key: cord-0789714-1g3xbfgp authors: Schütz, Ramona; Müller, Martin; Geisslinger, Franz; Vollmar, Angelika; Bartel, Karin; Bracher, Franz title: Synthesis, biological evaluation and toxicity of novel tetrandrine analogues date: 2020-09-04 journal: Eur J Med Chem DOI: 10.1016/j.ejmech.2020.112810 sha: f7ac47e48e3e549b6265da18c8425c4b6794770f doc_id: 789714 cord_uid: 1g3xbfgp In this work, we present the design and synthesis of novel fully synthetic analogues of the bisbenzylisoquinoline tetrandrine, a molecule with numerous pharmacological properties and the potential to treat life-threatening diseases, such as viral infections and cancer. Its toxicity to liver and lungs and the underlying mechanisms, however, are controversially discussed. Along this line, novel tetrandrine analogues were synthesized and biologically evaluated for their hepatotoxicity, as well as their antiproliferative and chemoresistance reversing activity on cancer cells. Previous studies suggesting CYP-mediated toxification of tetrandrine prompted us to amend/replace the suspected metabolically instable 12-methoxy group. Of note, employing several in vitro models showed that the proposed CYP3A4-driven metabolism of tetrandrine and analogues is not the major cause of hepatotoxicity. Biological characterization revealed that some of the novel tetrandrine analogues sensitized drug-resistant leukemia cells by inhibition of the P-glycoprotein. Interestingly, direct anticancer effects improved in comparison to tetrandrine, as several compounds displayed a markedly enhanced ability to reduce proliferation of drug-resistant leukemia cells and to induce cell death of liver cancer cells. Those enhanced anticancer properties were linked to influences on activation of the kinase Akt and mitochondrial events. In sum, our study clarifies the role of CYP3A4-mediated toxicity of the bisbenzylisoquinoline alkaloid tetrandrine and provides the basis for the exploitation of novel synthetic analogues for their antitumoral potential. The natural product tetrandrine (1) (Fig. 1) , isolated from the plant Stephania tetrandra [1, 2] , belongs to the class of bisbenzylisoquinoline alkaloids. Tetrandrine has a wide range of pharmacological activities [3, 4] , most interestingly antiviral [5] [6] [7] [8] , anticancer [9, 10] , multidrug resistance reversing [11] [12] [13] [14] [15] and calcium channel blocking [6, [16] [17] [18] effects. (2) (1R,1'S) -Numbering of the skeleton according to Shamma [19] . Recently, it was shown that tetrandrine (1) blocks endolysosomal two-pore channels (TPCs, voltage-gated calcium channels) and thereby reduces cellular entry of Ebola [6] , MERS-CoV [7] , SARS-CoV-2 [5] viruses or pseudoviruses into host cells. Currently, a clinical trial (NCT04308317) is announced to investigate tetrandrine (1) as adjuvant treatment of COVID-19 patients in China [20] . Moreover, tetrandrine displayed strong antiproliferative and cytotoxic properties against several cancer cell lines [4, 9] , arrested cell cycle progression [21] , induced pro-apoptotic signaling pathways [22] and reduced migration [23] of tumor cells. Additionally, tetrandrine was shown to resensitize chemoresistant tumors by inhibition of Pglycoprotein (P-gp) [11, 12, 14] , a universal efflux pump for xenobiotics, which is a key factor of drug resistance [24] frequently causing treatment failure of tumor therapy. All these multiple pharmacological activities make tetrandrine (1) an interesting lead compound and a potential drug candidate for diverse applications. Unfortunately, its clinical application as a drug is limited by its toxicity. Several toxic side effects have been reported for tetrandrine and other bisbenzylisoquinolines so far [25] . In animal models, mainly a damage of liver and lungs was observed [26] [27] [28] [29] [30] . The molecular mechanism of tetrandrine-induced toxicity has not been entirely elucidated yet. Li and coworkers [31] hypothesized that an interaction of tetrandrine with p38α MAPK (mitogen-activated protein kinase) led to liver injury, whereas several other studies suggest the involvement of cytochrome P450 (CYP) enzymes in tetrandrine-induced pulmonary [26, 27] and hepatic toxicity [32] . Qi et al. [32] consider CYP2E1 as primary reason for mitochondrial dysfunction of rat hepatocytes after tetrandrine exposure, which they relate to reactive oxygen species (ROS) that were generated in the course of CYP2E1 metabolism. However, it is unknown which molecular mechanisms are connected to CYP2E1-mediated toxicity. In a metabolic study by Jin et al. [26] a CYP3A4-and CYP3A5-mediated metabolism equally leading to a potentially toxic intermediate is described. Hereby one particular structural element is suggested to be responsible for tetrandrine's toxicity. In the hypothesized metabolic pathway, the methoxy group at C-12 in para-position to a benzylic methylene group is first demethylated to give the corresponding phenol 3. This phenol then undergoes enzymatic oxidation to the para-quinone methide 4, which is an electrophilic intermediate prone to attack bio-nucleophiles (see Scheme 1) . After incubation of tetrandrine (1) with human liver mircosomes in