QSAR model, which might be used to create novel compounds with stronger tubulin inhibitory action, also offered further knowledge. An evaluation of the antiproliferative activities of the synthesized derivatives along with a comparison was made between different moieties. Different functional groups had specific actions (either as an activator or as a poison) and were recorded. The significance of this study was the activities shown by the compounds on a large scale such as antiproliferative activity (Scheme 7).

Scheme 7
scheme 7

Synthetic route for an indole-sulfonamide moiety

Analysis of antibacterial activities of indole-sulfonamide analogs

Recent work in 2017 described the production of sulfonamide analogs with the pyrimidine nucleus and tested them for cytotoxicity and antiamoebic activity [50]. Given the significance of these functional moieties, the author built a structure that included all the crucial components previously reported, with the idea that these structures would cooperate to increase therapeutic benefits. This work reported the synthesis of a novel compound named N-[5-(substituted-phenylsulfonyl)-1H-indol-3-yl] following the computational screening of 1, 3, and 4-thiadiazol-2-yl-substituted benzenesulfonamide that was also tested. Gram (+ ve & −ve) pathogens like S. epidermidis, E. coli, and P. mirabilis were the ones against which the synthesized analogs were assessed for their antibacterial activity. The results showed that every component showed a wide range of activity based on Lipinski’s rule of five. The artificial counterparts outperformed “Ciprofloxacin” in terms of their activity. The significance of this study is the involvement of the indole nuclei in generating desired sulfonamide scaffolds as products (Scheme 8).

Scheme 8
scheme 8

Systematic route adapted for the synthesis of compounds

Synthesis of bis- and tris-indole sulfonamides via alkylation

Chemists have previously reported new sulfonamide-indole derivatives containing bis-indoles and tris-indoles [39, 51]. The main reaction involved the alkylation followed by condensation of N-benzylsulfonyl halides with tryptamines that led to the production of several novel indole-sulfonamide derivatives in moderate to good yields (40–68%) [52]. The target protein, aromatase, and the generated indole derivatives were examined for their binding modes using molecular docking. Molecular docking was the main characterization technique to verify the formed compound. This study helped generate the indole-sulfonamide scaffolds via a simple alkylation method of the alkyl halides. The addition of benzaldehyde in this reaction gave different moieties as described in the scheme below. Bis-indoles and tris-indoles were obtained in very good yield, and this reaction paved the way for the scientist to explore more of such compounds (Scheme 9).

Scheme 9
scheme 9

Bis- and tris-indole derivatives with sulfonamide (11–44)

Analyzing IR stretching vibrations in indole-sulfonamide derivatives

3-Phenyl-5-sulfonamide-1H-indole-2-carbohydrazide synthesis was reported by a team of scientists led by Kübra Demir-Yazıcı who synthesized this compound from sulfanilamide by diazotization reaction [53]. The reaction involved the generation of diazonium salt followed by condensation, cyclization, and ultimately treating it with hydrazines [54]. Further processing of the material with the proper carbonyl component (ketone or aldehyde) produced hydrazone derivatives. Melting temperatures and spectrum analysis were used to describe newly synthesized hydrazones. Compounds IR spectra displayed NH stretching bands for hydrazine and sulfonamide groups at the indole ring at 3423 and 3138 cm−1. Strong C=O stretching bands in the range of 1643–685 cm−1, whereas the main compound as expected had a band at 1627 cm−1, confirming the novel compounds’ carbonyl functionalities. The sulfonamide group's symmetric and asymmetric SO2 stretching vibrations displayed IR band values in the 1344–1311 cm–1 and 1176–1147 cm–1 range, respectively [55]. The reactivity was checked, but due to the time-lapse, it was left over. However, it was seen that a few of the sulfonamide-indole scaffolds showed carbonic anhydrase activities that were later examined (Scheme 10).

