1,053
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      Acta Materia Medica now indexed by SCOPUS from May 2024. Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
      0
      0
      0
      0
      Smart Citations
      0
      0
      0
      0
      Citing PublicationsSupportingMentioningContrasting
      View Citations

      See how this article has been cited at scite.ai

      scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.

       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Research advances in anti-cancer activities and mechanism of action of neocryptolepine and its derivatives

      Published
      review-article
      Bookmark

            Abstract

            Cancer has been a severe public health and social problem, a leading disease that has diminished the quality of life, and a barrier to improving life expectancy. Neocryptolepine is an indole-quinoline alkaloid isolated from Cryptolepis sanguinolenta which grows in some African countries. This review summarizes the structures of 228 neocryptolepine derivatives, including 84 neocryptolepine derivatives synthesized by our laboratory, and analyzed the cytotoxic effects and mechanism of action at the cellular level. Neocryptolepine derivatives 43, 65, 93, and 96 have good cytotoxicity against gastric cancer AGS cells and the IC50 value reached 43 nM, 148 nM, 2.9 μM, and 4.5 μM, respectively. The IC50 values of compounds 64 and 69 on colorectal cancer HCT116 cells reached 0.33 and 0.35 μM, respectively. In addition, the structure-activity relationship of these compounds is discussed in this review. Topoisomerase II is discussed as a possible inhibition target of neocryptolepine derivatives in several cancer cell lines by binding DNA. The structures of the reported neocryptolepine derivatives and the possible cytotoxic mechanisms are analyzed. This review provides a fundamental reference for anticancer drug development of neocryptolepine and its derivatives as anti-tumor agents.

            Main article text

            1. INTRODUCTION

            The emergence and spread of cancer are considered major threats to global public health by the World Health Organization. Cancer is a non-communicable disease and one of the leading causes of human mortality [1, 2]. The International Agency for Research on Cancer (IARC) reported that the estimated number of cancer cases worldwide in 2018 has risen to 18.1 million new cases, resulting in 9.6 million deaths [3, 4]. In addition, due to the high contribution of Asia to the global population, nearly one-half of the estimated cases and greater than one-half of cancer deaths in men and women occur in Asia. Greater than 60% of cancers occur in low- and middle-income countries and approximately 70% of cancer deaths occur in these countries [5]. The traditional cancer treatment modalitiess include surgery, radiation, and chemotherapy. The negative side effects associated with these treatment options, such as high cost and toxicity, increase the demand for the development of less toxic and less expensive anticancer drugs from natural sources [6]. Because the cancer death rates are rising in developing and industrialized countries, there is an urgent need to explore novel anticancer drugs. Moreover, there is a need to develop new anticancer drugs to treat cancer and generate interest in the pharmaceutical industry [7].

            Cancer cells have the ability of uncontrolled proliferation and can migrate to other tissues and organs [8]. Although research and development of anticancer drugs have made significant advances in recent decades, anti-cancer therapy still has challenges. Chemotherapy, radiotherapy, and surgery are important therapeutic methods for cancer patients [9, 10]. Small molecular chemical drugs have many advantages compared to biological drugs and antibodies, including low cost, more forms of administration, and good patient compliance [11]. However, most clinical anticancer drugs are expensive and toxic, and the side effects of chemical drugs cannot be ignored. Although chemotherapy drugs clearly improve overall survival among cancer patients, treatment can be painful. Therefore, it is essential to find drugs with highly effective and low toxicity for cancer treatment.

            Natural products have a role in the development of anticancer drugs, which are a significant source of bioactive molecules in anti-cancer drugs [12]. There were 321 anti-cancer drugs approved between 1946 and 2019, among which greater than 30% belong to natural products or their derivatives [13]. There are many anti-cancer drugs that originated from natural products, such as taxol [14], camptothecin [15], vincristine [16], topotecan [17], and etoposide [18] ( Figure 1 ), that have been shown to have good inhibitory effects on cancer. Neocryptolepine, an indoquinoline alkaloid, is derived from Cryptolepis sanguinolenta, which grows in some African countries [19]. Neocryptolepine is widely used in traditional medicine in many Central and Western countries [2022]. In fact, cryptolepine, isocryptolepine, and neocryptolepine ( Figure 1 ) are the main alkaloids isolated from the roots of C. sanguinolenta. They are mainly used for the treatment of hypertension and fever [23] and have anti-muscarinic [24], anti-bacterial [25], anti-fungal [26], and anti-inflammatory effects [24]. Therefore, neocryptolepine and its derivatives are promising resources for the development of anti-cancer drugs.

            Next follows the figure caption
            Figure 1 |

            Chemical structures of FDA-approved anti-cancer drugs (taxol, camptothecin, vincristine, topotecan, and etoposide), cryptolepine, isocryptolepine, and neocryptolepine.

            To develop anti-cancer drugs with high efficacy and low toxicity, the cytotoxicity and mechanism of action underlying a series of neocryptolepine derivatives were reviewed in different cancer cells, such as colorectal, gastric, liver, lung, ovarian, breast, and cervical cancer, Ehrlich ascites carcinoma (EAC), and melanoma. Although many studies have reported that neocryptolepine derivatives exert good cytotoxicity, a systematic summary of structure-activity relationships and the mechanism of action for neocryptolepine and its derivatives are lacking. The cytotoxic mechanism underlying neocryptolepine and its derivatives are analyzed in this review ( Figure 2A and 2B ) and the neocryptolepine derivatives structures modified with the neocryptolepine parent nucleus at A, B, C, and D are discussed ( Figure 2B ). This review lays the foundation to develop anticancer drugs with high efficacy and low toxicity from neocryptolepine and its derivatives and provides ideas for developing novel anti-cancer drugs by structure modification based on the structure of neocryptolepine.

            Next follows the figure caption
            Figure 2 |

            The cytotoxic effects and potential mechanism of action of neocryptolepine and its derivatives (Figure A was drawn by Figdraw, ID: WSOSO688f1).

            2. NATURAL PRODUCTS AS ANTICANCER AGENTS

            Cancer is one of the major diseases that threatens human life and health, so the isolation and synthesis of compounds with antitumor activity are areas of intense research. Derivatization of the skeleton structure of some natural products is a common method by which to obtain active compounds. Studies have shown that plants are natural sources of novel drugs with unique medicinal activities [2731]. Natural products based on plants and their derivatives have been widely used as sources for the development of anticancer agents and have been extensively documented for the treatment of various diseases. Alkaloids, which are nitrogen-containing compounds in plants, fungi, and bacteria, are among the most important natural products [3235]. Most alkaloids are found in vascular plants and a few alkaloids occur in lower plants [35]. The chemical structures of alkaloids in the same plant are similar. These alkaloids are mainly categorized as follows: pyridines; terpenoids; indoles; steroids; and isoquinolines. Many kinds of alkaloids have been shown to have anti-cancer activity [36]. With further research involving alkaloids, more novel alkaloids with anti-cancer activity have been identified.

            2.1 Indole-quinoline alkaloids

            Indole-quinoline alkaloids are tetracyclic heterocycles formed by the fusion of indoles and quinoline. Cryptolepine is the natural product of indole-quinoline alkaloids, which were isolated in 1929. The exact chemical structure of cryptolepine was assigned in 1951 as a linearly-fused 5-methyl-10H-indolo [3,2-b]quinoline [37]. Cryptolepine has been investigated as a promising anti-malarial and anti-cancer agent [38]. In fact, indole-quinoline alkaloids have potential as anti-cancer drugs and it has been reported that quinoline and indole-quinoline alkaloids generally have good anti-bacterial and cytotoxic activities [39, 40]. However, different studies have shown that the toxicity of cryptolepine cannot be ignored, which hinders further progress.

            2.2 Neocryptolepine and its derivatives

            Neocryptolepine is a polycyclic quinoline compound and has cytotoxic effects in different cancer cells, including liver cancer, cholangiocarcinoma, lung cancer, EAC, and leukemia. Neocryptolepine causes cell cycle arrest, induces apoptosis, and has a good anti-oxidant effect [7, 41, 42]. Some neocryptolepine derivatives (9 and 10; Figure 3 ) have strong cytotoxicity against the A549 cell line with IC50 of 0.197 and 0.1988 μM, respectively, and IC50 of 0.138 and 0.117 μM against BALB/3T3 cells, respectively [43].

            Next follows the figure caption
            Figure 3 |

            The chemical structures of neocryptolepine derivatives, indoquinazoline alkaloids, and indole alkaloids [4345].

            2.3 Other alkaloids

            Nine novel indoquinazoline alkaloids (11-19; Figure 3 ) were isolated and characterized from Euodiae fructus and shown to have significant cytotoxicity against HL-60 human promyelocyte leukemia and N-87 human gastric cancer cells [44]. Some synthetic indole derivatives have been confirmed to have promising anti-cancer activities against various human cancer cell lines, such as colorectal cancer HCT116 cells, breast cancer MCF-7 cells, and cervical cancer Hela cells [44]. An indole alkaloid (20; Figure 3 ) isolated from Tabernaemontana catharinensis showed significant cytotoxic activity against laryngeal carcinoma Hep-2 cells (IC50 = 54.47 μg/mL) [45]. Indole-quinoline alkaloids are considered promising frameworks for the development of anti-cancer drugs, which can be further developed into effective anti-cancer drugs [46, 47]. Neocryptolepine and its derivatives also have great potential as cytotoxic agents. This review was mainly focused on the advances in anti-cancer effects of neocryptolepine and its derivatives in different cancer lines by inhibiting proliferation, migration, inducing apoptosis, and arresting cell cycle. A summary of anti-cancer effects and the mechanism of action of neocryptolepine derivatives in various cancer lines is presented.

            3. NEOCRYTOLEPINE AND NEOCRYTOLEPINE DERIVATIVES AS ANTI-CANCER AGENTS

            3.1 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against gastric cancer

            Alkaloids as the main anti-tumor active substances undergoing broad development and research. The application of alkaloid anti-cancer drugs will reduce the toxicity and side effects of chemotherapeutic drugs and improve the efficacy of anti-tumor activity. To help the preliminary screening of alkaloids of compounds with anti-tumor activity for future studies, we developed a database of alkaloids with anti-tumor activity [DAAA] (http://www.gsbios.com). This database analyzed and extracted the relevant literature and integrated research data and results. This database contains the name, structure, molecular weight, molecular formula, activity, toxicity, patents, and corresponding references of alkaloids. Furthermore, this database contains many neocryptoleepine derivatives and will be used to develop promising compounds with anti-tumor activity.