the presence of glutathione (GSH), a corresponding GSH conjugate (5) was detected via LC-MS, which provides evidence for the formation of the para-quinone methide 4. Scheme 1. Postulated CYP-enzyme mediated oxidative metabolism of tetrandrine (1) adopted from Jin et al. [26] . A considerable number of pharmacologically interesting bisbenzylisoquinolines and seco analogues such as dauricine (6) [33] [34] [35] [36] , berbamine (7) [37] [38] [39] , fangchinoline (8) [7, 15] , cepharanthine (9) [40] and muraricine (10) [41, 42] contain the discussed paramethoxybenzyl motif like the one present in tetrandrine (1) or the equivalent parahydroxybenzyl moiety (see Fig. 2 ), which could potentially result in an unfavorable toxicity profile as well. So it is of great relevance to clarify if and to which extent this structural motif causes cytotoxicity after oxidation by CYP3A4/5 enzymes. Aim of this study was to investigate if expression of CYP3A4, the most abundant CYP enzyme in human liver [43] , substantially contributes to the toxicity of tetrandrine (1) and whether the para-methoxybenzyl moiety is indeed the crucial fracture point. In a combined medicinal chemistry and cell biology approach we evaluated if a reduction of the discussed CYP3A4-mediated toxicity of tetrandrine can be achieved by replacing or eliminating the hypothesized metabolically instable 12-methoxy group. This motivated us to synthesize analogues of tetrandrine which are lacking the critical methoxy group or the entire benzyl unit. Subsequently, the toxicity of the obtained tetrandrine analogues to human hepatocyte like cells (HepaRG TM ) and human hepatocellular carcinoma cells (HepG2), with or without overexpression of CYP3A4, was investigated. Concurrently, we determined the impact of structural modifications on the biological activity of compounds and focused on inhibition of cancer cell proliferation and interaction with the efflux pump P-gp, as an application in tumor therapy is desired. As mentioned above, we aimed at investigating the role of the metabolically labile 12methoxy group of tetrandrine (1). Thus we designed analogues of tetrandrine, in which this methoxy group is either deleted (target compounds RMS1-2) or replaced by metabolically stabile trifluoromethoxy (RMS3-4) or chlorine substituents (RMS7-8). Further we replaced the methoxybenzyl residue by a thienylmethyl (target compound RMS5-6) and by a non-aromatic butylidene unit (RMS9-10). The synthesis of these tetrandrine analogues is based on our recently published racemic total synthesis of tetrandrine [44] following the therein described synthetic protocol of "Route 1b". The intermediate 11 (no. 19 in ref. [44] ) of the (iso)tetrandrine synthesis served as a valuable starting material, since it already comprises one tetrahydrobenzylisoquinoline unit (rings A'-C') and ring A of the second half of the bisbenzylsisoquinoline scaffold (see Fig. 1 ). Detailed information for the preparation of intermediate 11 is provided in the Supporting Information. The second tetrahydroisoquinoline moiety (rings A and B) was constructed via trifluoromethanesulfonic acid-mediated intermolecular N-acyl Pictet-Spengler reaction of arylethylamino carbamate intermediate 11 and enol ethers 12a-12d or the aliphatic aldehyde 12e, respectively, to obtain the seco-bisbenzylisoquinolines 13a-13d and bromobutyl analogue 13e (Scheme 3). The required enol ethers 12a-d were conveniently prepared via a Wittig olefination of the corresponding commercially available aromatic aldehydes (Scheme 2), 5-bromopentanal (12e) was obtained by PCC oxidation of the corresponding primary alcohol [45] . The intermediates 13a-13e were further processed in intramolecular Ullmanntype cross-coupling reactions or an S N 2 reaction (for the ω-bromobutyl intermediate 13e) respectively to access the diaryl ether bridges (connecting rings C and C' in 14a-d) or the aliphatic bridge in 14e, furnishing macrocyclic bisbenzylisoquinoline analogues. Final step was the simultaneous reduction of both carbamate groups using lithium alanate to give the desired N-methylated compounds RMS1-6 and RMS9-10. In case of the chloroarene variations RMS7-8 this method led to substantial dechlorination, even when using the milder reduction reagent Red-Al ® (sodium bis(2-methoxyethoxy)aluminum dihydride). To circumvent this problem, the carbamate groups were first removed by treatment with methyl lithium and the resulting secondary amino groups was subsequently N-methylated via reductive amination using formaldehyde/NaBH 3 CN. Since the N-acyl Pictet-Spengler condensations expectedly proceeded with no or only rather poor diastereoselectivity [44] (with a tendency for formation of the R,R/S,S isomers), we obtained racemic mixtures of diastereomers in every case. Luckily we were able to separate the open-chain diastereomers obtained in the N-acyl Pictet-Spengler reactions by flash column chromatography to provide, in the end, each tetrandrine analogue eventually as racemic compound with the relative stereoconfiguration of either tetrandrine (1) or isotetrandrine (2) (Fig. 1 ) in high diastereomeric ratios (d.r., determined by HPLC). The relative stereochemistry