Scheme 10
scheme 10

Synthesis via diazotization of indole-sulfonamide scaffolds

Synthesis of sulfonamide-indole compounds with aromatase-inhibitory activity

By reacting the suitable commercial indolamine and the commercial arylsulfonamide with N(CH3)3 in dry CH2CL2, holding the mixture for 2 h at the ice point, followed by 18 to 22 h standing at room temperature, the desired compounds were created[56]. Liquid chromatography provided the refined chemicals during the purification process. Aromatase inhibitors are used in the pharmaceutical management of postmenopausal women with early and metastatic breast cancer. Although aromatase inhibitors [57] are now used in medicine, generating new compounds with greater clinical efficacy and fewer negative effects is difficult. The docking study revealed that the active substances had interactions with the aromatase’s residues. The synthesized indole-sulfonamide derivatives [58] showed aromatase inhibition activity, and it was observed by studying the heme group interactions of the aromatase enzyme with indole nuclei. This research is very significant because for the very first time, the AI is used parallel to the chemistry, and the products were screened for future significance (Scheme 11).

Scheme 11
scheme 11

Sulfonamide-indole derivatives synthesis containing aromatase inhibition activity

Indole-sulfonamide derivatives as potential acetylcholinesterase inhibitors

A new class of indole-sulfonamide derivatives exhibiting strong acetylcholinesterase (AChEs) inhibition was prepared using hydrazine and pyridine [59]. The molecular docking research showed that newly synthesized drugs interacted with enzymes very well at the active site. The IC50 values were by the data collected and reflected strong AChEs inhibition [60]. The significance of this study was mainly associated with the different aromatic residues attached to the parent indole-sulfonamide moiety, each of which had a specific AChEs action. The amount and placement, which different functional groups occupied inside the benzene ring, differed between the chosen compounds. These results marked the beginning of the creation of an effective analog that can treat Alzheimer’s [61]. Further optimizing the most active molecules can create lead compounds. The simplicity of this study is reflected in the scheme illustrated below (Scheme 12).

Scheme 12
scheme 12

Sulfonamide analogs synthesis containing indole

Synthesis of sulfonamide-indole derivatives with carbonic anhydrase inhibition activity

Priti Singh and her team designed a new strategy to produce a new moiety of sulfonamide-indole derivatives with carbonic anhydrase inhibition activity [62]. This strategy provided a new route for the synthesis of desired products. Indole-3-sulfonamide derivatives were synthesized using this strategy. The main synthetic approach involved the N-alkylation of derivatives of 5-nitroindoles by using sodium hydride in a solvent of dimethylformamide (DMF) in the presence of anhydrous conditions [63]. Chlorosulfonic acid is used for the chlorosulfonation of the indole derivatives obtained in the previous step. N-alkylated indole-3-sulfonamide derivatives were synthesized in aqueous ammonia through an amination reaction with the amines [64]. In the presence of acetic acid and iron, the derivatives were reduced to obtain an intermediate that was then reacted with the isocyanates to get the desired product in good yield [65, 66]. Then the successful characterization was done, and it was revealed that the reaction obtained products in good amounts and the final product showed good biological activity, the carbonic anhydrase inhibition activity. The solvents and reagents used in this experiment were purified. The results were very much promising (Scheme 13).

Scheme 13
scheme 13

Carbonic anhydrase moieties of indole-sulfonamide

Synthesis and antimicrobial evaluation of sulfonamide-based indole derivatives

In the study carried out by Khushbu Agarwal et al., a starting material with the name of 1H-indole-2 carboxylic acid was used to synthesize several novel indole derivatives based on sulfonamides [67]. By using specific characterization techniques like 1H NMR [68], the structures of produced sulfonamide-based indole derivatives were verified. The most common indole derivative with acid was employed to carry out the reaction. The strategy involved the di-methylation of the N-alkylated indole-2-carboxylic acid [69], and then it was treated with methyl iodide (MeI) and K2CO3 leading to the formation of indole ester [70] which in the presence of tetrahydrofuran undergoes deprotection. The formed indole-acid derivative was then reacted with a commercially available reagent with the name of 4-ter-Butyloxycarbonyl (4-boc) piperidine amine followed by deblocking reaction to yield the final product [71]. The synthetic scheme shown below demonstrates the strategy that accompanied this reaction and how the desired products were obtained. The strategy is quite simple and easy to apply. The sulfonamide-indole derivatives were prepared by treating reactants in the presence of trimethylamine with alkyl sulfonamide halide in a very friendly environment. The produced indole-sulfonamide scaffolds showed to be effective antimicrobials. All of the produced derivatives exhibited positive activity against Gram Positive Staphylococcus aureus while showing inactivity against Bacillus megaterium, according to biological evaluation. Most of the products showed strong biological activity against Gram-negative bacteria excluding a few, while all of them showed an extremely strong effect against Klebsiella pneumonia. This study contributed to the field's ongoing indole derivative research (Scheme 14).

