We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Tetrahydroquinoline: an efficient scaffold as mTOR inhibitor for the treatment of lung cancer

    Udit Chaube

    *Author for correspondence: Tel.: +91 797 165 2718;

    E-mail Address: uditchoube@gmail.com

    Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India

    ,
    Rajdeep Dey

    Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India

    ,
    Suman Shaw

    Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India

    ,
    Bhumika D Patel

    Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India

    &
    Hardik G Bhatt

    Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India

    Published Online:https://doi.org/10.4155/fmc-2022-0204

    Efforts have been made to find an efficient scaffold (and its substitution) that can be used for the treatment of lung cancer via mTOR inhibition. A detailed literature search was carried out for previously reported mTOR inhibitors. The present review is focused on lung cancer; therefore, descriptions of some mTOR inhibitors that are currently in clinical trials for the treatment of lung cancer are provided. Based on previous research findings, tetrahydroquinoline was found to be the most efficient scaffold to be explored for the treatment of lung cancer. A possible efficient substitution of the tetrahydroquinoline scaffold could also be beneficial for the treatment of lung cancer.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Srivastava S, Koay EJ, Borowsky AD et al. Cancer overdiagnosis: a biological challenge and clinical dilemma. Nat. Rev. Cancer 19(6), 349–358 (2019). • Definition and characteristics of malignant cells.
    • 2. Cancer Stat Facts: Lung and Bronchus Cancer. https://seer.cancer.gov/statfacts/html/lungb.html#content
    • 3. Ferlay J, Shin H-R, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer 127(12), 2893–2917 (2010).
    • 4. Ridge C, McErlean A, Ginsberg M. Epidemiology of lung cancer. Semin. Interv. Radiol. 30(02), 093–098 (2013). •• Statistics and the significance of research in lung cancer.
    • 5. Jiang M, Li X, Quan X, Li X, Zhou B. Single nucleotide polymorphisms in HMGB1 correlate with lung cancer risk in the northeast Chinese Han population. Molecules 23(4), 832 (2018).
    • 6. Wood SL, Pernemalm M, Crosbie PA, Whetton AD. Molecular histology of lung cancer: from targets to treatments. Cancer Treat. Rev. 41(4), 361–375 (2015).
    • 7. Chaube U, Chhatbar D, Bhatt H. 3D-QSAR, molecular dynamics simulations and molecular docking studies of benzoxazepine moiety as mTOR inhibitor for the treatment of lung cancer. Bioorg. Med. Chem. Lett. 26(3), 864–874 (2016). •• Knowledge-based pharmacophore and virtual screening suggested tetrahydroquinoline (THQ) is one of the best choices for mTOR inhibitors.
    • 8. Kan Z, Jaiswal BS, Stinson J et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466(7308), 869–873 (2010).
    • 9. Govindan R, Ding L, Griffith M et al. Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell 150(6), 1121–1134 (2012).
    • 10. Li H, Wang S, Takayama K et al. Nicotine induces resistance to erlotinib via cross-talk between α 1 nAChR and EGFR in the non-small cell lung cancer xenograft model. Lung Cancer 88(1), 1–8 (2015).
    • 11. Yip PY. Phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin (PI3K-Akt-mTOR) signaling pathway in non-small cell lung cancer. Transl. Lung Cancer Res. 4(2), 165–176 (2015).
    • 12. Fumarola C, Bonelli MA, Petronini PG, Alfieri RR. Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer. Biochem. Pharmacol. 90(3), 197–207 (2014).