of the final racemic compounds RMS1-8 was determined by analysis of NMR data utilizing the comprehensive investigation performed by Guinaudeau et al. on more than 100 bisbenzylisoquinolines [46] . Herein the chemical shifts of distinctive protons in the NMR spectra of bisbenzylisoquinolines can be used for a reliable determination of the relative configurations at both asymmetric centers. The relative configurations of all seco-precursors were then assigned retrospectively. For the variations RMS9-10, which contain an alkyl bridge instead of an aromatic ring (ring C), this method is not applicable. Unfortunately, attempts of crystallization for crystallographic analysis were unsuccessful, and the determination of the stereoconfiguration in this case was therefore not possible. To assess general toxicity, HepaRG TM cells, non-cancerous hepatic stem cells which possess various characteristics of healthy liver cells and which have a high activity of P450 enzymes [47, 48] were treated with 10 and 20 µM of tetrandrine (1) and the analogues RMS1-10. Unlike expected, none of the new compounds was less toxic to HepaRG TM cells than tetrandrine (1) at 10 µM (Fig. 3a) . In contrast, RMS4 and RMS8 slightly, but RMS2 strongly decreased cell viability. Similarly, at 20 µM, RMS2, RMS4 and RMS8 were significantly more toxic to HepaRG TM cells than tetrandrine (1) (Fig. 3b ). Of note, 20 µM of RMS6 and RMS9 were significantly less toxic than tetrandrine, whereas toxic effects of the other compounds did not substantially differ from those of tetrandrine (1) . It should be noted that depending on the substituent at C-12 of ring C, oxidation to a para-quinone methide cannot be excluded for 12-unsubstituted compounds RMS1/RMS2 (via initial CYP-mediated ring hydroxylation), whereas in trifluoromethoxy compounds RMS3/RMS4 and chloro compounds RMS7/RMS8 oxidation processes are prevented by metabolically stable substituents. To specifically determine the influence of CYP3A4 activity on toxicity in vitro, CYP3A4 with a C-terminal EGFP tag was cloned (CYP3A4-EGFP) and its physiological function was successfully validated by conversion of a proluminogenic CYP3A4 substrate in transiently transfected HepG2 cells, which was strongly reduced by the CYP3A4 inhibitor ketoconazole [49] (Suppl. Fig. 1a ). Of note, treatment of CYP3A4 overexpressing HepaRG TM cells with respective compounds did not significantly increase cell death (Fig. 3c ). As observed for the non-transfected HepaRG TM cells, RMS2 still had the strongest effect on cell viability. Next, transgenic HepG2 cells, a liver cancer cell line frequently used for recombinant expression of CYP enzymes and hepatotoxicity studies [49] [50] [51] , were generated. Stable transfection and overexpression of CYP3A4 were confirmed by PCR methods (Suppl. Fig. 1b,c) . Again, no correlation between the level of cellular CYP3A4 expression and cytotoxicity was observed. Interestingly, most tetrandrine analogues (RMS1, RMS2, RMS3, RMS4, RMS5, RMS7, RMS8) exerted generally increased cytotoxicities against cancerous HepG2 cells, mostly independent of their stereochemistry, in comparison to tetrandrine (1) . Surprisingly, the diastereomers RMS9 and RMS10 slightly, while RMS5 and RMS6 substantially differed in their cytotoxic potencies. Of note, RMS3 and RMS5 influenced cell viability of HepaRG TM cells similarly to tetrandrine (1) (Fig. 3a-c) , but they exerted strongly increased cytotoxicites to cancerous HepG2 cells (Fig. 3d ). To exclude that the observed similar cytotoxic effects on vector and CYP3A4-EGFP transfected cells were caused by insufficient initial demethylation that is required for tetrandrine (1) for being oxidized to a putatively toxic para-quinone methide, related alkaloids berbamine (7) and dauricine (6) were also tested. Both alkaloids (Fig. 2) bear a parahydroxybenzyl moiety at the region of interest, which theoretically can be directly oxidized by CYP3A4 to a para-quinone methide with no need for previous O-demethylation. Similarly, for both berbamine (7) and dauricine (6), cell death was not significantly increased by cellular CYP3A4 overexpression (Fig. 3d) . Taken together, the toxicity of tetrandrine (1) was not considerably decreased or even increased by variation of the para-methoxybenzyl moiety in several cellular models. However, no influence of the level of cellular CYP3A4 expression was found. Consequently, we conclude that the proposed CYP3A4-mediated generation of a para-quinone methide [26] does not substantially contribute to the hepatotoxicity of tetrandrine (1). To decipher the correlation between the structure of quinone methides and their liver toxicity, Thompson and coworkers [52] investigated the toxicity of a series of 4-alkyl-2methoxyphenols using an in vitro hepatotoxicity model. Although all tested molecules were converted into quinone methides, only little correlation between the rate of quinone methide formation in microsomes and relative toxicities of the alkylphenols was found [52] . It was suggested that primarily the reactivity of the quinone methides being formed and their stability towards solvolysis are the determining factors for their