Scheme 14
scheme 14

Sulfonamide-indole-based derivatives synthesis

Conclusion

The current review emphasizes the synthesis and biological evaluation of indole-sulfonamide analogs from 2010 to 2023. The various synthetic routes including the Friedel–Craft reaction, cascade sulfa Michael addition reaction, enantioselective cascade cyclization reaction, and alkylation reactions are schematically visualized for the better understanding of drug chemists. The infrared stretching vibrations are also reported. Furthermore, pharmacological activities such as antiproliferative, antibacterial, and antimicrobial are demonstrated with corresponding structure–activity relationships. Additionally, some of their inhibitory effects like aromatase, acetylcholinesterase, and carbonic anhydrase are also described. Using the literature findings mentioned above as a summary, we can state that indole exhibits a wide range of biological activities. Future research in the production of indole scaffolds is growing, and this literature will give a direction for drug design chemists to synthesize a new variety of active drugs.

Availability of data and materials

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

DMF:

Dimethylformamide

THF:

Tetrahydrofuran

SAR:

Structure–activity relationship

3D-QSAR:

Three-dimensional quantitative structure–activity relationship

AIs:

Aromatase inhibitors

AChEs:

Acetylcholinesterase

1H NMR:

Proton nuclear magnetic resonance

References

  1. Kumar S (2020) A brief review of the biological potential of indole derivatives. Future J Pharm Sci 6(1):1–19

    CAS  Google Scholar 

  2. Van Order R, Lindwall H (1942) Indole. Chem Rev 30(1):69–96

    Article  Google Scholar 

  3. Kumari A, Singh RK (2019) Medicinal chemistry of indole derivatives: current to future therapeutic prospectives. Bioorgn Chem 89:103021

    Article  CAS  Google Scholar 

  4. Jia Y et al (2020) Current scenario of indole derivatives with potential anti-drug-resistant cancer activity. Eur J Med Chem 200:112359

    Article  CAS  PubMed  Google Scholar 

  5. Kaushik NK et al (2013) Biomedical importance of indoles. Molecules 18(6):6620–6662

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Shaker AM et al (2020) Synthesis and biological evaluation of 2-(4-methylsulfonyl phenyl) indole derivatives: multi-target compounds with dual antimicrobial and anti-inflammatory activities. BMC Chem 14(1):1–15

    Article  Google Scholar 

  7. Al-Ostoot FH et al (2020) Design-based synthesis, molecular docking analysis of an anti-inflammatory drug, and geometrical optimization and interaction energy studies of an indole acetamide derivative. J Mol Struct 1202:127244

    Article  CAS  Google Scholar 

  8. Reddy GS, Pal M (2021) Indole derivatives as anti-tubercular agents: an overview on their synthesis and biological activities. Curr Med Chem 28(22):4531–4568

    Article  CAS  PubMed  Google Scholar 

  9. Thanikachalam PV et al (2019) An insight into the medicinal perspective of synthetic analogs of indole: a review. Eur J Med Chem 180:562–612

    Article  CAS  PubMed  Google Scholar 

  10. Khan A et al (2018) Enhancement in anti-tubercular activity of indole based thiosemicarbazones on complexation with copper (I) and silver (I) halides: structure elucidation, evaluation and molecular modelling. Bioorgn Chem 80:303–318

    Article  CAS  Google Scholar 

  11. Nomura S et al (2014) Novel indole-N-glucoside, TA-1887 as a sodium glucose cotransporter 2 inhibitor for treatment of type 2 diabetes. ACS Med Chem Lett 5(1):51–55

    Article  CAS  PubMed  Google Scholar 

  12. Omar F et al (2021) Plant-based indole alkaloids: a comprehensive overview from a pharmacological perspective. Molecules 26(8):2297

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Al-Salahi R et al (2018) Screening and evaluation of antioxidant activity of some 1, 2, 4-triazolo [1, 5-a] quinazoline derivatives. Future Med Chem 10(4):379–390