    • 13. Kaur A, Sharma S. Mammalian target of rapamycin (mTOR) as a potential therapeutic target in various diseases. Inflammopharmacology 25(3), 293–312 (2017).
    • 14. Dos D Sarbassov, Ali SM, Kim D-H et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr. Biol. 14(14), 1296–1302 (2004).
    • 15. Kim D-H, Sarbassov DD, Ali SM et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 110(2), 163–175 (2002).
    • 16. Sancak Y, Thoreen CC, Peterson TR et al. PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase. Mol. Cell. 25(6), 903–915 (2007).
    • 17. Peterson TR, Laplante M, Thoreen CC et al. DEPTOR Is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival. Cell 137(5), 873–886 (2009). • Describes mTORC1 and mTORC2 pathways to regulate cell growth, proliferation and survival and a detailed description of DEPTOR.
    • 18. Zhao Y, Xiong X, Sun Y. DEPTOR, an mTOR inhibitor, is a physiological substrate of SCFβTrCP E3 ubiquitin ligase and regulates survival and autophagy. Mol. Cell. 44(2), 304–316 (2011).
    • 19. Xu T, Sun D, Chen Y, Ouyang L. Targeting mTOR for fighting diseases: a revisited review of mTOR inhibitors. Eur. J. Med. Chem. 199, 112391 (2020). • Describes the PI3K/AKT/mTOR pathways and clinical development of some rapalogs.
    • 20. Asati V, Mahapatra DK, Bharti SK. PI3K/Akt/mTOR and Ras/Raf/MEK/ERK signaling pathways inhibitors as anticancer agents: structural and pharmacological perspectives. Eur. J. Med. Chem. 109, 314–341 (2016).
    • 21. Reddy GL, Guru SK, Srinivas M et al. Synthesis of 5-substituted-1H-pyrazolo[4,3-d]pyrimidin-7(6H)-one analogs and their biological evaluation as anticancer agents: mTOR inhibitors. Eur. J. Med. Chem. 80, 201–208 (2014).
    • 22. Polivka J, Janku F. Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol. Ther. 142(2), 164–175 (2014).
    • 23. Zhang Y-J, Duan Y, Zheng XFS. Targeting the mTOR kinase domain: the second generation of mTOR inhibitors. Drug Discov. Today 16(7–8), 325–331 (2011).
    • 24. Jerusalem G, Rorive A, Collignon J. Use of mTOR inhibitors in the treatment of breast cancer: an evaluation of factors that influence patient outcomes. Breast Cancer Targets Ther. 43 (2014).
    • 25. Yang H, Rudge DG, Koos JD, Vaidialingam B, Yang HJ, Pavletich NP. mTOR kinase structure, mechanism and regulation. Nature 497(7448), 217–223 (2013).
    • 26. Chen X, Liu M, Tian Y et al. Cryo-EM structure of human mTOR complex 2. Cell Res. 28(5), 518–528 (2018).
    • 27. Chresta CM, Davies BR, Hickson I et al. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res. 70(1), 288–298 (2010).
    • 28. Liu Q, Chang JW, Wang J et al. Discovery of 1-(4-(4-propionylpiperazin-1-yl)-3-(trifluoromethyl)phenyl)-9-(quinolin-3-yl)benzo[h] [1,6]naphthyridin-2(1 H)-one as a highly potent, selective mammalian target of rapamycin (mTOR) inhibitor for the treatment of cancer. J. Med. Chem. 53(19), 7146–7155 (2010).
    • 29. Liu Q, Wang J, Kang SA et al. Discovery of 9-(6-aminopyridin-3-yl)-1-(3-(trifluoromethyl)phenyl)benzo[h] [1,6]naphthyridin-2(1 H)-one (Torin2) as a potent, selective, and orally available mammalian target of rapamycin (mTOR) inhibitor for treatment of cancer. J. Med. Chem. 54(5), 1473–1480 (2011).
    • 30. Sun S-Y. mTOR kinase inhibitors as potential cancer therapeutic drugs. Cancer Lett. 340(1), 1–8 (2013).
    • 31. Neuhaus P. mTOR inhibitors: an overview. Liver Transpl. 7(6), 473–484 (2001). • Describes the mechanism of action of mTOR-selective small-molecule inhibitors.
    • 32. Alqasoumi SI, Al-Taweel AM, Alafeefy AM, Ghorab MM, Noaman E. Discovering some novel tetrahydroquinoline derivatives bearing the biologically active sulfonamide moiety as a new class of antitumor agents. Eur. J. Med. Chem. 45(5), 1849–1853 (2010).