toxicity. These findings support the results of a former in vivo study [53] which observed differences in the toxicities of 2-methoxy-quinone methides that could be explained by their relative reactivities [54] .Thus, formation of quinone methides does not necessarily lead to toxicity in vitro and in vivo. For tetrandrine (1), other proposed mechanisms might play a more substantial role in toxicity, such as the generation of ROS by CYP2E1 [32] or the proposed interaction with p38α MAPK, a promotor of inflammatory processes in the liver [31] , or additional effects that remain to be elucidated. (1) and RMS1-RMS10 for 24 h and cell viability was determined by CellTiter-Blue ® cell viability assay. Cell viability was normalized to vehicle control. Bar graphs indicate means ± SEM of three independent experiments (One-Way ANOVA followed by Dunnett's multiple comparison test, relative cell viabilites were compared with that of tetrandrine, *P < 0.05, **P < 0.01, ***P < 0.001). (c) Differentiated HepaRG TM cells were transfected with pcDNA3-CYP3A4-EGFP or an empty vector control and treated with 10 µM of tetrandrine (1) and RMS1-RMS10 for 24 h. (d) HepG2 cells stably transfected with pcDNA3-CYP3A4-EGFP or empty vector control were treated with berbamine (7), dauricine (6), tetrandrine (1) and RMS1-RMS10 (15 µM) for 24 h. (c,d) Cell death was determined by propidium iodide staining and flow cytometry. Means ± SEM of three independent experiments are shown (unpaired t-test with Welch's correction, not significant). bbm: berbamine, dau: dauricine, Rel.: relative, tet: tetrandrine. As modification of the para-methoxybenzyl moiety of tetrandrine (1) led, depending on the kind of chemical modification and relative stereochemistry, to decreased, similar or elevated cytotoxicities to liver cells, we aimed to decipher whether and to which extent the modifications influenced known biological effects of tetrandrine on cancer cells. Firstly, their interactions with the efflux transporter P-gp [11] were investigated. Except for RMS9 and RMS10 (analogues lacking aromatic ring C), all tetrandrine analogues were able to prevent efflux of the model substrate calcein-AM from P-gp overexpressing, vincristine-resistant (VCR-R) CEM cells [55] equally to the parental molecule (Fig. 4a) . P-gp surface expression, evaluated by flow cytometry, was not diminished by any of the compounds at the same conditions (Fig. 4b) , suggesting that the observed increase in calcein fluorescence (Fig. 4a ) was caused by direct interaction with P-gp. Subsequently, the compounds were used at 1 µM in combination with varying doses of vincristine (VCR). As expected, all but RMS9 and RMS10 were able to sensitize VCR-R CEM cells to VCR, indicated by strongly increased apoptosis rates (Fig. 4c) . RMS8 was the most potent analogue as it significantly increased apoptosis at 0.01 µM VCR (Fig. 4c) . These data indicate that by replacing the paramethoxybenzyl moiety of tetrandrine with other substituted or unsubstituted aromatic residues, the potency to inhibit P-gp is maintained, while replacement with an alkyl chain (RMS9 and RMS10) strongly reduces it. Thereby, we add new information to structureactivity relationships of bisbenzylisoquinoline alkaloids with regards to their interference with the major efflux transporter P-gp. (a,c) Bar graphs indicate means ± SEM of three independent experiments (Two-Way ANOVA followed by Dunnett's multiple comparison test, *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001). (b) Bar graphs indicate means ± SEM of three independent experiments (One-Way ANOVA followed by Dunnett's multiple comparison test, *P < 0.05). -: solvent control, Rel.: relative, tet: tetrandrine, VCR: vincristine. In a next step, we determined direct antiproliferative effects of the compounds on VCR-R CEM cells, which are cross-resistant to a variety of chemotherapeutic drugs [55] . Interestingly, most tetrandrine analogues (RMS1-RMS5, RMS7-8) displayed a markedly enhanced ability to inhibit proliferation of VCR-R leukemia cells (Fig. 5a,b) . RMS6, RMS9 and RMS10 were nearly inactive. Thus, in contrast to the lead structure tetrandrine (1), RMS1-RMS5, RMS7 and RMS8 were also effective when applied as monotherapy and therefore represent potential candidates to treat multidrug resistant cancers. Notably, RMS6 had no direct effect on proliferation (Fig. 5a,b) , but successfully sensitized VCR-R CEM cells to VCR (Fig. 4c ) through inhibition of P-gp (Fig. 4a ). To gain more detailed insights into the underlying mechanisms, the influence of tetrandrine (1) and RMS1-RMS10 on pathways related to cell survival and apoptosis was investigated. Wan and coworkers [56] illustrated that tetrandrine and the multikinase inhibitor J o u r n a l P r e -p r o o f sorafenib have synergistic antitumor effects by reducing expression of anti-apoptotic proteins and activation of the kinase Akt. This prompted us to investigate how RMS1-RMS10 and tetrandrine (1) affect the intrinsic apoptosis pathway and phosphorylation of Akt, which is a key factor for proliferation and cell survial [56] . Upon treatment with 5 µM, phosphorylation of Akt and expression levels of the