    Article  CAS  PubMed  Google Scholar 

  14. Bentz EN et al (2018) Intrinsic antioxidant potential of the aminoindole structure: a computational kinetics study of tryptamine. J Phys Chem B 122(24):6386–6395

    Article  CAS  PubMed  Google Scholar 

  15. Dadashpour S, Emami S (2018) Indole in the target-based design of anticancer agents: a versatile scaffold with diverse mechanisms. Eur J Med Chem 150:9–29

    Article  CAS  PubMed  Google Scholar 

  16. Dhiman A, Sharma R, Singh RK (2022) Target-based anticancer indole derivatives and insight into structure-activity relationship: a mechanistic review update (2018–2021). Acta Pharm Sin B. https://doi.org/10.1016/j.apsb.2022.03.021

    Article  PubMed  PubMed Central  Google Scholar 

  17. Balakrishnan N et al (2023) Influence of Indole-N substitution of thiosemicarbazones in cationic Ru (II)(η6-Benzene) complexes on their anticancer activity. Organometallics 42(3):259–275

    Article  CAS  Google Scholar 

  18. Ali I et al (2018) Facile synthesis of indole heterocyclic compounds based micellar nano anti-cancer drugs. RSC Adv 8(66):37905–37914

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bingul M, Ercan S, Boga M (2020) The design of novel 4, 6-dimethoxyindole based hydrazide-hydrazones: Molecular modeling, synthesis and anticholinesterase activity. J Mol Struct 1213:128202

    Article  CAS  Google Scholar 

  20. Mahmoud E et al (2022) Recent progress in biologically active indole hybrids: a mini review. Pharmacol Rep 74(4):570–582

    Article  CAS  PubMed  Google Scholar 

  21. Sulthana S, Pandian P (2019) A review on Indole and Benzothiazole derivatives its importance. J Drug Deliv Ther 9(1-s):505–509

    Article  CAS  Google Scholar 

  22. Chadha N, Silakari O (2018) Indoles: as multitarget directed ligands in medicinal chemistry. In: Key heterocycle cores for designing multitargeting molecules, Elsevier, pp 285–321

  23. Ramesh D et al (2020) Indole chalcones: design, synthesis, in vitro and in silico evaluation against Mycobacterium tuberculosis. Eur J Med Chem 198:112358

    Article  CAS  PubMed  Google Scholar 

  24. Namasivayam V et al (2018) The journey of HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) from lab to clinic. J Med Chem 62(10):4851–4883

    Article  PubMed  PubMed Central  Google Scholar 

  25. Lynch SM, Bur SK, Padwa A (2002) Intramolecular amidofuran cycloadditions across an indole π-bond: an efficient approach to the aspidosperma and strychnos ABCE core. Org Lett 4(26):4643–4645

    Article  CAS  PubMed  Google Scholar 

  26. Bergman J, Venemalm L (1992) Efficient synthesis of 2-chloro-, 2-bromo-, and 2-iodoindole. J Org Chem 57(8):2495–2497

    Article  CAS  Google Scholar 

  27. Ximenes VF, Campa A, Catalani LH (2001) The oxidation of indole derivatives catalyzed by horseradish peroxidase is highly chemiluminescent. Arch Biochem Biophys 387(2):173–179

    Article  CAS  PubMed  Google Scholar 

  28. Shabica A et al (1946) Improved syntheses of indole-3-aldehyde. J Am Chem Soc 68(7):1156–1157

    Article  CAS  Google Scholar 

  29. Ma Q, Zhang X, Qu Y (2018) Biodegradation and biotransformation of indole: advances and perspectives. Front Microbiol 9:2625

    Article  PubMed  PubMed Central  Google Scholar 

  30. Chadha N, Silakari O (2017) Indoles as therapeutics of interest in medicinal chemistry: bird’s eye view. Eur J Med Chem 134:159–184

    Article  CAS  PubMed  Google Scholar 

  31. Apaydın S, Török M (2019) Sulfonamide derivatives as multi-target agents for complex diseases. Bioorgn Med Chem Lett 29(16):2042–2050

    Article  Google Scholar 

  32. Ovung A, Bhattacharyya J (2021) Sulfonamide drugs: structure, antibacterial property, toxicity, and biophysical interactions. Biophys Rev 13(2):259–272