    • 33. Parmenon C, Guillard J, Caignard D-H et al. 4,4-Dimethyl-1,2,3,4-tetrahydroquinoline-based PPARα/γ agonists. Part. II: synthesis and pharmacological evaluation of oxime and acidic head group structural variations. Bioorg. Med. Chem. Lett. 19(10), 2683–2687 (2009).
    • 34. Pagliero RJ, Lusvarghi S, Pierini AB, Brun R, Mazzieri MR. Synthesis, stereoelectronic characterization and antiparasitic activity of new 1-benzenesulfonyl-2-methyl-1,2,3,4-tetrahydroquinolines. Bioorg. Med. Chem. 18(1), 142–150 (2010).
    • 35. Di Fabio R, Alvaro G, Bertani B et al. Chiral tetrahydroquinoline derivatives as potent anti-hyperalgesic agents in animal models of sustained inflammation and chronic neuropathic pain. Bioorg. Med. Chem. Lett. 17(5), 1176–1180 (2007).
    • 36. Nishiyama T, Hashiguchi Y, Sakata T, Sakaguchi T. Antioxidant activity of the fused heterocyclic compounds, 1,2,3,4-tetrahydroquinolines, and related compounds–effect of ortho-substituents. Polym. Degrad. Stab. 79(2), 225–230 (2003).
    • 37. Zhang J, Zhan P, Wu J et al. Synthesis and biological evaluation of novel 5-alkyl-2-arylthio-6-((3,4-dihydroquinolin-1(2H)-yl)methyl)pyrimidin-4(3H)-ones as potent non-nucleoside HIV-1 reverse transcriptase inhibitors. Bioorg. Med. Chem. 19(14), 4366–4376 (2011).
    • 38. Guo T, Gu H, Hobbs DW et al. Design, synthesis, and evaluation of tetrahydroquinoline and pyrrolidine sulfonamide carbamates as γ-secretase inhibitors. Bioorg. Med. Chem. Lett. 17(11), 3010–3013 (2007).
    • 39. Rano TA, Sieber-McMaster E, Pelton PD, Yang M, Demarest KT, Kuo G-H. Design and synthesis of potent inhibitors of cholesteryl ester transfer protein (CETP) exploiting a 1,2,3,4-tetrahydroquinoline platform. Bioorg. Med. Chem. Lett. 19(9), 2456–2460 (2009).
    • 40. Spadoni G, Bedini A, Lucarini S et al. Highly potent and selective MT2 melatonin receptor full agonists from conformational analysis of 1-benzyl-2-acylaminomethyl-tetrahydroquinolines. J. Med. Chem. 58(18), 7512–7525 (2015).
    • 41. Nagata N, Kawai K, Nakanishi I. Subtle structural changes in tetrahydroquinolines, a new class of nonsteroidal selective androgen receptor modulators, induce different functions. J. Chem. Inf. Model. 52(8), 2257–2264 (2012).
    • 42. Nagata N, Miyakawa M, Amano S, Furuya K, Yamamoto N, Inoguchi K. Design and synthesis of tricyclic tetrahydroquinolines as a new series of nonsteroidal selective androgen receptor modulators (SARMs). Bioorg. Med. Chem. Lett. 21(6), 1744–1747 (2011).
    • 43. Nagata N, Furuya K, Oguro N et al. Lead evaluation of tetrahydroquinolines as nonsteroidal selective androgen receptor modulators for the treatment of osteoporosis. ChemMedChem 9(1), 197–206 (2014).
    • 44. Kakeya H, Kuranaga T. Precious microorganisms as productive resources: marine-derived microorganisms and combined-culture. In: Comprehensive Natural Products III. 7, 430–456 (2020). https://linkinghub.elsevier.com/retrieve/pii/B9780124095472146931
    • 45. Nammalwar B, Bunce R. Recent syntheses of 1,2,3,4-tetrahydroquinolines, 2,3-dihydro-4(1H)-quinolinones and 4(1H)-quinolinones using domino reactions. Molecules. 19(1), 204–232 (2013).
    • 46. Jacquemond-Collet I, Bessière J-M, Hannedouche S, Bertrand C, Fourasté I, Moulis C. Identification of the alkaloids of Galipea officinalis by gas chromatography-mass spectrometry: ALKALOIDS IN GALIPEA OFFICINALIS. Phytochem. Anal. 12(5), 312–319 (2001).
    • 47. Houghton PJ, Woldemariam TZ, Watanabe Y, Yates M. Activity against mycobacterium tuberculosis of alkaloid constituents of angostura bark, Galipea officinalis. Planta Med. 65(3), 250–254 (1999).