            In our previous work we designed 84 neocryptolepine derivatives ( Figure 4A, B ) and evaluated the cytotoxic activity against gastric cancer AGS cells [19, 48, 49]. Some neocryptolepine derivarives showed good cytoxixc effects against AGS cells. Neocryptolepine derivative 43 had excellent cytoxicity. Specifically, the IC50 of compound 43 was 43 nM for 48 h in AGS cells. In addition, the IC50 of compounds 65, 93, and 96 reached 148 nM, 2.9 μM, and 4.5 μM, respectively. We also assessed the cytotoxicity of neocryptolepine derivatives in liver cancer SMMC7721 cells, colorectal cancer HCT116 cells, and pancreatic cancer PANC-1 cells. The IC50 of neocryptolepine derivatives in these cancer cells are listed in Table 1 . As seen in Table 1 and Figure 5 , in the A-ring substitutions of neocryptolepine, 2-halogenation (compounds 24, 25, and 26) showed better cytotoxicity than other positions (compounds 29, 30, 31, 33, and 35). 4-methyl substitution (compound 32) had better cytotoxic activity than 3- or 2-methyl substitution (compounds 22 and 27). Moreover, 2-methoxy (compound 23) substitution had better cytoxic effects than the 3-methoxy (compound 28). The 9-Cl substitution (compound 39) is better than the 7-, 8-, and 10-Cl substitution (compounds 44, 49 and 52) when the C1 atom in neocryptolepine is substituted. The 10-methyl substitution (compound 46) has better cytotoxicity than the 7-methyl substitution (compound 51) when the substituent group is methyl. When the 2-position substituent group is methoxy (compounds 54-59), 8-F, 8-methyl, and 8-methoxy (compounds 55, 58, 59) have better cytotoxicity than 8-Cl (compound 56) and 8-Br (compound 57) in the A- and D-ring substitutions. When the substituent group is methyl (compounds 60-64), 8-methyl (compound 63) and 8-methoxy (compound 64) have the best cytotoxic effects, and 8-F (compound 60) and 8-Cl (compound 61) are better than 8-Br (compound 62). When the substituent group is the Cl atom (compound 65-69), 8-Br (compound 67) has better cytotoxicity than 8-Cl (compound 66) and 8-methyl (compound 68). These results suggest that neocryptolepine derivatives have good cytotoxicity in AGS cells by regulating the PI3K/AKT/mTOR signaling pathway. Moreover, neocryptolepine derivatives induce cell apoptosis, arrest the cell cycle at G2/M, and inhibit the migration of cancer cells. The identify and verification of the neocryptolepine derivative targets are important and warrant further study.

            Next follows the figure caption
            Figure 4 |

            The structure of neocryptolepine derivatives and analogues [26, 48].

            Table 1 |

            Anti-proliferative effects (IC50, μM) of neocryptolepine and its derivatives (21-105) and CIS on AGS, SMMC7721, HCT116, and PANC-1 cells for 48 h.

            CompoundsSMMC7721AGSPANC-1HCT116CompoundsSMMC7721AGSPANC-1HCT116
            21 27.00 ± 7.1020.00 ± 0.77>506.30 ± 1.20 64 9.70 ± 2.003.60 ± 2.5018.40 ± 3.000.30 ± 0.00
            22 24.00 ± 0.0027.00 ± 7.40>5010.10 ± 0.60 65 24.00 ± 0.000.15 ± 0.006>5027.60 ± 16.30
            23 19.00 ± 5.6014.00 ± 8.3037.60 ± 0.0012.50 ± 5.60 66 23.00 ± 0.007.10 ± 3.3037.50 ± 11.8022.70 ± 0.00
            24 10.00 ± 5.504.00 ± 0.6014.00 ± 0.103.70 ± 2.40 67 19.00 ± 0.002.80 ± 3.6027.10 ± 0.007.10 ± 0.00
            25 12.00 ± 6.103.30 ± 0.508.90 ± 1.601.80 ± 0.90 68 26.00 ± 24.007.70 ± 5.7045.80 ± 0.007.500 ± 5.30
            26 19.00 ± 16.005.00 ± 2.5020.10 ± 5.807.60 ± 4.50 69 13.00 ± 0.0027.00 ± 38.00>500.40 ± 0.00
            27 25.00 ± 0.8014.00 ± 1.6035.40 ± 13.1011.40 ± 2.60 70 >50>50>50>50
            28 >5041.00 ± 0.00>5025.70 ± 11.20 71 >5015.00 ± 6.7024.00 ± 0.0012.50 ± 3.10
            29 22.00 ± 6.807.10 ± 0.20>5017.70 ± 13.50 72 >5026.00 ± 0.7057.70 ± 0.0049.60 ± 20.60
            30 >50>50>50>50 73 >5013.00 ± 7.026.80 ± 4.30ND
            31 37.00 ± 0.0036.00 ± 0.00>5020.50 ± 9.80 74 >508.90 ± 0.6022.60 ± 8.4020.30 ± 4.40
            32 13.00 ± 8.603.10 ± 0.3010.50 ± 5.407.20 ± 7.30 75 >5030.00 ± 5.60>5030.70 ± 5.10
            33 15.00 ± 8.706.00 ± 0.8025.00 ± 0.106.30 ± 3.70 76 >5028.00 ± 0.0042.30 ± 0.0026.00 ± 11.20
            34 17.00 ± 0.008.60 ± 1.00>5022.30 ± 16.80 77 >50>50>5035.50 ± 8.60
            35 17.00 ± 0.0014.00 ± 1.4035.40 ± 4.2011.70 ± 0.00 78 >509.30 ± 0.6025.60 ± 0.0019.80 ± 2.40
            36 18.00 ± 11.008.10 ± 3.0018.10 ± 0.006.30 ± 4.00 79 >5014.00 ± 4.7022.90 ± 0.00>50
            37 28.00 ± 19.007.90 ± 0.606.90 ± 0.0010.50 ± 10.50 80 39.00 ± 1304.50 ± 0.6038.50 ± 0.00160.20 ± 108.50
            38 28.00 ± 0.0038.00 ± 14.00>5016.50 ± 9.00 81 23.00 ± 1.901.50 ± 1.40>5020.70 ± 7.30
            39 22.00 ± 0.0018.00 ± 0.00>5011.50 ± 0.00 82 15.00 ± 6.202.20 ± 0.303.30 ± 0.007.60 ± 0.30
            40 22.00 ± 2.8017.50 ± 0.00>5011.50 ± 0.00 83 27.00 ± 5.803.50 ± 0.704.90 ± 0.0013.00 ± 3.10
            41 16.00 ± 10.003.10 ± 0.80>506.10 ± 3.00 84 29.00 ± 7.6026.00 ± 18.0019.70 ± 0.008.00 ± 4.10
            42 13.00 ± 1.3011.00 ± 9.4031.10 ± 21.507.60 ± 4.80 85 15.00 ± 1.203.30 ± 1.6013.30 ± 3.407.20 ± 0.40
            43 52.00 ± 45.000.043 ± 0.0039>5017.70 ± 0.00 86 38.00 ± 12.007.00 ± 0.6013.80 ± 0.0015.00 ± 3.40
            44 27.00 ± 0.00>50>5037.60 ± 0.00 87 >509.90 ± 0.00>5035.60 ± 14.30
            45 40.00 ± 0.00>50>5045.90 ± 0.00 88 41.00 ± 9.2016.00 ± 0.00>5012.70 ± 0.00
            46 12.00 ± 1.204.60 ± 0.3016.60 ± 5.007.20 ± 5.60 89 >505.10 ± 0.00>50ND
            47 16.00 ± 2.408.40 ± 2.7035.30 ± 3.009.40 ± 4.70 90 >507.30 ± 4.4023.00 ± 16.302.90 ± 0.80
            48 36.00 ± 0.0012.00 ± 7.70>5042.40 ± 0.00 91 >50>50>50>50
            49 38.00 ± 0.00>50>50>50 92 >504.20 ± 3.104.30 ± 0.001.00 ± 0.60
            50 30.00 ± 4.8015.00 ± 0.0019.90 ± 0.0040.80 ± 0.00 93 40.00 ± 18.002.90 ± 0.103.90 ± 0.101.90 ± 2.50
            51 22.00 ± 12.007.80 ± 1.4042.00 ± 0.009.80 ± 2.50 94 >50>50>50>50
            52 43.00 ± 10.0022.00 ± 0.00>50>50 95 >504.70 ± 0.504.40 ± 0.802.20 ± 2.40
            53 >5041.00 ± 0.00>50>50 96 12.00 ± 0.514.50 ± 2.503.60 ± 0.902.80 ± 0.00
            54 2.70 ± 0.003.00 ± 0.103.80 ± 0.001.60 ± 0.20 97 >505.70 ± 1.208.50 ± 0.008.30 ± 0.60
            55 13.00 ± 1.106.50 ± 1.4035.60 ± 0.006.70 ± 0.30 98 >506.40 ± 0.0015.80 ± 1.505.80 ± 2.80
            56 15.00 ± 2.4013.00 ± 3.00>506.60 ± 0.90 99 >5015.00 ± 0.00>50ND
            57 19.00 ± 2.7021.00 ± 12.00>504.10 ± 2.70 100 >503.90 ± 0.0020.10 ± 0.0027.30 ± 10.70
            58 4.60 ± 2.402.30 ± 0.805.60 ± 0.002.10 ± 2.20 101 >504.70 ± 0.0029.90 ± 5.7016.80 ± 3.80
            59 6.50 ± 5.502.70 ± 1.509.10 ± 1.902.90 ± 3.40 102 >5010.40 ± 2.0022.00 ± 0.003.70 ± 2.00
            60 11.00 ± 2.804.80 ± 2.20>508.70 ± 0.00 103 >50>50>50>50
            61 15.00 ± 7.205.00 ± 2.7026.30 ± 3.304.90 ± 3.50 104 >50>50>50ND
            62 21.00 ± 13.0016.00 ± 2.30>509.20 ± 7.40 105 >50>50>50ND
            63 8.00 ± 0.503.00 ± 1.4012.90 ± 2.002.90 ± 0.90CIS>5015.00 ± 1.40>507.80 ± 1.90

            ND, not detected.

            Next follows the figure caption
            Figure 5 |

            The structure-activity relationship analysis of neocryptolepine derivatives.

            3.2 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against colorectal cancer

            Several studies have attempted to synthesize a variety of neocryptolepine derivatives to find leading compounds with superior anti-colorectal cancer efficacy. Boddupally et al. [50] synthesized a series of 11-substituted derivatives of cryptolepine in 2012. Compound 107 ( Figure 6A ) had the strongest anti-cancer activity with IC50 of 0.97 μM against colorectal cancer HCT116 cells and 2.33 μM against Raji lymphoma cells in in vitro cytotoxicity tests. Compound 107 concurrently demonstrated a significant suppression of c-MYC expression [50]. Wang et al. [51] synthesized a series of 11-amino-substituted 5H- and 6H-indolo[2,3-b]quinolines, the anti-proliferative effects of which were assessed in human lung cancer A549 cells and colorectal cancer HCT116 cells. Compound 109 ( Figure 6B ) has good anti-proliferative activities against HCT116 cells (IC50 = 0.195 ± 0.044 μM) and lower toxicity [51]. Wang et al. [51] reported that the 11-chloroneocryptolepines are prone to undergo addition-elimination reactions of the amine nucleophiles of the linear primary amines at 70–130 °C, giving compound 109 good-to-excellent yields. Sidoryk et al. [56] synthesized a series of neocryptolepine derivatives that have an amino acid or a dipeptide at position C-9 and assessed anti-cancer activity in vitro and in vivo. The anti-proliferative effects of 5H-indolo[2,3b]quinoline against Lovo cells was demonstrated by its amino acid and peptide derivatives and cytotoxicity was as high as 0.20 ± 0.40 μM.

            Next follows the figure caption
            Figure 6 |

            The synthetic route and structures of compounds 106-123 [5055].