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Taha M et al (2021) Evaluation and docking of indole sulfonamide as a potent inhibitor of α-glucosidase enzyme in streptozotocin–induced diabetic albino wistar rats. Bioorgn Chem 110:104808

    Article  CAS  Google Scholar 

  34. Chohan ZH et al (2010) Identification of antibacterial and antifungal pharmacophore sites for potent bacteria and fungi inhibition: indolenyl sulfonamide derivatives. Eur J Med Chem 45(3):1189–1199

    Article  CAS  PubMed  Google Scholar 

  35. Garaj V et al (2005) Carbonic anhydrase inhibitors: novel sulfonamides incorporating 1, 3, 5-triazine moieties as inhibitors of the cytosolic and tumour-associated carbonic anhydrase isozymes I, II and IX. Bioorgn Med Chem Lett 15(12):3102–3108

    Article  CAS  Google Scholar 

  36. Verma SK et al (2020) Antibacterial activities of sulfonyl or sulfonamide containing heterocyclic derivatives and its structure-activity relationships (SAR) studies: a critical review. Bioorgn Chem 105:104400

    Article  Google Scholar 

  37. Nirogi RV et al (2011) Indole-3-piperazinyl derivatives: novel chemical class of 5-HT6 receptor antagonists. Bioorgn Med Chem Lett 21(1):346–349

    Article  CAS  Google Scholar 

  38. Anilkumar GN et al (2012) II. Novel HCV NS5B polymerase inhibitors: discovery of indole C2 acyl sulfonamides. Bioorgn Med Chem Lett 22(1):713–717

    Article  CAS  Google Scholar 

  39. Pingaew R et al (2013) Synthesis and structure–activity relationship of mono-indole-, bis-indole-, and tris-indole-based sulfonamides as potential anticancer agents. Mol Divers 17:595–604

    Article  CAS  PubMed  Google Scholar 

  40. Ahmed A et al (2023) Synthesis and spectroscopic characterization of nicotinaldehyde based derivatives: SC-XRD, linear and non-linear optical studies. J Mol Struct 1273:134236

    Article  CAS  Google Scholar 

  41. Li Y-H et al (2014) Asymmetric synthesis of 3-substituted indole derivatives containing tetrahydrothiophene via cascade sulfa-Michael/Michael additions catalyzed by a chiral squaramide catalyst. Tetrahedron Asymmetry 25(23):1513–1519

    Article  CAS  Google Scholar 

  42. Alemán J et al (2011) Squaramides: bridging from molecular recognition to bifunctional organocatalysis. Chem A Eur J 17(25):6890–6899

    Article  Google Scholar 

  43. Meninno S, Croce G, Lattanzi A (2013) Asymmetric synthesis of trisubstituted tetrahydrothiophenes bearing a quaternary stereocenter via double Michael reaction involving dynamic kinetic resolution. Org Lett 15(13):3436–3439

    Article  CAS  PubMed  Google Scholar 

  44. Gregory AW et al (2013) Gold and BINOL-phosphoric acid catalyzed enantioselective hydroamination/N-sulfonyliminium cyclization cascade. Org Lett 15(17):4330–4333

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Muratore ME et al (2009) Enantioselective Brønsted acid-catalyzed N-acyliminium cyclization cascades. J Am Chem Soc 131(31):10796–10797

    Article  CAS  PubMed  Google Scholar 

  46. Hadfield JA et al (2003) Tubulin and microtubules as targets for anticancer drugs. Prog Cell Cycle Res 5:309–326

    PubMed  Google Scholar 

  47. Man R-J et al (2016) Synthesis and biological evaluation of novel indole derivatives containing sulfonamide scaffold as potential tubulin inhibitor. MedChemComm 7(9):1759–1767

    Article  CAS  Google Scholar 

  48. Chripkova M, Zigo F, Mojzis J (2016) Antiproliferative effect of indole phytoalexins. Molecules 21(12):1626

    Article  PubMed  PubMed Central  Google Scholar 

  49. Brancale A, Silvestri R (2007) Indole, a core nucleus for potent inhibitors of tubulin polymerization. Med Res Rev 27(2):209–238