anti-apoptotic proteins B-cell lymphoma-extra large (Bcl-xL) and myeloid cell leukemia sequence-1 (Mcl-1) were barely affected by any of the compounds (Fig. 5c ). However, PARP cleavage, a marker for cells undergoing apoptosis [56] , was mostly detected for several RMS compounds (RMS1, RMS3, RMS4, RMS8). Mitochondrial depolarization is an indicator for early stage apoptosis [57] through the intrinsic, mitochondria-initiated pathway and mitochondrial health can be visualized with the fluorescent dye JC-1. Along this line, a shift towards green fluorescence was observed for RMS2, RMS3, RMS4, RMS5 and RMS8, indicating disruption of the mitochondrial membrane potential ΔΨm, whereas tetrandrine (1), RMS1, RMS6, RMS7, RMS9 and RMS10 had no such an effect (Fig. 5d ,e). Thus, these findings indicate that impaired mitochondrial functions partially account for the enhanced antiproliferative and cytotoxic effects of numerous tetrandrine analogues in comparison to tetrandrine (1) on VCR-R CEM cells. In HepG2 cells, p-Akt levels were reduced by RMS4 and RMS5, whereas no PARP cleavage was detected (Suppl. Fig. 2a) by any of the other tested compounds. Additionally, RMS5 slightly diminished the expression of the anti-apoptotic Bcl-2 family proteins Bcl-XL and Mcl-1. In accordance with the absence of PARP cleavage, none of the compounds significantly disrupted ΔΨm at 5 µM (Suppl. Fig. 2b ). Taken together, amendment or replacement of the 12-methoxy group of tetrandrine (1) by metabolically stable substituents can lead to enhanced induction of the intrinsic apoptosis pathway or activation of Akt, depending on the cancer cell line. Bar graphs indicate means ± SEM of at least three independent experiments (One-Way ANOVA followed by Dunnett's multiple comparison test, *P < 0.05, **P < 0.01, ***P < 0.001) The mitochondrial uncoupler CCCP (100 µM, 1 h) served as positive control. -: solvent control, Rel.: relative, tet: tetrandrine (1). Considering both the antitumoral potential and the toxicity profile, RMS3 and RMS5 have markedly enhanced antiproliferative and cytotoxic effects on cancer cells (Fig. 3d, Fig. 5 ), while their toxicity to non-malignant hepatocytes is equal in comparison with tetrandrine ( Fig. 3a ,b). Consequently, additional toxicity studies with primary cells were conducted to further elucidate their therapeutic potential. In line with the observations made with HepaRG TM cells ( Fig. 3a,b) , toxicities of RMS3 and RMS5 to human umbilical vein endothelial cells (HUVECs) and peripheral blood mononuclear cells (PBMCs) were similar to those caused by tetrandrine and only minor differences were found ( Fig. 6a,b) . While RMS3 had a slightly stronger effect on the viability of HUVECs than tetrandrine at 10 µM ( Fig. 6a) , the highest percentage of dead PBMCs was detected after tetrandrine exposure at 5 and 10 µM (Fig. 6b ). Taken together, the toxicity profile of the tetrandrine analogues RMS3 and RMS5 is largely equal to that of tetrandrine, but the improved anticancer properties theoretically enable dose reduction, providing incentive for further in vivo investigations. (1), RMS3 and RMS5 was determined by propidium iodide staining and flow cytometry. Experiment was performed in triplicate (Two-Way ANOVA followed by Dunnett's multiple comparison test, *P < 0.05, **P < 0.01, ***P < 0.001). The plant bisbenzylisoquinoline alkaloid tetrandrine (1) has diverse pharmacological properties [3, 4] , including antiviral [5] [6] [7] [8] , anticancer [9, 10] , multidrug resistance reversing [11] [12] [13] [14] [15] and calcium channel blocking [6, [16] [17] [18] activities, making this alkaloid an attractive drug candidate. Unfortunately, its clinical application is limited by its toxicity, mainly affecting liver and lungs [26] [27] [28] [29] [30] . Five diastereomeric pairs of novel analogues of tetrandrine and its natural diastereomer isotetrandrine, respectively, were synthesized and biologically evaluated with respect to their toxicity, their antiproliferative and multidrug resistance reversing activity. The design of these new analogues was driven by the published hypothesis that one particular structural element, the methoxy group at C-12 in para-position to a benzylic methylene group, is responsible for toxicity due to its susceptibility to undergo enzymatic oxidation to a reactive (toxic) para-quinone methide, which in turn can react with bio-nucleophiles. The shape of the new compounds was intended to prevent this oxidative toxification process. Although all of the analogues lack the putative problematic structure motif, no significant decrease of toxicity could be observed without a concurrent reduction of anticancer activity. Compounds RMS2, RMS4 and RMS8 were found to be even more toxic to healthy cells than tetrandrine (1). Moreover, CYP3A4 overexpression in two cellular models that are established for recombinant hepatotoxicity studies suggested that the propagated mechanism of CYP3A4-mediated toxification involving the para-quinone methide 4 (see Scheme 1) is no primary cause for the cytotoxicity of tetrandrine and related (bis)benzylisoquinoline alkaloids. Consequently, the structure variations made in our approach