    • 48. Jacquemond-Collet I, Benoit-Vical F, Valentin A, Stanislas E, Mallié M, Fourasté I. Antiplasmodial and cytotoxic activity of galipinine and other tetrahydroquinolines from Galipea officinalis. Planta Med. 68(1), 68–69 (2002).
    • 49. Sugiyama R, Nishimura S, Ozaki T, Asamizu S, Onaka H, Kakeya H. 5-Alkyl-1, 2, 3, 4-tetrahydroquinolines, new membrane-interacting lipophilic metabolites produced by combined culture of Streptomyces nigrescens and Tsukamurella pulmonis. Org. Lett. 17(8), 1918–1921 (2015).
    • 50. Jo H, Choi M, Kumar AS et al. Development of novel 1,2,3,4-tetrahydroquinoline scaffolds as potent NF-κB inhibitors and cytotoxic agents. ACS Med. Chem. Lett. 7(4), 385–390 (2016).
    • 51. Chung P-Y, Tang JC-O, Cheng C-H et al. Synthesis of hexahydrofuro[3,2-c]quinoline, a martinelline type analogue and investigation of its biological activity. SpringerPlus 5(1), 271 (2016).
    • 52. Wang X-F, Wang S-B, Ohkoshi E et al. N-Aryl-6-methoxy-1,2,3,4-tetrahydroquinolines: a novel class of antitumor agents targeting the colchicine site on tubulin. Eur. J. Med. Chem. 67, 196–207 (2013).
    • 53. Kouznetsov VV, Merchan-Arenas DR, Tangarife-Castaño V, Correa-Royero J, Betancur-Galvis L. Synthesis and cytotoxic evaluation of novel 2-aryl-4-(4-hydroxy-3-methoxyphenyl)-3-methyl-6,7-methylendioxy-1,2,3,4-tetrahydroquinolines, podophyllotoxin-like molecules. Med. Chem. Res. 25(3), 429–437 (2016).
    • 54. Madácsi R, Kanizsai I, Fehér L et al. Aromatic sulfonamides containing a condensed piperidine moiety as potential oxidative stress-inducing anticancer agents. Med. Chem. 9(7), 911–919 (2013).
    • 55. Chen C, Zingales S, Wang T et al. Synthesis and in vitro evaluation of 4-substituted furano[3,2-c] tetrahydroquinolines as potential anti-cancer agents. J. Enzyme Inhib. Med. Chem. 31(5), 853–858 (2016).
    • 56. Chaube UJ, Rawal R, Jha AB, Variya B, Bhatt HG. Design and development of tetrahydro-quinoline derivatives as dual mTOR-C1/C2 inhibitors for the treatment of lung cancer. Bioorg. Chem. 106, 104501 (2021).
    • 57. García-Echeverría C. Allosteric and ATP-competitive kinase inhibitors of mTOR for cancer treatment. Bioorg. Med. Chem. Lett. 20(15), 4308–4312 (2010).
    • 58. Pike KG, Malagu K, Hummersone MG et al. Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: the discovery of AZD8055 and AZD2014. Bioorg. Med. Chem. Lett. 23(5), 1212–1216 (2013).
    • 59. Pei Z, Blackwood E, Liu L et al. Discovery and biological profiling of potent and selective mTOR inhibitor GDC-0349. ACS Med. Chem. Lett. 4(1), 103–107 (2013).
    • 60. Bhagwat SV, Gokhale PC, Crew AP et al. Preclinical characterization of OSI-027, a potent and selective inhibitor of mTORC1 and mTORC2: distinct from rapamycin. Mol. Cancer Ther. 10(8), 1394–1406 (2011).
    • 61. Guo Q, Yu C, Zhang C et al. Highly selective, potent, and oral mTOR inhibitor for treatment of cancer as autophagy inducer. J. Med. Chem. 61(3), 881–904 (2018).
    • 62. Chen D, Soh CK, Goh WH, Wang H. Design, synthesis, and preclinical evaluation of fused pyrimidine-based hydroxamates for the treatment of hepatocellular carcinoma. J. Med. Chem. 61(4), 1552–1575 (2018).
    • 63. Zhao B, Lei F, Wang C et al. Design, Synthesis and biological evaluation of novel phenylsulfonylurea derivatives as PI3K/mTOR dual inhibitors. Molecules 23(7), 1553 (2018).
    • 64. Liu Y, Xia Q, Fang L. Design and synthesis of alkyl-substituted pyridino[2,3-D]pyrimidine compounds as PI3Kα/mTOR dual inhibitors with improved pharmacokinetic properties and potent in vivo antitumor activity. Bioorg. Med. Chem. 26(14), 3992–4000 (2018).