            Wang et al. [52] tested the 84-neocryptolepine and 44-isocryptolepine derivatives and found that the −NH(CH2)3NH2 side chain substituents were the most powerful and potent compounds. Compound 108 ( Figure 6A ) exerts good cytotoxicity with an IC50 of 0.117 μM against HCT116 cells. New anti-proliferative neocryptolepine derivatives 113-118 were effectively synthesized based on neocryptolepine and were explained using spectroscopic techniques in the presence of lithium perchlorate as a Lewis acid catalyst [53]. Compound 118 had the best cytotoxicity against the HCT116 cells and the IC50 was 2.4 μM compared to the reference drug, doxorubicin (IC50 = 10.90 μM). Håheim [54] presented modifications to previous synthetic strategies, which allowed for the realization of novel tetracyclic ring-systems (compounds 119-123; Figure 6F ) along with the N-alkylation of several compounds to furnish novel analogues. Both parent alkaloids of neocryptolepine (compound 8) and isocryptolepine (compound 7) performed good cytotoxicity against HCT116 cells (compound 8: IC50 = 6.22 μM; compound 7: IC50 = 0.67 μM).

            A number of studies have been conducted to investigate the anti-colorectal cancer bioactivity of the coumarin compound, 3,3′-(3,4-dichlorobenzylidene)-bis-(4-hydroxycoumarin), which is termed DCH and its derivatives. Matsui et al. [57] showed that cryptolepine induced cell cycle arrest in MG63 cells through the p53-independent activation of p21WAF1/CIP1 and the activator is mediated through the specific Sp1 site in the promoter region. These findings suggested that cryptolepine arrested the growth of MG63 cells by activating the p21WAF1/CIP1 promoter through the specific Sp1 site in a p53-independent manner. The unregulated wingless integrated type-1 (WNT)/β-catenin signaling pathway is the cause of >90% of colorectal cancers. Quarshie et al. [58] showed that cryptolepine inhibits WNT3a, a WNT activator-mediated activation of the WNT/β-catenin signaling pathway, which inhibits the proliferation, stemness, and metastasis of colorectal cancer cells. Cryptolepine (compound 6) had excellent anti-cancer effects in colorectal adenocarcinoma epithelial DLD1 cells and colorectal cancer COLO205 cells with IC50 of 2.45 and 1.16 μM after 48 h, respectively. Cryptolepine also reduces WNT3a-induced OCT4 and CD133 expression and suppresses colony formation of the cells. Cryptolepine suppresses cell stemness. Nagy et al. [55] evaluated the cytotoxic activity of 11(4-aminophenylamino) neocryptolepine [APAN] (compound 110; Figure 6C ) in HCT116 cells. Treatment of compound 110 caused cytotoxicity in HepG2 and HCT116 cells with IC50 values of 2.60 and 1.82 μg/mL, respectively. Compound 110 caused damage and severe morphologic alterations in the cells, such as membrane blebbing, cytoplasmic condensation, shrinking of the nucleus with increased condensed chromatin, and removal of microvilli [55].

            Abd Elrahman et al. [59] evaluated the cytotoxic activity of 11-(1,4-bisaminopropylpiperazinyl)5-methyl-5H-indolo[2,3-b] quinoline [BAPPN] (compound 112; Figure 6D ). Compound 112 had cytotoxic activity against colorectal cancer cells (IC50 = 23.00 μg/mL) by upregulation of apoptotic proteins (caspase-3 and p53) and downregulation of proliferative proteins (VEGF, PCNA, and Ki67) [59]. In addition, compound 112 induced cell injury and morphologic changes in ultracellular structure, including cellular delayed activity, vanishing of membrane blebbing, microvilli, cytoplasmic condensation, and shrunken nuclei with more condensed chromatin autophagosomes.

            In our previous studies, 8-methoxy-2,5-dimethyl-5H-indolo[2,3-b] quinoline (compound 64) was tested for cytotoxicity in colorectal cells. The results showed that compound 64 inhibited the growth of HCT116 and Caco-2 cells, arrested the cell cycle in the G2/M phase, reduced the mitochondrial membrane potential, and induced apoptosis. In addition, western blot analysis suggested that compound 64 may inhibit the proliferatiion of colorectal cancer cells by reglulating the PI3K/AKT/mTOR signaling pathway [60].

            3.3 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against ovarian cancer

            Ovarian cancer (OC) is the eighth most common cancer in women worldwide, accounting for 3.7% of all cancers and 4.7% of all cancer deaths [61]. In the early 2000s, the incidence of OC was highest in North America and northern Europe. OC can be subdivided into at least five different histologic subtypes with different identifiable risk factors, origin cells, molecular composition [62], clinical features, and treatment methods. OC is a global problem that is typically diagnosed in the late stages and lacks effective screening strategies [63].

            Neocryptolepine and its derivatives have been shown to have biological activity and promising application in the clinical setting [26]. The antitumor and anti-fungal activities suggest that neocryptolepine and its derivatives could be potential anti-cancer drug candidates [26, 49]. It has been shown that neocryptolepine derivatives exhibit significantly anti-proliferative and pro-apoptotic effects in OC cells and high selectivity towards normal cells [22].

            One study found that a neocryptolepine derivative (compound 134; Figure 7C ) had good cytotoxic activity against OC cells and the IC50 value of compound 134 against SKOV3 cells was 4.60 ± 0.10 μM [64]. Overall, neocryptolepine derivatives have shown potential as promising leading compounds for the treatment of OC [7]. Neocryptolepine derivatives have a significant inhibitory effect on OC cells by cytotoxicity or induction of cell apoptosis. Further research and evaluation are needed for the clinical application of neocryptolepine derivatives, including studies on pharmacokinetics, toxicology, and clinical efficacy. In summary, studies involving the treatment of OC have made progress but there are still many challenges. An anti-cancer study of neocryptolepine and its derivatives may provide novel ideas for the development of anti-OC drugs in the future.

            Next follows the figure caption
            Figure 7 |

            The synthesis and structure of neocryptolepine derivatives 124-144 [42, 46, 56, 64].

            3.4 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against liver cancer

            Liver cancer is the fourth most common cancer globally. The occurrence of liver cancer is mainly concentrated in cases of cirrhosis, hepatitis B or C virus infection, or non-alcoholic steatohepatitis. The potential liver disease also limits the therapeutic effect [65]. Hepatocellular carcinoma (HCC) is the most common primary liver cancer, accounting for 85–90% of all cases. Since 1980 the global incidence of liver cancer has tripled and the growth rate of cancer for men and women is faster than that of any other cancer [66]. Since 2000 the HCC mortality rate has increased year-after-year, increasing by 3% every year. Until 2019 liver cancer, including HCC, was the fifth and seventh leading cause of cancer deaths in men and women, respectively. In the past decade significant progress has been made in the systemic treatment of advanced HCC. However, newly developed treatment strategies have not achieved widespread success and patients with advanced liver cancer often exhibit resistance to these therapies.

            The indole-quinoline alkaloids are representative examples of natural products and the indole-quinoline alkaloids exhibit various biological activities by DNA binding and inhibition of topoisomerase II [22]. The indole-quinoline alkaloids also have cytotoxic, anti-bacterial, antitumor, and molluscicidal effects, as well as anti-protozoan activity, especially against Trypanosoma, Leishmania, and Plasmodium [6769]. Some studies have suggested that neocryptoolepine derivatives exhibit significant anti-proliferative and pro-apoptotic effects in liver cancer cells. In one study five of seven neocryptoolepine derivatives (compounds 135-139; Figure 7E and 7F ) showed significant efficacy against human liver cancer HepG-2 cells with an IC50 of 27.70 ± 3.80 μM for compound 135. The IC50 of compound 136 is 25.40 ± 3.30 mM [46].

            3.5 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against ascites carcinoma

            El-Aarag et al. [70] investigated the toxicity of cryptolepine in EAC cells at the cellular level. The analog (compound 140; Figure 7G ) exerted cytotoxic effects in EAC cells and reduced the ascites volume. Moreover, compound 140 induced oxidative stress in EAC by increasing the level of malonaldehyde and decreasing the level of total antioxidant capacity and catalase activity. Compound 140 also induced apoptosis by elevating the level of caspase-8 expression in EAC. Furthermore, compound 140 decreased the level of AKT and mTOR protein expression and upregulated PTEN expression in EAC [70].

            Altwaijry et al. [42] synthesized a series of derivatives and the results of an acute toxicity study in mice showed that the LD50 of compound 144 ( Figure 7G ) was 1000 mg/kg, which indicated that compound 144 could be considered as a low toxicity compound with higher safety margins. The maximum reduction in tumor volume was seen in treatment groups with compound 144 compared to a positive control. In addition, compound 144 might exert anti-tumor activity by reducing the peroxidation of lipids, scavenging the free radicals, and increasing the level of anti-oxidant enzymes.

            Nofal et al. [71] studied the anti-tumor effects of APAN in mice (compound 144). Nofal et al. [71] found that compound 144 had ameliorative activity against Ehrlich solid tumors and hepatic toxicity in mice and the greatest improvement occurred with the combined treatment of compound 144 and chemotherapeutic drug, etoposide. TNF-α was highly expressed in the solid tumor and liver tissues of animals with Ehrlich solid tumors (ESTs) and was markedly downregulated in mono- and/or dual-EST-treated groups compared to untreated mice with EST. Therefore, it is of great significance to develop promising leading compounds against anti-ascites cancer from neocryptolepine and its derivatives.

            3.6 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against osteosarcoma

            Osteosarcoma is the most common form of primary bone cancer in children and young people [72]. Bone malignancy mainly occurs in adolescent patients. Although current effective treatment options greatly improve patient outcomes, >20% of patients die due to tumor metastasis [73]. One of the most important reasons for osteosarcoma treatment failure is resistance of the tumor to chemotherapy drugs [74]. Neoryptolepine and cryptolepine derived from C. sanguinolenta [42] have shown significant anti-plasmodium and cytotoxic activity and have become an important scaffold for drug discovery. Cryptolepine (compound 5) has been reported to inhibit cancer cell growth in vitro by inhibiting cell cycle progression in human osteosarcoma cells [75]. Compound 5 has been shown to have anti-proliferative activity in vitro against several types of cancer cells (KB, MCF-7, A549, and Lovo) [76]. Further research has identified compound 5 as a potential anti-microbial agent against the most common pathogenic bacteria and fungi that cause cancer and transplant-associated infections in humans [77].

            3.7 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against lung cancer

            Lung cancer is one of the cancers with the highest incidence and highest mortality rates [78]. Chemotherapy is still the main treatment modality for lung cancer and compounds extracted and isolated from natural products have an important role in the treatment of lung cancer [79].