are not a suitable approach in order to reduce the alkaloid's toxicity, and different strategies remain to be investigated in this respect. On the contrary, further biological characterization revealed distinct differences between tetrandrine and the synthesized analogues. While P-gp inhibitory potency was maintained by most of the performed structure variations, seven compounds exerted strongly enhanced antiproliferative potential against drug-resistant leukemia cells and increased cytotoxicities to liver cancer cells as compared with tetrandrine (1). Mechanistically, those compounds acted on the intrinsic, mitochondriainitiated apoptosis pathway and/or on activation of the kinase Akt. Considering both their toxicity and the pharmacological profile, compounds RMS3 and RMS5 were found to be most promising, since they possess stronger anticancer properties, while displaying no increased toxicity to healthy liver cells. Similar toxicities to primary cells were confirmed using nonmalignant blood and endothelial cells. Side effects might therefore be reduced by lowering the required therapeutic dose and, therefore, our study strongly suggests their investigation using in vivo models. Moreover, based on these encouraging data and facilitated by our recent work on effective total synthesis of the bisbenzylisoquinoline alkaloids tetrandrine and isotetrandrine [44] , further investigations regarding structure-activity relationships in this chemical class should be part of future medicinal chemistry projects. MM and RMS developed the concept. RMS, MM and FG conducted experiments. RMS and MM wrote the paper. AMV, FB, FG and KB substantially revised the manuscript. . HUVECs were cultured for a maximum of six passages. All cells were cultured at 37 °C with 5% CO 2 with constant humidity are proven to be mycoplasma-free on a quarterly basis. The cDNA template was generated by isolation of mRNA from HepaRG TM progenitor cells (QIAGEN RNeasy Mini Kit) and subsequent reverse transcription (High Capacity cDNA Reverse Transcription Kit, Applied Biosystems, Waltham, USA). cDNA was amplified by PCR (Thermo Scientific Phusion Green Hot Start II High-Fidelity Polymerase, Thermo Fisher, Waltham, USA) (CYP3A4-FW: 5'-ATATATGGTACCGCCACCATGGCTCTCATCCCA-3', CYP3A4-RV: 5'-ATCTCGAGGGCTCCACTTACGGTGCCA-3'). The obtained PCR product was cloned into the pcDNA3-EGFP vector using FastDigest KpnI, FastDigest XhoI and T4 DNA Ligase (all purchased from Thermo Fisher) as indicated by the manufacturer. pcDNA3-EGFP was a gift from Doug Golenbock (Addgene plasmid #13031; http://n2t.net/addgene:13031; RRID:Addgene_13031). Correct insertion of the insert was confirmed by PCR, restriction digestion and Sanger sequencing. Sequencing services were provided by Eurofins Genomics (Munich, Germany). Primers were purchased from Metabion (Planegg, Germany). Metabolic activity of CYP3A4-EGFP was confirmed using a CYP3A4 P450-Glo TM assay (Promega, Madison, USA). HepG2 cells were seeded at a density of 0.75 × 10 6 cells per well into a 24 well plate and allowed to adhere overnight. On the following day, cells were transfected with either pcDNA3-CYP3A4-EGFP or pcDNA3-EGFP using the Lipofectamine TM 3000 (Invitrogen, Waltham, USA) transfection reagent according to the manufacturer's instructions. On the subsequent day, cells were incubated with luciferin-IPA (3 µM) in the presence or absence of ketoconazole (Santa Cruz biotechnology, Dallas, USA) (10 µM) for 60 min. Luminescence was detected using the nonlytic method as described by the manufacturer. Isolation of mRNA (RNeasy Mini Kit, QIAGEN, Venlo, Netherlands), synthesis of cDNA templates (High Capacity cDNA Reverse Transcription Kit, Applied Biosystems, Waltham, USA) and quantitative real-time PCR (ABI 7300 Real-Time PCR System, Applied Biosystems) with human actin serving as housekeeping gene were performed as described before [58] . Primers were purchased from Metabion. Primer sequences for detecting CYP3A4 mRNA expression were taken from the work of Nozaki and coworkers [59] (FW primer: 5'-GTATGGAAAAGTGTGGGGCT-3', RV primer: 5'-GACCATCTCCTTGAGTTTTCCA-3'). For assessing the toxicity of tetrandrine and the analogues, several cellular models and approaches were used: differentiated HepaRG TM cells without transfection, differentiated HepaRG TM cells with transient transfection, HepG2 cells with stable transfection, human umbilical vein endothelial cells (HUVECs) and peripheral blood mononuclear cells (PBMCs). Williams' medium E supplemented with GlutaMAX and HepaRG Maintenance/Metabolism Medium Supplement (Maintenance/Metabolism Working Medium) (all purchased from Life Technologies) as indicated by the manufacturer. After 24 h treatment with the respective compound concentrations, cell viability was determined by CellTiter-Blue ® cell viability assay as described by the manufacturer. HepaRG TM cells were differentiated as described above. 