    • 65. Zhang L, Bu T, Bao X et al. Design, synthesis and biological evaluation of novel 3 H-imidazole [4,5- b ] pyridine derivatives as selective mTOR inhibitors. Bioorg. Med. Chem. Lett. 27(15), 3395–3398 (2017).
    • 66. Mao B, Gao S, Weng Y, Zhang L, Zhang L. Design, synthesis, and biological evaluation of imidazo[1,2- b ]pyridazine derivatives as mTOR inhibitors. Eur. J. Med. Chem. 129, 135–150 (2017).
    • 67. Zhang J, Lv X, Ma X, Hu Y. Discovery of a series of N -(5-(quinolin-6-yl)pyridin-3-yl)benzenesulfonamides as PI3K/mTOR dual inhibitors. Eur. J. Med. Chem. 127, 509–520 (2017).
    • 68. Lv X, Ying H, Ma X et al. Design, synthesis and biological evaluation of novel 4-alkynyl-quinoline derivatives as PI3K/mTOR dual inhibitors. Eur. J. Med. Chem. 99, 36–50 (2015).
    • 69. Zhu W, Chen C, Sun C et al. Design, synthesis and docking studies of novel thienopyrimidine derivatives bearing chromone moiety as mTOR/PI3Kα inhibitors. Eur. J. Med. Chem. 93, 64–73 (2015).
    • 70. Le PT, Cheng H, Ninkovic S et al. Design and synthesis of a novel pyrrolidinyl pyrido pyrimidinone derivative as a potent inhibitor of PI3Kα and mTOR. Bioorg. Med. Chem. Lett. 22(15), 5098–5103 (2012).
    • 71. Han F, Lin S, Liu P, Tao J, Yi C, Xu H. Synthesis and structure–activity relationships of PI3K/mTOR dual inhibitors from a series of 2-amino-4-methylpyrido[2,3- d ]pyrimidine derivatives. Bioorg. Med. Chem. Lett. 24(18), 4538–4541 (2014).
    • 72. Ma X, Lv X, Qiu N, Yang B, He Q, Hu Y. Discovery of novel quinoline-based mTOR inhibitors via introducing intra-molecular hydrogen bonding scaffold (iMHBS): the design, synthesis and biological evaluation. Bioorg. Med. Chem. 23(24), 7585–7596 (2015).
    • 73. Lee W, Ortwine DF, Bergeron P et al. A hit to lead discovery of novel N-methylated imidazolo-, pyrrolo-, and pyrazolo-pyrimidines as potent and selective mTOR inhibitors. Bioorg. Med. Chem. Lett. 23(18), 5097–5104 (2013).
    • 74. Liu KK-C, Bagrodia S, Bailey S et al. 4-Methylpteridinones as orally active and selective PI3K/mTOR dual inhibitors. Bioorg. Med. Chem. Lett. 20(20), 6096–6099 (2010).
    • 75. Rageot D, Bohnacker T, Melone A et al. Discovery and preclinical characterization of 5-[4,6-Bis({3-oxa-8-azabicyclo[3.2.1]octan-8-yl})-1,3,5-triazin-2-yl]-4-(difluoromethyl)pyridin-2-amine (PQR620), a highly potent and selective mTORC1/2 inhibitor for cancer and neurological disorders. J. Med. Chem. 61(22), 10084–10105 (2018).
    • 76. Yu T, Li N, Wu C et al. Discovery of pyridopyrimidinones as potent and orally active dual inhibitors of PI3K/mTOR. ACS Med. Chem. Lett. 9(3), 256–261 (2018).
    • 77. Zhang J, Ma X, Lv X et al. Identification of 3-amidoquinoline derivatives as PI3K/mTOR dual inhibitors with potential for cancer therapy. RSC Adv. 7(4), 2342–2350 (2017).
    • 78. Saurat T, Buron F, Rodrigues N et al. Design, synthesis, and biological activity of pyridopyrimidine scaffolds as novel PI3K/mTOR dual inhibitors. J. Med. Chem. 57(3), 613–631 (2014).
    • 79. Takeuchi CS, Kim BG, Blazey CM et al. Discovery of a novel class of highly potent, selective, ATP-competitive, and orally bioavailable inhibitors of the mammalian target of rapamycin (mTOR). J. Med. Chem. 56(6), 2218–2234 (2013).
    • 80. D'Angelo ND, Kim T-S, Andrews K et al. Discovery and optimization of a series of benzothiazole phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) dual inhibitors. J. Med. Chem. 54(6), 1789–1811 (2011).
    • 81. Koehler MFT, Bergeron P, Blackwood E et al. Potent, selective, and orally bioavailable inhibitors of the mammalian target of rapamycin kinase domain exhibiting single agent antiproliferative activity. J. Med. Chem. 55(24), 10958–10971 (2012).