    Article  CAS  PubMed  Google Scholar 

  50. Roouf Bhat A et al (2013) Synthesis, characterization, and anti-amoebic activity of N-(Pyrimidin-2-yl) benzenesulfonamide derivatives. Chem Biodivers 10(12):2267–2277

    Article  PubMed  Google Scholar 

  51. Pingaew R et al (2012) Synthesis and cytotoxicity of novel 2, 2′-bis-and 2, 2′, 2 ″-tris-indolylmethanes-based bengacarboline analogs. Arch Pharmacal Res 35:949–954

    Article  CAS  Google Scholar 

  52. Ratchanok Pingaew PM, Prachayasittikul V, Prachayasittikul S, Ruchirawat S, Prachayasitti V (2018) Synthesis, molecular docking, and QSAR study of sulfonamide-based indoles as aromatase inhibitors. Eur J Med Chem 143:1604–1615

    Article  PubMed  Google Scholar 

  53. Demir-Yazıcı K, Bua S, Akgüneş NM, Akdemir A, Supuran CT, Güzel-Akdemir Ö (2019) Indole-based hydrazones containing a sulfonamide moiety as selective inhibitors of tumor-associated human carbonic anhydrase isoforms IX and XII. Int J Mol Sci 20(9):2354

    Article  PubMed  PubMed Central  Google Scholar 

  54. Güzel Ö, Temperini C, Innocenti A, Scozzafava A, Salman A, Supuran CT (2008) Carbonic anhydrase inhibitors. Interaction of 2-(hydrazinocarbonyl)-3-phenyl-1H-indole-5-sulfonamide with 12 mammalian isoforms: kinetic and X-ray crystallographic studies. Bioorgn Med Chem Lett 18:152–158

    Article  Google Scholar 

  55. Ergenç N, Salman A, Gürsoy A, Pharmazie G (1990) Synthesis and antifungal evaluation of some 3-phenyl-2, 5-disubstituted indoles derived from new ethyl-2-benzyl-2-[N-(aryl)hydrazono] ethanoates. Die Pharm 45:346–347

    Google Scholar 

  56. Fantacuzzi M, De Filippis B, Gallorini M, Ammazzalorso A, Giampietro L, Maccallini C, Aturki Z, Donati E, Ibrahim RS, Shawky E, Cataldi A, Amoroso R (2020) Synthesis, biological evaluation, and docking study of indole aryl sulfonamides as aromatase inhibitors. Eur J Med Chem 185:111815

    Article  CAS  PubMed  Google Scholar 

  57. Uddin A et al (2019) Encapsulation of organic and perovskite solar cells: a review. Coatings 9(2):65

    Article  Google Scholar 

  58. Caporuscio F, Rastelli G, Imbriano C, Del Rio A (2011) Structure-based design of potent aromatase inhibitors by high-throughput docking. J Med Chem 54(12):4006–4017

    Article  CAS  PubMed  Google Scholar 

  59. Taha M, Alshamrani FJ, Rahim F, Uddin N, Chigurupati S, Almandil NB, Farooq RK, Iqbal N, Aldubayan M, Venugopal V, Khan KM (2021) Synthesis, characterization, biological evaluation, and kinetic study of indole base sulfonamide derivatives as acetylcholinesterase inhibitors in search of potent anti-Alzheimer agent. J King Saud Univ Sci 33(3):101401

    Article  Google Scholar 

  60. Insaf Filali JB, Znati M, Garah F-E, Jannet HB (2015) Synthesis of new isoxazoline derivatives from harmine and evaluation of their anti-Alzheimer, anti-cancer and anti-inflammatory activities. J Enzyme Inhib Med Chem 30(3):371–376

    Article  PubMed  Google Scholar 

  61. Rayala Swetha DK, Gupta SK, Ganeshpurkar A, Singh R, Gutti G, Kumar D, Jana S, Krishnamurthy S, Singh SK (2019) Multifunctional hybrid sulfonamides as novel therapeutic agents for Alzheimer’s disease. Future Med Chem 11(24):3161–3178

    Article  PubMed  Google Scholar 

  62. Singh P, Choli A, Swain B, Angeli A, Sahoo SK, Yaddanapudi VM, Supuran CT, Arifuddin M (2022) Design and development of novel series of indole-3-sulfonamide ureido derivatives as selective carbonic anhydrase II inhibitors. Arch Pharm 355(1):2100333