16 h prior to stimulation, cells were transfected with either pcDNA3-CYP3A4-EGFP or pcDNA3-EGFP using Lipofectamine TM 3000 (Invitrogen) transfection reagent according to the manufacturer's instructions. Cells were treated as indicated for 24 h. Cell death was assessed by propidium iodide (5 µg/mL in PBS; Carl Roth, Karlsruhe, Germany) staining and flow cytometry using a BD FACS Canto II (BD Biosciences, Becton Dickinson, Franklin Lakes, USA). Data were analyzed using FlowJo 7.6 (BD Biosciences). No FSC/SSC gating was performed. Determination of the percentage of PI-A positive cells was conducted as stated below. Specific cell death was calculated as follows: ℎ % = ℎ % − ℎ %. HepG2 cells were transfected with either pcDNA3-CYP3A4-EGFP or pcDNA3-EGFP using Lipofectamine TM 3000 (Invitrogen) transfection reagent according to the manufacturer's instructions. Transfected cells were constantly cultivated in the presence of 0.5 mg/mL G418 (Sigma Aldrich, St. Louis, USA) for four weeks. Presence of plasmids was confirmed by PCR (pcDNA3-forward: 5'-TACATCAATGGGCGTGGATAG-3', pcDNA3-reverse: 5'-AGGAAGGGAAGAAAGCGAAAG-3'). Primers were purchased from metabion. HepG2 cells stably expressing either CYP3A4-EGFP or pcDNA3-EGFP were seeded at a density of 0.1 × 10 6 cells per well of a 24-well plate and allowed to adhere overnight. Treatment, flow cytometry and data analysis were performed as described for HepaRG TM cells. The natural product tetrandrine was kindly donated by Prof. P. Pachaly and used as free base in all assays. Berbamine (dihydrochloride) was purchased from Sigma-Aldrich (now Merck, Darmstadt, Germany) and Dauricine (free base) from Carbosynth (Compton, Berkshire, United Kingdom). PBMCs: PBMCs were isolated from anticoagulated whole blood from healthy donors by density gradient centrifugation using Ficoll-Paque PLUS density gradient medium (GE Healthcare, Chicago, USA) as described by the manufacturer. Isolated PBMCs were cultivated in RPMI 1640 supplemented with 20% FCS and 1% penicillin/streptomycin (all purchased from PAN Biotech). 4 hours after seeding, cells were treated as indicated for 48 h. Cell death was analyzed by propidium iodide (Carl Roth, Karlsruhe, Germany; 5 µg/mL in PBS) staining and flow cytometry using a BD FACS Canto II (BD Biosciences). For evaluation, cell debris was excluded and propidium iodide positive cells were determined using FlowJo 7.6 (BD Biosciences). HUVECs: HUVECs were seeded at a density of 0.125 × 10 3 cells per well of a 96-well plate and allowed to adhere overnight. After 24 h treatment with the respective compound concentrations, cell viability was determined by CellTiter-Blue ® cell viability assay as described by the manufacturer. VCR-R CEM cells were seeded at a density of 0.02 × 10 6 cells per well of a 96-well plate, incubated for 4 h before stimulation with the indicated concentrations of compounds for 48 h. 4 h after seeding, initial metabolic activity was determined and used as zero value. 2 h before the end of stimulation time, CellTiter-Blue ® reagent (Promega) was added and fluorescence at 590 nm was detected with a Sunrise ELISA reader (Tecan, Männedorf, Switzerland). Halfmaximal inhibitory concentrations (IC 50 ) values were calculated by nonlinear regression using GraphPad Prism 8.4.0 software. The calcein-AM retention assay was performed as described previously [42] . VCR-R CEM cells were seeded at a density of 0.125 × 10 6 cells per well of a 24-well plate and incubated for 4 h. Treatment was performed with the indicated concentrations for 48 h. Apoptosis was determined by propidium iodide staining as described before [60] on a BD FACS Canto II (BD Biosciences). HepG2 were seeded at a density of 0.5 × 10 6 cells per well of a 6-well plate and allowed to adhere overnight. VCR-R CEM cells were seeded at a density of 2 × 10 6 cells per well of a 6well plate 4 h prior to treatment. Afterwards, cells were treated with DMSO, tetrandrine or the respective RMS compounds (5 µM) for 24 or 48 h as indicated. Cell lysis was performed with RIPA (radioimmunoprecipiation) buffer as described before [58] . The cationic dye JC-1 (Thermo Fisher) was used to measure mitochondrial membrane potential (ΔΨm). When ΔΨm is intact, JC-1 accumulates in mitochondria, yielding a red fluorescence. In contrast, mitochondrial depolarization is indicated by a decrease in the red/green fluorescence intensity. HepG2 cells were seeded at a density of 0.1 × 10 6 cells per well of a 24-well plate and allowed to adhere overnight. VCR-R CEM cells were seeded at a density of 0.125 × 10 6 cells per well of a 24-well plate 4 h before treatment. Subsequently, cells were treated for 24 h or 48 h with DMSO or 5 µM of the respective compounds. Cells were incubated with JC-1 (1 µg/mL) for 1 h before cells were harvested (for staining of VCR-R CEM cells, the P-gp inhibitor elacridar (5 µM; Sellekchem Chemicals, Houston, USA) was present to prevent efflux of the mitochondrial uncoupler CCCP (100 µM)), washed with PBS and resuspended in PBS for flow cytometric analysis on a BD FACS Canto II (BD Biosciences). In parallel, compensation samples were prepared using Anti-Mouse Ig, κ/Negative Control (FBS) Compensation Particles Set (BD Biosciences) and a BD PE Mouse IgG1, κ/ Isotype Control (BD Biosciences #555749) and a BD Alexa Fluor ® 488 Mouse IgG1 κ Isotype Control (BD Biosciences #557721) as described by the manufacturer. Prior to analysis of cellular samples, compensation