            Neocryptolepine and its derivatives have been reported to exert good cytotoxic effects in lung cancer in recent decades. Akkachairin and his co-workers found novel compounds containing five-membered ring fused quinoline core structures as anti-cancer and anti-malarial agents. Akkachairin et al. innovated synthetic methods for the synthesis of neocryptolepine derivatives and carbocycle-fused quinolines and the cytotoxicity of all neocryptolepine derivatives (145-167; Figure 8A ) against human lung cancer A549 cells were evaluated using the MTT assay. Among these compounds, compounds 149 (IC50 = 15.58 ± 0.78 μM), 150 (IC50 = 13.63 ± 1.57 μM), 151 (IC50 = 14.89 ± 0.46 μM), 152 (IC50 = 15.31 ± 0.57 μM), 153 (IC50 = 10.80 ± 1.57 μM), and 164 (IC50 = 14.00 ± 1.37 μM) displayed good cytotoxic activity. Compound 153 exhibited the best cytotoxic activity and compound 152 had the greatest selectivity. Topoisomerase II has been shown to be be a target of inhibition by neocryptolepine derivatives in several cancer cells via DNA intercalation [8183]. Therefore, the mode of inhibiting the proliferation of compound 152 involved inhibition of topoisomerase II in A549 cells as well [7]. Ahmed et al. [53] synthesized a series of neocryptolepine compounds 113-118 ( Figure 6E ) and the cytotoxicity of these compounds in A549 cells was examined using the MTT assay. Compound 113 displayed the best cytotoxic activity against A549 cells with an IC50 of 23.10 μM compared to doxorubicin [53]. Wang et al. [84] designed and synthesized a series of neocryptolepine derivatives and the results of the MTT assay revealed that the compounds had good anti-proliferative activity (IC50 = 0.20–4.54 μM) against A549 cells and lower cytotoxicity against normal fibroblasts (BALB/3T3 cells). Inokuchi and his co-workers obtained a series of neocryptolepine derivatives 168-193 ( Figures 8B and 9A ) by the structural modification of neocryptolepine. The anti-proliferative activities of these compounds were tested in vitro against A549 cells. Most compounds showed good cytotoxicity and 11-(3-amino-2-hydroxy) propylamino derivatives 172 and 173 had the best cytotoxicity with IC50 values of 0.20 and 0.19 μM against the A549 cells, respectively. The computer-assisted database analysis, COMPARE, suggested that compounds 171 and 176 have a mode of action similar to actinomycin D, while compound 175 has a mode of action similar to vincristine sulfate or aclarubicin hydrochloride [52].

            Next follows the figure caption
            Figure 8 |

            The synthetic route of compounds 145-167 and compounds 168-185 [7, 80].

            Next follows the figure caption
            Figure 9 |

            The chemical structures and synthetic route of neocryptolepine derivatives 186-212 [43, 52, 80, 86].

            3.8 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against breast cancer

            Neocryptolepine has become a focus of research because of its good cytotoxicity and is considered a promising backbone for drug development. Some studies have reported that neocryptolepine has good cytotoxic effects on a variety of cancer cells, including lung cancer, cholangiocarcinoma, liver cancer, and leukemia [56]. Neocryptolepine arrests the cell cycle at the G2/M phase, induces apoptosis, and affects mitochondrial function [48]. In recent years it has been shown that some neocryptolepine derivatives have better anti-proliferative activity against cancer cells, such as lung, breast, colorectal, and oral epidermoid cancer, after the structural modification of neocryptolepine [46, 53, 55, 56].

            Cryptolepine has been demonstrated to have good cytotoxic activity in breast cancer by affecting cyclins D1, D2, and D3 and cyclin E, which regulate the cell cycle [22, 49, 85]. Plant-derived 5-Me-indolo[2,3-b]quinolines with substituents at C11 and C2 were assayed for anti-proliferative activity against several cancer cells. Specifically, the 11-(3-aminopropylamino)-substituted compounds 194 and 195 ( Figure 9B ) had the best cytotoxic activity against breast cancer MDA-MB-453 cells (IC50 = 0.30–0.50 μM) [52, 80, 87]. A synergistic effect was observed with an electron-donating group, such as methoxyl at C2 in compound 195 (IC50 = 0.20 μM) [60, 71, 88]. Further modification of the terminal free amino group of the lariat attachment at C11 into the corresponding acylamides and 2,3-dihydrobenzo[e] [1, 3] thiazin-4-ones was not effective with respect to anti-proliferative activity [26]. However, the novel compounds may have other unique mode of actions, which would also be an interesting possibility to examine in corollary studies [89].

            3.9 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against leukemia

            Leukaemia is a malignant clonal disease originating from hematopoietic stem cells with a complex pathogenesis involving multiple biological, physical, chemical, genetic, and other hematologic factors, such as myelodysplasia, myelofibrosis, multiple myeloma [90]. The most common and studied leukemias are acute myeloid leukemia (AML), acute lymphoblastic leukemia/lymphoma (ALL), chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), and chronic myeloid leukemia (CML) [91]. The alkaloids, which differ in the orientation of the indole and quinoline rings, have good cytotoxic activity against cancer cells and possess anti-bacterial and anti-parasitic properties. As early as 1991, Harker et al. [92] investigated the cytotoxic effects of cryptolepine and neocryptolepine on HL-60/MX2 cells and mitoxantrone-resistant HL-60/MX2 cells. Dassonneville et al. [41] assessed the cytotoxicity and effects of these compounds on the cell cycle in mouse and human leukemia cells. Cryptolepine and neocryptolepine were shown to significantly arrest mouse leukemia P388 cells at the G2/M phase. In human leukemia HL-60 cells, cryptolepine treatment resulted in the appearance of a sub-G1 peak below the diploid DNA content and representative of the apoptotic cell population. However, the use of an HL-60/MX2 cell line resistant to the anti-cancer drug, mitoxantrone, indicated that topoisomerase II may not be the primary cellular target of these alkaloids because both alkaloids were only two-fold less toxic to drug-resistant HL-60rMX2 cells than parental cells. The study also examined the ability of these drugs to induce apoptosis in human leukemia HL-60 cells. Western blot analysis showed that cryptolepine induced cleavage of poly-ADP-ribose polymerase and cryptolepine and neocryptolepine induced cytochrome C release from mitochondria. Cleavage of poly-ADP-ribose polymerase was associated with a strong activation of the caspase pathway, which correlated with the significant sub-G1 peak seen in cryptolepine-treated cell cycle experiments. Despite activation of the caspase cascade reaction, the investigators did not detect inter-nucleosome cleavage of DNA in these alkaloid-treated HL-60 cells.

            Zhou et al. [86] synthesized quinoline derivatives 198-207 ( Figure 9C ) by modification and compound 200 showed the most potent inhibition (50% inhibition at 0.44 mM in the cell-free telomeric report amplification protocol [TRAP]). Compound 200 is highly selective for telomerase and K562 cells have significant cell growth arrest and the cell senescence phenotype after 35 days of treatment with compound 200. Compound 200 reproducibly inhibited telomerase activity of cancer cells, leading to telomere shortening and subsequent cellular senescence. As early as 1988 Kaczmarek et al. [93] synthesized tetracyclic or pentacyclic benzo-iso-alpha-carboline-based compounds and evaluated the anti-tumor properties. The results clearly showed that the size and shape of the compounds had a significant influence on the biological activity. Among the compounds, compound 208 ( Figure 9C ), a neocryptolepine-like compound with a linear tetracyclic portion bearing two methyl groups at both N-5 and C-11 positions, was the best cytotoxic compound. Further studies showed that compound 208 significantly inhibited tumor growth in mice using the P388 and B16 cell-derived xenograft tumour model in vivo.

            Wang et al. [52] synthesized a series of neocryptolepine (210 and 211) and isocryptolepine derivatives (212; Figure 9C ). The anti-proliferative activity of these compounds were then tested against MV4-11, HCT116, A549, and BALB/3T3 cells in vitro. The cytotoxicity was much better in MV4-11 cells than HCT116 and A549 cells [52]. The anti-proliferative effects of ester groups in vitro and structure-activity relationship studies of 5-methyl-5H-indolo[2,3-b]quinoline (neocryptolepine) derivatives was described by Lu et al. [94]. C-2 and/or C-9 ester-substituted neocryptolepine derivatives were synthesized, starting from indole-3-carboxylates with ester groups and N-methylaniline. Various aminoalkylamino substituents were further attached to the C-11 position of these ester-substituted neocryptolepine and anti-proliferative assays in vitro were carried out by varying the substituents at the C-11 position of neocryptolepine and the position of the ester group in the A and/or D rings. The anti-proliferative activity of the compounds was improved by introducing an ester substituent at the C-9 position ( Figure 5 ). Among the compounds, methyl 11-(3-aminopropylamino)-5-methyl-5H-indolo[2,3-b]quinoline-9-carboxylate (compound 213; Figure 10A ) was the most potent compound with an IC50 value of 0.04 μM against human leukemia MV4-11 cells. Shaban et al. [43] prepared a series of 11-substituted neocryptolepine derivatives with branched ω-aminoalkylamino chains with different linkage lengths between the two nitrogen atoms and evaluated the anti-proliferative activity using MV4-11, A549, HCT116, and BALB/3T3 cells. All the synthesized compounds showed potent anti-proliferative activity against MV4-11 cells compared to 11-chloro substituted precursors in vitro. Due to the diversification of ω-amino alkyl amino chains, the 11-(3-amino-2-hydroxy)propylamino substituted compounds (214 and 215; Figure 10B ) were shown to have the best anti-proliferative activity with mean IC50 values of 0.04 μM and 0.06 μM against MV4-11 cells, respectively. Peng et al. [87] described the design and synthesis of a series of novel 11-aminochromeno[2,3-b]indole derivatives and evaluated the anti-proliferative activity of these derivatives using MV4-11, A549, HCT116, and BALB/3T3 cell lines. Compound 216 ( Figure 10C ) showed good anti-proliferative activity against MV4-11 leukemia cells with an IC50 of 0.12 μM [87].

            Next follows the figure caption
            Figure 10 |

            Synthetic route of compounds 213-219 [43, 64, 87, 94].

            3.10 Anti-cancer activity of neocrytolepine and neocrytolepine derivatives against other cancer

            As natural products with good anticancer activity, neocryptolepine and its derivatives are manifested in the treatment of common generalized cancers such as gastric cancer, colorectal cancer and liver cancer [49, 55]. Several studies have shown that neocryptolepine still exhibited good cytotoxicity in other cancers, such as melanoma, cholangiocarcinoma, cervical cancer, and oral epithelial cancer [22]. In addition, researchers obtained different neocryptolepine derivatives by structural modification of neocryptoleine. Overall, neocryptolepine and its derivatives have great prospects for the development of antitumor drugs and provide promising ideas for the combination therapy of cancer.

            3.10.1 Melanoma

            Melanoma is a highly malignant skin tumor caused by melanocytes or primitive nevus cells of the skin. The incidence of melanoma is increasing year-after-year and is difficult to treat [95]. Early-stage melanoma can be successfully treated with surgical intervention but advanced metastatic melanoma has a poor prognosis and requires targeted therapy, chemotherapy, and immunotherapy [96]. The efficacy of current treatments is limited by drug resistance or immune tolerance in melanoma patients [97, 98]. In recent years, the application of neocryptolepine and its derivatives in the treatment of melanoma has received increasing attention. Majhi et al. [64] obtained a series of derivatives by structural modification of neocryptolepine and evaluated the cytotoxic activity in various cancer cells. Compounds 6, 217, 218, and 219 ( Figures 1 and 10D ) had better cytotoxicity in melanoma B16F10 cells [64]. Wang et al. [52] also obtained a variety of new derivatives by the introduction of different groups in the parent nucleus of cryptolepine or neocryptolepine. Compound 175 showed strong cytotoxic activity against melanoma LOX-IMVI cells with an IC50 of 0.75 μM [52].