    • 82. Lin Z, Wang Z, Zhou X et al. Discovery of new fluorescent thiazole–pyrazoline derivatives as autophagy inducers by inhibiting mTOR activity in A549 human lung cancer cells. Cell Death Dis. 11(7), 551 (2020).
    • 83. Lawal B, Lo W-C, Mokgautsi N et al. A preclinical report of a cobimetinib-inspired novel anticancer small-molecule scaffold of isoflavones, NSC777213, for targeting PI3K/AKT/mTOR/MEK in multiple cancers. Am. J. Cancer Res. 11(6), 2590–2617 (2021).
    • 84. Sun C-Y, Li Y-Z, Cao D, Zhou Y-F, Zhang M-Y, Wang H-Y. Rapamycin and trametinib: a rational combination for treatment of NSCLC. Int. J. Biol. Sci. 17(12), 3211–3223 (2021).
    • 85. Yu J, Zhang L, Peng J et al. Dictamnine, a novel c-Met inhibitor, suppresses the proliferation of lung cancer cells by downregulating the PI3K/AKT/mTOR and MAPK signaling pathways. Biochem. Pharmacol. 195, 114864 (2022).
    • 86. Ryczkowska M, Maciejewska N, Olszewski M, Witkowska M, Makowiec S. New tetrahydroquinoline derivatives with anticancer activity: design, synthesis, and biological evaluation. www.researchsquare.com/article/rs-1497725/v1
    • 87. Faheem Karan Kumar B, Venkata Gowri Chandra Sekhar K, Chander S, Kunjiappan S, Murugesan S. 1, 2, 3, 4-Tetrahydroisoquinoline (THIQ) as privileged scaffold for anticancer de novo drug design. Expert Opin. Drug Discov. 16(10), 1119–1147 (2021).
    • 88. Fathy U, Azzam MA, Mahdy F, El‐Maghraby S, Allam RM. Synthesis and in vitro anticancer activity of some novel tetrahydroquinoline derivatives bearing pyrazol and hydrazide moiety. J. Heterocycl. Chem. 57(5), 2108–2120 (2020).
    • 89. He G, Chen C, Wang T et al. Combining structure-based pharmacophore modeling, virtual screening, and in silico ADMET analysis to discover novel tetrahydro-quinoline based pyruvate kinase isozyme M2 activators with antitumor activity. Drug Des. Devel. Ther. 8, 1195 (2014).
    • 90. Chaube UJ, Rawal R, Jha AB, Variya B, Bhatt HG. Design and development of tetrahydro-quinoline derivatives as dual mTOR-C1/C2 inhibitors for the treatment of lung cancer. Bioorganic Chem. 106, 104501 (2021). •• Design and development of THQ as mTOR inhibitors for the treatment of lung cancer.
    • 91. Kouznetsov VV, Merchan-Arenas DR, Tangarife-Castaño V, Correa-Royero J, Betancur-Galvis L. Synthesis and cytotoxic evaluation of novel 2-aryl-4-(4-hydroxy-3-methoxyphenyl)-3-methyl-6,7-methylendioxy-1,2,3,4-tetrahydroquinolines, podophyllotoxin-like molecules. Med. Chem. Res. 25(3), 429–437 (2016).
    • 92. Wang S-B, Wang X-F, Qin B et al. Optimization of n-aryl-6-methoxy-1,2,3,4-tetrahydroquinolines as tubulin polymerization inhibitors. Bioorg. Med. Chem. 23(17), 5740–5747 (2015).
    • 93. Lam K-H, Lee KK-H, Gambari R et al. Preparation of Galipea officinalis Hancock type tetrahydroquinoline alkaloid analogues as anti-tumour agents. Phytomedicine 20(2), 166–171 (2013).
    • 94. Wang X-F, Wang S-B, Ohkoshi E et al. N-aryl-6-methoxy-1,2,3,4-tetrahydroquinolines: a novel class of antitumor agents targeting the colchicine site on tubulin. Eur. J. Med. Chem. 67, 196–207 (2013).
    • 95. Sonawane YA, Zhu Y, Garrison JC et al. Structure–activity relationship studies with tetrahydroquinoline analogs as EPAC inhibitors. ACS Med. Chem. Lett. 8(11), 1183–1187 (2017).
    • 96. Jo H, Choi M, Kumar AS et al. Development of novel 1,2,3,4-tetrahydroquinoline scaffolds as potent NF-κB inhibitors and cytotoxic agents. ACS Med. Chem. Lett. 7(4), 385–390 (2016).
    • 97. Muthukrishnan I, Sridharan V, Menéndez JC. Progress in the chemistry of tetrahydroquinolines. Chem. Rev. 119(8), 5057–5191 (2019). •• Importance of THQ in medicinal chemistry and use of the scaffold in multidisciplinary areas.