    Article  CAS  Google Scholar 

  63. Carmine Ostacolo VDS, Lauro G, Pepe G, Musella S, Ciaglia T, Vestuto V, Autore G, Bifulco G, Marzocco S, Campiglia P, Gomez-Monterrey IM, Bertamino A (2019) Identification of an indol-based multi-target kinase inhibitor through phenotype screening and target fishing using inverse virtual screening approach. Eur J Med Chem 167:61–75

    Article  PubMed  Google Scholar 

  64. Swain B, Angeli A, Angapelly S, Thacker PS, Singh P, Supuran CT, Arifuddin M (2019) Synthesis of a new series of 3-functionalised-1-phenyl-1, 2, 3-triazole sulfamoylbenzamides as carbonic anhydrase I, II, IV and IX inhibitors. J Enzyme Inhib Med Chem 34(1):1199–1209

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Thacker PS, Srikanth D, Angeli A, Singh P, Chinchilli KK, Arifuddin M, Supuran CT (2021) Coumarin-thiourea hybrids show potent carbonic anhydrase IX and XIII inhibitory action. ChemMedChem 16(8):1252–1256

    Article  CAS  PubMed  Google Scholar 

  66. Kurt BZ, Kandas NO, Dag A, Sonmez F, Kucukislamoglu M (2020) Synthesis and biological evaluation of novel coumarin-chalcone derivatives containing urea moiety as potential anticancer agents. Arab J Chem 13(1):1120–1129

    Article  CAS  Google Scholar 

  67. Agrawal K, Patel T, Patel R (2023) Synthesis, biological activity of newly designed sulfonamide based indole derivative as anti-microbial agent. Future J Pharm Sci 9(1):17

    Article  Google Scholar 

  68. Abdul Raouf MA, Khalid M, Ahmed A, Safdar K, Khan MU, Assiri MA, Imran M (2023) Exploration of electronic and non-linear optical properties of novel 4-Aryl-2-methylpyridine based compounds synthesized via high-yielding Pd (0) catalysed reaction. J Mol Struct 1274:134469

    Article  Google Scholar 

  69. Pillaiyar Thanigaimalai SK, Yamamoto T, Koiwai Y, Taguchi A, Takayama K, Yakushiji F, Akaji K, Chen S-E, Naser-Tavakolian A, Schön A, Freire E, Hayashi Y (2013) Development of potent dipeptide-type SARS-CoV 3CL protease inhibitors with novel P3 scaffolds: design, synthesis, biological evaluation, and docking studies. Eur J Med Chem 68:372–384

    Article  PubMed  PubMed Central  Google Scholar 

  70. Eduard Dolušić PL, Blanc S, Sapunaric F, Norberg B, Moineaux L, Colette D, Stroobant V, Pilotte L, Colau D, Ferain T, Fraser G, Galeni M, Frère J-M, Masereel B, Van den Eynde B, Wouters J, Frédérick R (2011) Indol-2-yl ethanones as novel indoleamine 2,3-dioxygenase (IDO) inhibitors. Bioorgn Med Chem 19(4):1550–1561

    Article  Google Scholar 

  71. Sei-ichi Tanuma KK, Oyama T, Yoshimori A, Shibasaki Y, Asawa Y, Yamazaki H, Makino K, Okazawa M, Ogino Y, Sakamoto Y, Nomura M, Sato A, Abe H, Nakamura H, Takahashi H, Tanuma N, Uchiumi F (2020) Structural basis of beneficial design for effective nicotinamide phosphoribosyltransferase inhibitors. Molecules 25(16):3633

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was not supported by any funding agencies.

Author information

Authors and Affiliations

Authors

Contributions

IM contributed to the design of the study and drafted the paper; AA contributed to drafting the paper and critical revision of the article; and all authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Irfan Mushtaq or Adnan Ahmed.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The authors declare no conflict of interest.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

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

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mushtaq, I., Ahmed, A. Synthesis of biologically active sulfonamide-based indole analogs: a review. Futur J Pharm Sci 9, 46 (2023). https://doi.org/10.1186/s43094-023-00500-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-023-00500-5

Keywords