            3.10.2 Cervical cancer

            Cervical cancer is one of the most common malignant tumors among women worldwide and has high morbidity and mortality rates [99]. Although traditional treatments, such as surgery, radiotherapy, and chemotherapy, have achieved some efficacy in the treatment of cervical cancer, many challenges still exist, such as side effects and drug resistance [100, 101]. Therefore, searching for novel leading compounds with high efficiency and low toxicity is a strategy to develop drugs for the treatment of cervical cancer [102]. Neocryptolepine and its derivatives have gradually shown potential for the treatment of cervical cancer because of unique pharmacologic effects and good anti-cancer activity [103]. Cybulski et al. [103] obtained a series of indoquinoline derivatives and had good inhibitory effects in pancreatic cancer. The IC50 of compounds 220 and 221 ( Figure 11A ) were 808.75 ± 91.29 and 203.15 ± 35.28 nM against cervical cancer Hela cells and the molecular docking results also verified the target affinity [103]. The potential of neocryptolepine derivatives to inhibit the progression of cervical carcinogenesis has also been demonstrated in a study published by Majhi et al. [64]. Compounds 6 and 139 showed significant cytotoxicity in Hela cells [64]. In the future, studies involving neocryptolepine and its derivatives in the treatment of cervical cancer will continue to deepen and be optimized.

            Next follows the figure caption
            Figure 11 |

            Synthesis routes of neocryptolepine derivatives 220-228 [103106].

            3.10.3 Cholangiocarcinoma

            Cholangiocarcinoma (CCA) is a highly malignant solid tumor of the digestive system and is the second most common primary hepatobiliary malignancy after HCC. The incidence and mortality of CCA have gradually increased worldwide in the last decade and surgical resection remains the most desirable radical therapy for CCA patients [107, 108]. However, the majority of patients are diagnosed in mid- to late-stage because of the atypical clinical presentation and insidious onset of this cancer. Patients with end-stage CCA are usually unable to undergo surgical resection or local intervention. The National Comprehensive Cancer Network (NCCN) guidelines recommend gemcitabine combined with cisplatin (GC) as first-line chemotherapy for end-stage CCA [109]. Therefore, it is necessary to search for effective drugs for the treatment of CCA and the continuous exploration of neocryptolepine derivatives has provided such a possibility. Neocryptolepine derivatives 151, 152, 153, 161, and 164 exhibit selective anti-cancer activity against CCA HuCCA-1 cells in vitro as demonstrated by Akkachairin et al. [7], who showed good cytotoxicity with and IC50 of 13.13 ± 1.82 μM, 13.39 ± 1.68 μM, 13.63 ± 0.00 μM, 13.53 ± 0.42 μM, and 9.65 ± 1.57 μM respectively. Moreover, topoisomerase II has been shown to be a target of these compounds by DNA binding in several cancer cells based on several studies involving C. alba and C. neoformans.

            3.10.4 Oral epidermal cancer

            In addition to good antitumor potential against solid tumors of organs, neocryptolepine and its derivatives have good inhibitory ability against some specific types of epidermal cancers. The incidence of oral epidermal cancer ranks fifth among all cancers globally and the prevalence has increased with the aging population [110]. Surgical intervention is usually the main choice for the treatment of oral squamous cell carcinomas (OSCC). However, after successful surgery, the 5-year survival rate for OSCC patients is still very low [111, 112]. In addition, long-term survivors may experience severe complications related to occlusion, swallowing, and word articulation, along with facial distortion and psychological disorders [113115]. Therefore, non-surgical treatment options, such as immunotherapy and gene therapy, are being actively investigated [111]. Currently, basic and clinical research are focused on the development of anti-cancer drugs with selective antitumor effects, especially for advanced or recurrent tumors. Several studies have demonstrated that neocryptolepine and its derivatives have the potential for development of oral epidermoid carcinoma. Sidoryk et al. [104] obtained a series of neocryptolepine derivatives and the results of cytotoxic assays demonstrated that a variety of compounds possess cytotoxic activity against oral epidermoid carcinoma KB cells. Compound 222 ( Figure 11B ) had the strongest inhibition against KB cells compared to other neocryptolepine derivatives and the IC50 against KB cells was 0.38 ± 0.11 μM [104]. The neocryptolepine derivatives synthesized by Li et al. [77] also had good antiproliferative effects in oral epidermal cancer KB cells with IC50 values of 0.08, 0.31, 0.15, 0.15, 0.64, and 0.36 μM for compounds 223-228 ( Figure 11C-E ) against KB cells, respectively [77]. These results also provided a new strategy for the optimization of neocryptolepine and provided promising leading compounds for the treatment of oral epidermal cancer.

            4. CONCLUSION AND PERSPECTIVE

            Overall, neocryptolepine and its derivatives exhibit broad-spectrum anti-tumor activity in different cancer cells, including gastric, colorectal, liver, lung, and esophageal cancer. Compounds 43, 65, 93, and 96 exert excellent cytotoxicity in gastric cancer cells and these compounds inhibit cell migration and proliferation, arrest the cell cycle, and induce cell apoptosis. Compound 43 showed better cytotoxicity in AGS cells with an IC50 of 43 nM. Moreover, compound 65 showed good anti-cancer activity in human gastric cancer organoid and low toxicity in mice.

            As shown in Table 2 and Figure 12 , most neocryptolepine derivatives exert good cytotoxicity by binding DNA or inhibiting topoisomerase II. Many compounds induce apoptosis of cancer cells and increase the levels of apoptotic protein expression, such as p53, caspase 8, and caspase 3. The PI3K/AKT and Wnt/β-catenin cell signaling pathways have an important role in growth inhibition of neocryptolepine derivatives. In a structure-activity relationship analysis, the double substitution of the B and D rings was more common in the substitution of the neocryptolepine as the parent nucleus. As seen in Figure 5 , the hydrazine and amide linkers have an important role in exerting good cytotoxicity in the B ring substitution. The 8-position F, methyl, or methoxy substitution is important to maintain the cytotoxicity in A and D ring substitutions. Among the B and D ring-substituted compounds, the IC50 of compounds 93 and 96 against gastric cancer AGS cells were 2.9 and 4.5 μM, respectively.

            Table 2 |

            Preclinical studies of neocryptolepine and its derivatives in different types of cancers.

            Type of cancerCompoundsExperiments
            Effects and mechanismsReferences
            In vivo In vitro
            Gastric cancerNeocryptolepine derivative 43 NAAGSE-cadherin↑, CDK1↓, Cleased-caspase3↑, PI3KCA↓, p-AKT↓, AKT↓[19]
            Gastric cancerNeocryptolepine derivative 65, 93, 96 NAAGS, HGC27, MKN45, SGC7901CDK1↓, Cyclin B1↓, caspase 3↑, PI3KCA↓, p-AKT↓, AKT↓, mTOR↓, p-mTOR↓[48, 49]
            Colorectal cancerCompound 6 NAHCT116β-catenin↓, c-MYC↓, WNT3a↓, WISP1↓[58]
            Colorectal cancerCompound 64 NAHCT116, Caco-2CDK1↓, Cyclin B1↓, PI3KCA↓, p-AKT↓, AKT↓, mTOR↓, p-mTOR↓[60]
            Colorectal cancerCompound 107 NAHCT116c-MYC↓[50]
            Colorectal cancerCompound 108 NAHCT116NA[52]
            Colorectal cancerCompound 109 NAHCT116DNA intercalation[51]
            Colorectal cancerCompound 110 NAHCT116caspase3↑, p53↑, PCNA↓, Ki-67↓, VEGF↓[55]
            Ovarian cancerCompound 134 NAOVCAR3DNA binding[64]
            Liver cancerCompound 135 and 136 NAHepG-2DNA binding[46]
            Ehrlich ascites carcinomaCompound 140 Xenograft mice for Ehrlich ascites carcinoma cellsEhrlich ascites carcinoma cellsMalonaldehyde↑, caspase-8↑, mTOR↓, PTEN↑, AKT↓[70]
            Ehrlich ascites carcinomaCompound 144 The cells were relocated in the peritoneum of Swiss albino female miceEAC cellsInduced apoptosis, cell cycle arrest, TNF-α↑[42, 71]
            Lung cancerCompound 152 NAA549Inhibition of topoisomerase II[7]
            Lung cancerCompound 113 NAA549NA[53]
            Lung cancerNeocryptolepine derivatives 168-193 NAA549Interacting with DNA[52]
            Breast cancerCompounds 194 and 195 NAMDA-MB-453Interacting with DNA[52, 80, 87]
            LeukemiaCompound 200 NAK562Telomerase inhibitors[86]
            LeukemiaCompound 208 Xenograft mice for P388, L1210 cellsP388, L1210NA[93]
            LeukemiaCompounds 214 and 215 NAMV4-11Inhibition of topoisomerase II[43]
            LeukemiaCompound 216 NAMV4-11NA[87]
            MelanomaCompounds 6, 217, 218, and 219 NAB16F10DNA binding[64]
            MelanomaCompound 175 NALOX-IMVIInteracting with DNA[52]
            Cervical cancerCompounds 220 and 221 NAHelabinding to the topoisomerase II–DNA complex[103]
            Cervical cancerCompounds 6 and 139 NAHelaDNA binding[64]
            CholangiocarcinomaCompounds 151, 152, 153, 161, and 164 NAHuCCA-1NA[7]
            Next follows the figure caption
            Figure 12 |

            The potential mechanism underlying the cytotoxic effects of neocryptolepine and its derivatives (this Figure was drawn by Figdraw, ID: SWRYU0002d).

            It is also very promising to develop more effective and less toxic anti-tumor drugs by structural modification and optimization of the parent nucleus with active neocryptolepine derivatives. Although progress has been made in the field of anti-tumor research involving neocryptolepine derivatives and some neocryptolepine derivatives with good activity have been found, there are still many challenges. As seen in Table 2 , the cytotoxicity and mechanism of action of most neocryptolepine derivatives have only been evaluated at the cellular level. It is crucial to further determine the anti-tumor activity of neocryptolepine derivatives at the animal and organoid levels, and even in clinical trials. There is no toxicity data on normal cells and animals or pharmacokinetic data in vivo in the anti-tumor studies involving neocryptolepine derivatives. Therefore, there is a long way to go before clinical trials and clinical use of neocryptolepine derivatives. Finally, it is extremely important to further clarify the underlying molecular mechanism and target of the anti-tumor effect of neocryptolepine derivatives, which is also a focus in the future.

            In conclusion, this review summarized the structure and cytotoxicity of neocryptolepine and its derivatives in the last few years. We found many neocryptolepine derivatives exert good cytoxicity and the cytotoxic effects in vitro and the toxicity and anti-cancer activity in vivo were evaluated. Some neocryptolepine derivatives in this review can be used as leading compounds for the development of anti-cancer drug in future studies.

            CONFLICTS OF INTEREST

            The authors declare no competing financial interests.

            REFERENCES

            1. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al.. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2024. Vol. 74:229–263

            2. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global Cancer Statistics. CA: A Cancer Journal for Clinicians. 2011. Vol. 61:69–90

            3. Siegel RL, Giaquinto AN, Jemal A. Cancer Statistics, 2024. CA: A Cancer Journal for Clinicians. 2024. Vol. 74:12–49

            4. Cao M, Tang Y, Luo Y, Gu F, Zhu Y, Liu X, et al.. Natural Compounds Modulating Mitophagy: Implications for Cancer Therapy. Cancer Letters. 2024. Vol. 582:216590

            5. Hanahan D, Weinberg RA. The Hallmarks of Cancer. Cell. 2000. Vol. 100:57–70

            6. Canu B, Fioravanti A, Orlandi P, Di Desidero T, Alì G, Fontanini G, et al.. Irinotecan Synergistically Enhances the Antiproliferative and Proapoptotic Effects of Axitinib In Vitro and Improves its Anticancer Activity In Vivo. Neoplasia (New York, N.Y.). 2011. Vol. 13:217–229

            7. Akkachairin B, Rodphon W, Reamtong O, Mungthin M, Tummatorn J, Thongsornkleeb C, et al.. Synthesis of Neocryptolepines and Carbocycle-Fused Quinolines and Evaluation of their Anticancer and Antiplasmodial Activities. Bioorganic Chemistry. 2020. Vol. 98:103732

            8. Steinmetz KA, Potter JD. Vegetables, Fruit, and Cancer Prevention: A Review. Journal of the American Dietetic Association. 1996. Vol. 96:1027–1039

            9. Monsuez JJ, Charniot JC, Vignat N, Artigou JY. Cardiac Side-Effects of Cancer Chemotherapy. International Journal of Cardiology. 2010. Vol. 144:3–15

            10. Schirrmacher V. From Chemotherapy to Biological Therapy: A Review of Novel Concepts to Reduce the Side Effects of Systemic Cancer Treatment (Review). International Journal of Oncology. 2019. Vol. 54:407–419

            11. Wang L, Li P, Feng K. EGCG Adjuvant Chemotherapy: Current Status and Future Perspectives. European Journal of Medicinal Chemistry. 2023. Vol. 250:115197

            12. Wang S, Fang K, Dong G, Chen S, Liu N, Miao Z, et al.. Scaffold Diversity Inspired by the Natural Product Evodiamine: Discovery of Highly Potent and Multitargeting Antitumor Agents. Journal of Medicinal Chemistry. 2015. Vol. 58:6678–6696

            13. Newman DJ, Cragg GM. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. Journal of Natural Products. 2020. Vol. 83:770–803

            14. Abu Samaan TM, Samec M, Liskova A, Kubatka P, Büsselberg D. Paclitaxel’s Mechanistic and Clinical Effects on Breast Cancer. Biomolecules. 2019. Vol. 9:789

            15. Zhang R, Yu J, Guo Z, Jiang H, Wang C. Camptothecin-Based Prodrug Nanomedicines for Cancer Therapy. Nanoscale. 2023. Vol. 15:17658–17697

            16. Defachelles AS, Bogart E, Casanova M, Merks JHM, Bisogno G, Calareso G, et al.. Randomized Phase II Trial of Vincristine-Irinotecan With or Without Temozolomide, in Children and Adults With Relapsed or Refractory Rhabdomyosarcoma: A European Paediatric Soft Tissue Sarcoma Study Group and Innovative Therapies for Children With Cancer Trial. Journal of Clinical Oncology. 2021. Vol. 39:2979–2990

            17. Alshammari MK, Alghazwni MK, Alharbi AS, Alqurashi GG, Kamal M, Alnufaie SR, et al.. Nanoplatform for the Delivery of Topotecan in the Cancer Milieu: An Appraisal of its Therapeutic Efficacy. Cancers (Basel). 2022. Vol. 15:65

            18. Paz-Ares L, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D, et al.. Durvalumab plus Platinum-Etoposide Versus Platinum-Etoposide in First-Line Treatment of Extensive-Stage Small-Cell Lung Cancer (CASPIAN): A Randomised, Controlled, Open-Label, Phase 3 Trial. Lancet. 2019. Vol. 394:1929–1939

            19. Ma Y-H, Ma W-T, Zhou Z-K, Huang X, Jiang X-R, Du K-J, et al.. Synthesis of 8-Fluoroneocryptolepine and Evaluation for Cytotoxic Activity against AGS Cancer Cells. Journal of Natural Products. 2022. Vol. 85:963–971

            20. Asnake S, Teklehaymanot T, Hymete A, Erko B, Giday M. Evaluation of the Antiplasmodial Properties of Selected plants in Southern Ethiopia. BMC Complementary and Alternative Medicine. 2015. Vol. 15:448

            21. Oliver-Bever B. Medicinal Plants in Tropical West Africa. II. Plants Acting on the Nervous System. Journal of Ethnopharmacology. 1983. Vol. 7:1–93

            22. Larghi EL, Bracca AB, Arroyo Aguilar AA, Heredia DA, Pergomet JL, Simonetti SO, et al.. Neocryptolepine: A Promising Indoloisoquinoline Alkaloid with Interesting Biological Activity. Evaluation of the Drug and its Most Relevant Analogs. Current Topics in Medicinal Chemistry. 2015. Vol. 15:1683–1707

            23. Tudu CK, Bandyopadhyay A, Kumar M, Radha, Das T, Nandy S, et al.. Unravelling the Pharmacological Properties of Cryptolepine and its Derivatives: A Mini-Review Insight. Naunyn-Schmiedeberg’s Archives of Pharmacology. 2023. Vol. 396:229–238

            24. Guittat L, Alberti P, Rosu F, Van Miert S, Thetiot E, Pieters L, et al.. Interactions of Cryptolepine and Neocryptolepine with Unusual DNA Structures. Biochimie. 2003. Vol. 85:535–547

            25. Cimanga K, De Bruyne T, Pieters L, Totte J, Tona L, Kambu K, et al.. Antibacterial and Antifungal Activities of Neocryptolepine, Biscryptolepine and Cryptoquindoline, Alkaloids Isolated from Cryptolepis Sanguinolenta. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 1998. Vol. 5:209–214

            26. Zhu JK, Gao JM, Yang CJ, Shang XF, Zhao ZM, Lawoe RK, et al.. Design, Synthesis, and Antifungal Evaluation of Neocryptolepine Derivatives against Phytopathogenic Fungi. Journal of Agricultural and Food Chemistry. 2020. Vol. 68:2306–2315

            27. Mir PA, Mohi-Ud-Din R, Banday N, Maqbool M, Raza SN, Farooq S, et al.. Anticancer Potential of Thymoquinone: A Novel Bioactive Natural Compound from Nigella Sativa L. Anti-cancer Agents in Medicinal Chemistry. 2022. Vol. 22:3401–3415

            28. Atanasov AG, Zotchev SB, Dirsch VM; International Natural Product Sciences Taskforce; Supuran CT. Natural Products in Drug Discovery: Advances and Opportunities. Nature Reviews Drug Discovery. 2021. Vol. 20:200–216

            29. Nasim N, Sandeep IS, Mohanty S. Plant-Derived Natural Products for Drug Discovery: Current Approaches and Prospects. The Nucleus: An International Journal of Cytology and Allied Topics. 2022. Vol. 65:399–411

            30. Luo Z, Yin F, Wang X, Kong L. Progress in Approved Drugs from Natural Product Resources. Chinese Journal of Natural Medicines. 2024. Vol. 22:195–211

            31. Wu L, Ye M, Zhang J. Vincamine Prevents Lipopolysaccharide Induced Inflammation and Oxidative Stress via Thioredoxin Reductase Activation in Human Corneal Epithelial Cells. American Journal of Translational Research. 2018. Vol. 10:2195–2204

            32. Qiu S, Sun H, Zhang AH, Xu HY, Yan GL, Han Y, et al.. Natural Alkaloids: Basic Aspects, Biological Roles, and Future Perspectives. Chinese Journal of Natural Medicines. 2014. Vol. 12:401–406

            33. Gaur S, Wang Y, Kretzner L, Chen L, Yen T, Wu X, et al.. Pharmacodynamic and Pharmacogenomic Study of the Nanoparticle Conjugate of Camptothecin CRLX101 for the Treatment of Cancer. Nanomedicine: Nanotechnology, Biology, and Medicine. 2014. Vol. 10:1477–1486

            34. Moudi M, Go R, Yien CY, Nazre M. Vinca Alkaloids. International Journal of Preventive Medicine. 2013. Vol. 4:1231–1235

            35. Dang TT, Onoyovwi A, Farrow SC, Facchini PJ. Biochemical Genomics for Gene Discovery in Benzylisoquinoline Alkaloid Biosynthesis in Opium Poppy and Related Species. Methods in Enzymology. 2012. Vol. 515:231–266

            36. Joshi P, Vishwakarma RA, Bharate SB. Natural Alkaloids as P-gp Inhibitors for Multidrug Resistance Reversal in Cancer. European Journal of Medicinal Chemistry. 2017. Vol. 138:273–292

            37. Gellért E, Raymond-Hamet, Schlittler E. Die Konstitution des Alkaloids Cryptolepin. Helvetica Chimica Acta. 1951. Vol. 34:642–651

            38. Aroonkit P, Thongsornkleeb C, Tummatorn J, Krajangsri S, Mungthin M, Ruchirawat S. Synthesis of Isocryptolepine Analogues and their Structure-Activity Relationship Studies as Antiplasmodial and Antiproliferative Agents. European Journal of Medicinal Chemistry. 2015. Vol. 94:56–62

            39. Shang XF, Morris-Natschke SL, Yang GZ, Liu YQ, Guo X, Xu XS, et al.. Biologically Active Quinoline and Quinazoline Alkaloids Part II. Medicinal Research Reviews. 2018. Vol. 38:1614–1660

            40. Nuthakki VK, Mudududdla R, Sharma A, Kumar A, Bharate SB. Synthesis and Biological Evaluation of Indoloquinoline Alkaloid Cryptolepine and its Bromo-Derivative as Dual Cholinesterase Inhibitors. Bioorganic Chemistry. 2019. Vol. 90:103062

            41. Dassonneville L, Lansiaux A, Wattelet A, Wattez N, Mahieu C, Van Miert S, et al.. Cytotoxicity and Cell Cycle Effects of the Plant Alkaloids Cryptolepine and Neocryptolepine: Relation to Drug-Induced Apoptosis. European Journal of Pharmacology. 2000. Vol. 409:9–18

            42. Altwaijry N, El-Ghlban S, El Sayed IE, El-Bahnsawye M, Bayomi AI, Samaka RM, et al.. In Vitro and In Vivo Antitumor Activity of Indolo[2,3-b]quinolines, Natural Product Analogs from Neocryptolepine Alkaloid. Molecules. 2021. Vol. 26:754

            43. Shaban E, Świtalska M, Wang L, Wang N, Xiu F, Hayashi I, et al.. Synthesis and In Vitro Antiproliferative Activity of 11-Substituted Neocryptolepines with a Branched ω-Aminoalkylamino Chain. Molecules. 2017. Vol. 22:1954

            44. Huang X, Zhang YB, Yang XW. Indoloquinazoline Alkaloids from Euodia Rutaecarpa and their Cytotoxic Activities. Journal of Asian Natural Products Research. 2011. Vol. 13:977–983

            45. Rizo WF, Ferreira LE, Colnaghi V, Martins JS, Franchi LP, Takahashi CS, et al.. Cytotoxicity and Genotoxicity of Coronaridine from Tabernaemontana Catharinensis A.DC in a Human Laryngeal Epithelial Carcinoma Cell Line (Hep-2). Genetics and Molecular Biology. 2013. Vol. 36:105–110

            46. El-Bahnsawye M, Hussein MKA, Elmongy EI, Awad HM, Tolan AAE, Moemen YS, et al.. Design, Synthesis, and Antiproliferative Activity of Novel Neocryptolepine-Rhodanine Hybrids. Molecules. 2022. Vol. 27:7599

            47. Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A. Alkaloids for Cancer Prevention and Therapy: Current Progress and Future Perspectives. European Journal of Pharmacology. 2019. Vol. 858:172472

            48. Ma Y, Tian Y, Zhou Z, Chen S, Du K, Zhang H, et al.. Design, Synthesis and Biological Evaluation of Neocryptolepine Derivatives as Potential Anti-Gastric Cancer Agents. International Journal of Molecular Sciences. 2022. Vol. 23:11924

            49. Ma Y, Xu H, Zhou Z, Tian Y, Du K, Zhang H, et al.. CFNC, a Neocryptolepine Derivative, Inhibited the Growth of Gastric Cancer AGS Cells by Inhibiting PI3K/AKT Signaling Pathway. European Journal of Pharmacology. 2023. Vol. 938:175408

            50. Boddupally PV, Hahn S, Beman C, De B, Brooks TA, Gokhale V, et al.. Anticancer Activity and Cellular Repression of c-MYC by the G-Quadruplex-Stabilizing 11-Piperazinylquindoline is Not Dependent on Direct Targeting of the G-Quadruplex in the c-MYC Promoter. Journal of Medicinal Chemistry. 2012. Vol. 55:6076–6086

            51. Wang L, Świtalska M, Mei ZW, Lu WJ, Takahara Y, Feng XW, et al.. Synthesis and In Vitro Antiproliferative Activity of New 11-Aminoalkylamino-Substituted 5H- and 6H-Indolo[2,3-b]Quinolines; Structure-Activity Relationships of Neocryptolepines and 6-Methyl Congeners. Bioorganic & Medicinal Chemistry. 2012. Vol. 20:4820–4829

            52. Wang N, Świtalska M, Wang L, Shaban E, Hossain MI, El Sayed IET, et al.. Structural Modifications of Nature-Inspired Indoloquinolines: A Mini Review of their Potential Antiproliferative Activity. Molecules. 2019. Vol. 24:2121

            53. Ahmed AAS, Awad HM, El-Sayed IE-T, El Gokha AA. Synthesis and Antiproliferative Activity of New Hybrids Bearing Neocryptolepine, Acridine and α-Aminophosphonate Scaffolds. Journal of the Iranian Chemical Society. 2020. Vol. 17:1211–1221

            54. Håheim KS, Lindbäck E, Tan KN, Albrigtsen M, Urdal Helgeland IT, Lauga C, et al.. Synthesis and Evaluation of the Tetracyclic Ring-System of Isocryptolepine and Regioiso-Mers for Antimalarial, Antiproliferative and Antimicrobial Activities. Molecules. 2021. Vol. 26:3268

            55. Nagy ET, Ahmed AAS, Elmongy EI, El-Gendy SM, Elmadbouh I, El Sayed IET, et al.. Design and Cytotoxic Evaluation via Apoptotic and Antiproliferative Activity for Novel 11(4-Aminophenylamino)Neocryptolepine on Hepatocellular and Colorectal Cancer Cells. Apoptosis. 2023. Vol. 28:653–668

            56. Sidoryk K, Świtalska M, Wietrzyk J, Jaromin A, Piętka-Ottlik M, Cmoch P, et al.. Synthesis and Biological Evaluation of New Amino Acid and Dipeptide Derivatives of Neocryptolepine as Anticancer Agents. Journal of Medicinal Chemistry. 2012. Vol. 55:5077–5087

            57. Matsui TA, Sowa Y, Murata H, Takagi K, Nakanishi R, Aoki S, et al.. The Plant Alkaloid Cryptolepine Induces p21WAF1/CIP1 and Cell Cycle Arrest in a Human Osteosarcoma Cell Line. International Journal of Oncology. 2007. Vol. 31:915–922

            58. Quarshie JT, Fosu K, Offei NA, Sobo AK, Quaye O, Aikins AR. Cryptolepine Suppresses Colorectal Cancer Cell Proliferation, Stemness, and Metastatic Processes by Inhibiting WNT/β-Catenin Signaling. Pharmaceuticals (Basel, Switzerland). 2023. Vol. 16:1026

            59. Abd Elrahman SF, Ahmed AAS, Abd Elsatar D, Elkady S, Elgendy A, Alnakeeb F, et al.. Cytotoxic Potential of Novel Quinoline Derivative: 11-(1,4-Bisaminopropylpiperazinyl)5-methyl-5H-indolo[2,3-b]quinoline against Different Cancer Cell Lines via Activation and Deactivation of the Expression of Some Proteins. International Journal of Molecular Sciences. 2023. Vol. 24:14336

            60. Ma Y, Zhu H, Jiang X, Zhou Z, Zhou Y, Tian Y, et al.. Biological Evaluation of 8-Methoxy-2,5-dimethyl-5H-indolo[2,3-b]quinoline as a Potential Antitumor Agent via PI3K/AKT/mTOR Signaling. International Journal of Molecular Sciences. 2023. Vol. 24:15142

            61. Webb PM, Jordan SJ. Global Epidemiology of Epithelial Ovarian Cancer. Nature Reviews. Clinical Oncology. 2024. Vol. 21:389–400

            62. Stewart C, Ralyea C, Lockwood S. Ovarian Cancer: An Integrated Review. Seminars in Oncology Nursing. 2019. Vol. 35:151–156

            63. McMullen M, Karakasis K, Rottapel R, Oza AM. Advances in Ovarian Cancer, from Biology to Treatment. Nature Cancer. 2021. Vol. 2:6–8

            64. Majhi B, Ganguly S, Palit S, Parwez A, Saha R, Basu G, et al.. Sequence-Specific Dual DNA Binding Modes and Cytotoxicities of N-6-Functionalized Norcryptotackieine Alkaloids. Journal of Natural Products. 2023. Vol. 86:1667–1676

            65. Centers for Disease Control and Prevention (CDC). Hepatocellular Carcinoma - United States, 2001-2006. MMWR. Morbidity and Mortality Weekly Report. 2010. Vol. 59:517–520

            66. Grieb BC, Goff LW, Goyal L, Denlinger CS. Evolving Landscape of Systemic Therapy for Hepatocellular Carcinoma: Breakthroughs, Toxicities, and Future Frontiers. American Society of Clinical Oncology Educational Book. American Society of Clinical Oncology. Annual Meeting. 2019. Vol. 39:248–260

            67. Luo M-L, Huang W, Zhu H-P, Peng C, Zhao Q, Han B. Advances in Indole-Containing Alkaloids as Potential Anticancer Agents by Regulating Autophagy. Biomedicine & Pharmacotherapy. 2022. Vol. 149:112827

            68. Cushnie TPT, Cushnie B, Lamb AJ. Alkaloids: An Overview of their Antibacterial, Antibiotic-Enhancing and Antivirulence Activities. International Journal of Antimicrobial Agents. 2014. Vol. 44:377–386

            69. Dey P, Kundu A, Kumar A, Gupta M, Lee BM, Bhakta T, et al.. Analysis of Alkaloids (Indole Alkaloids, Isoquinoline Alkaloids, Tropane Alkaloids)Recent Advances in Natural Products Analysis. Sanches Silva A, Nabavi SF, Saeedi M, Nabavi SM. Elsevier. 2020. p. 505–567

            70. El-Aarag B, Shalaan ES, Ahmed AAS, El Sayed IET, Ibrahim WM. Cryptolepine Analog Exhibits Antitumor Activity against Ehrlich Ascites Carcinoma Cells in Mice via Targeting Cell Growth, Oxidative Stress, and PTEN/Akt/mTOR Signaling Pathway. Anti-Cancer Agents in Medicinal Chemistry. 2024. Vol. 24:436–442

            71. Nofal AE, Elmongy EI, Hassan EA, Tousson E, Ahmed AAS, El Sayed IET, et al.. Impact of Synthesized Indoloquinoline Analog to Isolates from Cryptolepis sanguinolenta on Tumor Growth Inhibition and Hepatotoxicity in Ehrlich Solid Tumor-Bearing Female Mice. Cells. 2023. Vol. 12:1024

            72. Tian Y, Zhang YZ, Chen W. MicroRNA-199a-3p and microRNA-34a Regulate Apoptosis in Human Osteosarcoma Cells. Bioscience Reports. 2014. Vol. 34:479–485

            73. Harisi R, Dudás J, Tímár F, Pogány G, Paku S, Tímár J, et al.. Antiproliferative and Antimigratory Effects of Doxorubicin in Human Osteosarcoma Cells Exposed to Extracellular Matrix. Anticancer Research. 2005. Vol. 25:805–813

            74. Ma Q, Li X, Wang H, Xu S, Que Y, He P, et al.. HOXB5 Promotes the Progression and Metastasis of Osteosarcoma Cells by Activating the JAK2/STAT3 Signalling Pathway. Heliyon. 2024. Vol. 10:e30445

            75. Matsuda H, Yoshida K, Miyagawa K, Nemoto Y, Asao Y, Yoshikawa M. Nuphar Alkaloids with Immediately Apoptosis-Inducing Activity from Nuphar pumilum and their Structural Requirements for the Activity. Bioorganic & Medicinal Chemistry Letters. 2006. Vol. 16:1567–1573

            76. Ahmed F, Toume K, Ohtsuki T, Rahman M, Sadhu SK, Ishibashi M. Cryptolepine, Isolated from Sida acuta, Sensitizes Human Gastric Adenocarcinoma Cells to TRAIL-Induced Apoptosis. Phytotherapy Research. 2011. Vol. 25:147–150

            77. Sidoryk K, Jaromin A, Edward JA, Świtalska M, Stefańska J, Cmoch P, et al.. Searching for New Derivatives of Neocryptolepine: Synthesis, Antiproliferative, Antimicrobial and Antifungal Activities. European Journal of Medicinal Chemistry. 2014. Vol. 78:304–313

            78. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al.. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021. Vol. 71:209–249

            79. Lovly CM. Expanding Horizons for Treatment of Early-Stage Lung Cancer. The New England Journal of Medicine. 2022. Vol. 386:2050–2051

            80. Okada M, Mei ZW, Hossain MI, Wang L, Tominaga T, Takebayashi T, et al.. Synthesis and In Vitro Cancer Cell Growth Inhibition Evaluation of 11-Amino-Modified 5-Me-Indolo[2,3-b]quinolines and their COMPARE Analyses. Medicinal Chemistry Research. 2016. Vol. 25:879–892

            81. Riou JF, Helissey P, Grondard L, Giorgi-Renault S. Inhibition of Eukaryotic DNA Topoisomerase I and II Activities by Indoloquinolinedione Derivatives. Molecular Pharmacology. 1991. Vol. 40:699–706

            82. Bonjean K, De Pauw-Gillet MC, Defresne MP, Colson P, Houssier C, Dassonneville L, et al.. The DNA Intercalating Alkaloid Cryptolepine Interferes with Topoisomerase II and Inhibits Primarily DNA Synthesis in B16 Melanoma Cells. Biochemistry. 1998. Vol. 37:5136–5146

            83. Van Miert S, Jonckers T, Cimanga K, Maes L, Maes B, Lemière G, et al.. In Vitro Inhibition of Beta-haematin Formation, DNA Interactions, Antiplasmodial Activity, and Cytotoxicity of Synthetic Neocryptolepine Derivatives. Experimental Parasitology. 2004. Vol. 108:163–168

            84. Wang L, Świtalska M, Mei ZW, Lu WJ, Takahara Y, Feng XW, et al.. Synthesis and In Vitro Antiproliferative Activity of New 11-Aminoalkylamino-Substituted 5H- and 6H-Indolo[2,3-b]quinolines; Structure–Activity Relationships of Neocryptolepines and 6-Methyl Congeners. Bioorganic & Medicinal Chemistry. 2012. Vol. 20:4820–4829

            85. Amer M. Improved Synthesis of Neocryptolepine-type Indoloquinolines: Applications for New Fluorescent Labelled Derivatives and Anticancer Activity Evaluations. United Kingdom: The University of Manchester. 2016

            86. Zhou JM, Zhu XF, Lu YJ, Deng R, Huang ZS, Mei YP, et al.. Senescence and Telomere Shortening Induced by Novel Potent G-Quadruplex Interactive Agents, Quindoline Derivatives, in Human Cancer Cell Lines. Oncogene. 2006. Vol. 25:503–511

            87. Peng W, Świtalska M, Wang L, Mei ZW, Edazawa Y, Pang CQ, et al.. Synthesis and In Vitro Antiproliferative Activity of New 11-Aminoalkylamino-Substituted Chromeno[2,3-b]Indoles. European Journal of Medicinal Chemistry. 2012. Vol. 58:441–451

            88. Le Gresley A, Gudivaka V, Carrington S, Sinclair A, Brown JE. Synthesis, Analysis and Biological Evaluation of Novel Indolquinonecryptolepine Analogues as Potential Anti-Tumour Agents. Organic & Biomolecular Chemistry. 2016. Vol. 14:3069–3079

            89. Jonckers TH, van Miert S, Cimanga K, Bailly C, Colson P, De Pauw-Gillet MC, et al.. Synthesis, Cytotoxicity, and Antiplasmodial and Antitrypanosomal Activity of new Neocryptolepine Derivatives. Journal of Medicinal Chemistry. 2002. Vol. 45:3497–3508

            90. Malard F, Mohty M. Acute Lymphoblastic Leukaemia. Lancet. 2020. Vol. 395:1146–1162

            91. Whiteley AE, Price TT, Cantelli G, Sipkins DA. Leukaemia: A Model Metastatic Disease. Nature Reviews Cancer. 2021. Vol. 21:461–475

            92. Harker WG, Slade DL, Drake FH, Parr RL. Mitoxantrone Resistance in HL-60 Leukemia Cells: Reduced Nuclear Topoisomerase II Catalytic Activity and Drug-Induced DNA Cleavage in Association with Reduced Expression of the Topoisomerase II Beta Isoform. Biochemistry. 1991. Vol. 30:9953–9961

            93. Kaczmarek L, Balicki R, Nantka-Namirski P, Peczyńska-Czoch W, Mordarski M. Cancerostatics, VI. Synthesis and Antineoplastic Properties of Some Benzo-Iso-Alpha-Carbolines. Arch Pharm (Weinheim). 1988. Vol. 321:463–467

            94. Lu WJ, Świtalska M, Wang L, Yonezawa M, El-Sayed IET, Wietrzyk J, et al.. In Vitro Antiproliferative Activity of 11-Aminoalkylamino-Substituted 5H-Indolo[2,3-b]quinolines; Improving Activity of Neocryptolepines by Installation of Ester Substituent. Medicinal Chemistry Research. 2013. Vol. 22:4492–4504

            95. Malekan M, Haass NK, Rokni GR, Gholizadeh N, Ebrahimzadeh MA, Kazeminejad A. VEGF/VEGFR Axis and its Signaling in Melanoma: Current Knowledge Toward Therapeutic Targeting Agents and Future Perspectives. Life Sciences. 2024. Vol. 345:122563

            96. Sussman TA, Ott PA. Adjuvant Immunotherapy for Melanoma Patients: Progress and Opportunities. ESMO Open. 2024. Vol. 9:102962

            97. Hua HK, Zhu HM, Zhang ZG. Clinical Significance of Downregulated NISCH Expression in Skin Cutaneous Melanoma: Modulation of Tumor Cell Invasion, Migration, and EMT Via PAK1 Inhibition. Tissue & Cell. 2024. Vol. 88:102399

            98. Swisher SK, Delman KA. Management of Locoregional Melanoma. Advances in Oncology. 2024. Vol. 4:9–18

            99. Guillén-Meléndez GA, Pérez-Hernández RA, Chávez-Montes A, Castillo-Velázquez U, de Jesús Loera-Arias M, Montes-de-Oca-Saucedo CR, et al.. Nanoencapsulation of Extracts and Isolated Compounds of Plant Origin and their Cytotoxic Effects on Breast and Cervical Cancer Treatments: Advantages and New Challenges. Toxicon. 2024. Vol. 244:107753

            100. Wang J, Wang Y, Bi P, Chen Z, Qiao J, Li J, et al.. Binary Organic Solar Cells with 19.2% Efficiency Enabled by Solid Additive. Advanced Materials. 2023. Vol. 35:e2301583

            101. Tao T, Zhang P, Zeng Z, Wang M. Advances in Autophagy Modulation of Natural Products in Cervical Cancer. Journal of Ethnopharmacology. 2023. Vol. 314:116575

            102. Huang J, Yang J, Zhang Y, Lu D, Dai Y. FTO Promotes Cervical Cancer Cell Proliferation, Colony Formation, Migration and Invasion Via the Regulation of the BMP4/Hippo/YAP1/TAZ Pathway. Experimental Cell Research. 2023. Vol. 427:113585

            103. Cybulski M, Sidoryk K, Zaremba-Czogalla M, Trzaskowski B, Kubiszewski M, Tobiasz J, et al.. The Conjugates of Indolo[2,3-b]quinoline as Anti-Pancreatic Cancer Agents: Design, Synthesis, Molecular Docking and Biological Evaluations. International Journal of Molecular Sciences. 2024. Vol. 25:2573

            104. Sidoryk K, Świtalska M, Jaromin A, Cmoch P, Bujak I, Kaczmarska M, et al.. The Synthesis of Indolo[2,3-b]quinoline Derivatives with a Guanidine Group: Highly Selective Cytotoxic Agents. European Journal of Medicinal Chemistry. 2015. Vol. 105:208–219

            105. Luniewski W, Wietrzyk J, Godlewska J, Świtalska M, Piskozub M, Peczynska-Czoch W, et al.. New Derivatives of 11-Methyl-6-[2-(dimethylamino)ethyl]-6H-indolo[2,3-b]quinoline as Cytotoxic DNA Topoisomerase II Inhibitors. Bioorganic & Medicinal Chemistry Letters. 2012. Vol. 22:6103–6107

            106. Li D, Yang R, Wu J, Zhong B, Li Y. Comprehensive Review of α-Carboline Alkaloids: Natural Products, Updated Synthesis, and Biological Activities. Frontiers in Chemistry. 2022. Vol. 10:988327

            107. Pascale A, Rosmorduc O, Duclos-Vallée J-C. New Epidemiologic Trends in Cholangiocarcinoma. Clinics and Research in Hepatology and Gastroenterology. 2023. Vol. 47:102223

            108. Bragazzi MC, Venere R, Ribichini E, Covotta F, Cardinale V, Alvaro D. Intrahepatic Cholangiocarcinoma: Evolving Strategies in Management and Treatment. Digestive and Liver Disease. 2024. Vol. 56:383–393

            109. Liu J, Shu J. Immunotherapy and Targeted Therapy for Cholangiocarcinoma: Artificial Intelligence Research in Imaging. Critical Reviews in Oncology/Hematology. 2024. Vol. 194:104235

            110. Lemos LM, Silva GGD, Pinheiro JC, Morais EFD, Barros CCDAS, Leite RB. Clinical and Epidemiologic Profile of Oral Epidermoid Carcinoma in Young Adults Patients: Systematic Review. Oral Surgery, Oral Medicine, Oral Pathology and Oral Radiology. 2020. Vol. 130:e274–e275

            111. Park HK, Ryu MH, Hwang DS, Kim GC, Jang MA, Kim UK. Effects of Melatonin Receptor Expression on Prognosis and Survival in Oral Squamous Cell Carcinoma Patients. International Journal of Oral and Maxillofacial Surgery. 2022. Vol. 51:713–723

            112. Fan QC, Tian H, Wang Y, Liu XB. Integrin-α5 Promoted the Progression of Oral Squamous Cell Carcinoma and Modulated PI3K/AKT Signaling Pathway. Archives of Oral Biology. 2019. Vol. 101:85–91

            113. Kim M, Kim G-H, Lee TK, Choi IW, Choi HW, Jo Y, et al.. Methylammonium Chloride Induces Intermediate Phase Stabilization for Efficient Perovskite Solar Cells. Joule. 2019. Vol. 3:2179–2192

            114. Stathopoulos P, Smith WP. Close Resection Margins Do Not Influence Local Recurrence in Patients with Oral Squamous Cell Carcinoma: A Prospective Cohort Study. Journal of Oral and Maxillofacial Surgery. 2018. Vol. 76:873–876

            115. Andrade ACDM, Brito CPD, Felix FA, França GMD, Assis GMD, Oliveira PTD, et al.. Papillary Squamous Cell Carcinoma of the Oral Cavity: A Case Report with Emphasis on Histopathologic Features. Oral Surgery, Oral Medicine, Oral Pathology and Oral Radiology. 2020. Vol. 130:e181

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            13 December 2024
            : 3
            : 4
            : 436-461
            Affiliations
            [a ]School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
            Author notes
            *Correspondence: yqliu@ 123456lzu.edu.cn (Y. Liu); chenpeng@ 123456lzu.edu.cn (P. Chen); Tel.: +86-931-8915686; Fax: +86-931-8915686
            Article
            10.15212/AMM-2024-0054
            f4b435df-3248-4bd0-a9a5-5738456d43e5
            2024 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 08 September 2024
            : 06 November 2024
            : 15 November 2024
            Page count
            Figures: 12, Tables: 2, References: 115, Pages: 26
            Funding
            Funded by: Gansu Provincial Science and Technology Major Project
            Award ID: 24ZDFA001
            Funded by: Lanzhou Municipal Science and Technology Program
            Award ID: 2022-2-52
            Funded by: Lanzhou Municipal Science and Technology Program
            Award ID: 2024-8-27
            Funded by: Lanzhou Municipal Science and Technology Program
            Award ID: 2024-8-30
            Funded by: Lanzhou Municipal Science and Technology Program
            Award ID: 2024-4-2
            Funded by: College Students’ Innovation and Entrepreneurship Program of Lanzhou University, China
            Award ID: 20240260001
            Funded by: College Students’ Innovation and Entrepreneurship Program of Lanzhou University, China
            Award ID: 20240260017
            This work was supported by Gansu Provincial Science and Technology Major Project (Grant No. 24ZDFA001), the Lanzhou Municipal Science and Technology Program (Grant Nos. 2022-2-52, 2024-8-27, 2024-8-30, and 2024-4-2), and the College Students’ Innovation and Entrepreneurship Program of Lanzhou University, China (Grant Nos. 20240260001 and 20240260017).
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            anti-tumor,neocryptolepine,anti-proliferation,cancer,alkaloid

            Comments

            Comment on this article