of spectral overlap was performed on a BD FACS Canto II (BD Biosciences) according to the manufacturer's instructions. The percentage of Alexa-Fluor ® -488-A positive populations was determined using FlowJo 7.6 (BD Biosciences) All solvents and reagents were purchased from commercial suppliers and were used without further purification, unless mentioned otherwise. TLC was carried out on 0.2 mm silica gel polyester plates with a fluorescence indicator (POLYGRAM SIL G/UV254, Macherey-Nagel). NMR spectra were recorded with a 400 MHz (400 MHz for 1 H and 101 MHz for 13 C) , 500 MHz (500 MHz for 1 H and 126 MHz for 13 C) or 800 MHz Bruker Biospin Avance spectrometer (800 MHz for 1 H and 201 MHz for 13 C) . Peak assignments were based on 2D NMR experiments using standard pulse programs (COSY, HSQC/HMQC, DEPT, HMBC and NOESY). Chemical shifts were referenced to the residual solvent signal (CDCl 3 : δ H = 7.26 ppm, δ C = 77.16 ppm). For the characterization of rotamers a temperature program was employed for recording both 1D and 2D spectra. Hereby chemical shifts were referenced to the signal of tetramethylsilane in deuterated tetrachloroethane (Tcl 2 [100 °C]: δ H = 5.92 ppm, δ C = 74.0 ppm). IR spectra were recorded using a Jasco FT/IR-4100 (type A) instrument equipped with a diamond ATR unit (Jasco PRO450-S). High resolution mass spectra (HR-MS) were recorded using a Jeol Mstation 700 or JMS GCmate II Jeol instrument for electron impact ionisation (EI). Thermo Finnigan LTQ was used for electrospray ionisation (ESI). Reaction monitoring by mass spectrometry was performed by atmospheric pressure solids analysis probe (ASAP) via atmospheric-pressure chemical ionization (APCI) on an Advion expression L CMS device. Purification by flash column chromatography (FCC) was performed using Merck silica gel 60 (0.040 -0.063 mm, 230-400 mesh ASTM). For the determination of purity HPLC was performed on a HP Agilent 1100 system equipped with an Agilent 1100/1200 Diode Array Detector and an Agilent Zorbax Eclipse Plus C18-column (5.0 µm, 150 x 4.6 mm) using following method: flow 0.8 mL/min; temperature 50 °C; eluent system for compounds RMS1-10, 13c-d and 14c-e: 80% MeOH, 20% water, NaOH buffer pH 9; for compounds 13a and 14a: 50% ACN, 49.9% water, 0.1% THF; for compounds 13b and 14b: 70% ACN, 30% water). The purity of all final compounds was >95%. General procedure 1: Wittig olefination: A suspension of (methoxymethyl)triphenylphosphonium chloride (1.2 equiv.) in anhydrous THF (2 mL per mmol aromatic aldehyde) was cooled to 0 °C under nitrogen atmosphere. A solution of lithium diisopropylamide (1.4 equiv., 2.0 M solution in THF) was added dropwise and the resulting mixture stirred for 45 min. A solution of the aromatic aldehyde (1.0 equiv.) in anhydrous THF (2 mL per mmol) was added with stirring. The mixture was allowed to warm up to ambient temperature and stirred for 4 h. The reaction was then quenched with deionized water and extracted 3 x with ethyl acetate. The combined organic phases were washed with brine, dried over anhydrous Na 2 SO 4 and concentrated in vacuo to afford the crude product which was purified by column chromatography. General procedure 2: N-Acyl Pictet-Spengler condensation: A solution of carbamate (1.0 equiv.) and enol ether (1.2 -2.0 equiv.) in dichloromethane (10 mL per mmol carbamate) was cooled to 0 °C under nitrogen atmosphere. Trifluoromethanesulfonic acid (TfOH, 0.1 equiv., 0.113 mol/L in acetonitrile) was added dropwise. The reaction mixture was allowed to warm up to ambient temperature and stirred for 6 -12 h. After adding TfOH (0.1 equiv.) A saturated NaHCO 3 solution was then added for neutralization and the mixture extracted 3 x with dichloromethane. The combined organic phases were dried over anhydrous Na 2 SO 4 and concentrated in vacuo to afford the crude product which was purified by column chromatography. According to a modified procedure of Wang et al. [62] , bromoarene (1.0 -1.2 equiv.), phenol (1.0 -2.0 equiv.), CuBr·Me 2 S (1 equiv.) and Cs 2 CO 3 (3 equiv.) were placed in a pressure tube or a flask closed with a screwcap with septum inlet and sealed with PTFE tape. Anhydrous pyridine (the reaction was carried out in a concentration of 0.02 mM) was added and after 5 min of prestirring the reaction mixture was heated to 110 °C for 2-7 days under nitrogen atmosphere. The reaction mixture was concentrated in vacuo, diluted with ethyl acetate and filtered over a small plug of silica gel in order to remove the catalyst and the excess base, followed by washing with ethyl acetate. The filtrate was concentrated in vacuo to afford a brown oil as crude product which was purified by column chromatography. General procedure 4: Carbamate reduction: Lithium alanate (12 equiv.) was suspended in 1 mL anhydrous THF under nitrogen atmosphere. A solution of carbamate (1.0 equiv.) in anhydrous THF (1 mL per 0.02 mmol carbamate) was added dropwise and the resulting mixture was heated at 50 °C for 4 -12 h. The reaction mixture was cooled to 0 °C and slowly quenched with water. After alkalizing to pH 12 -14 with a 2.0 M sodium hydroxide solution, the mixture was extracted 3 x with ethyl acetate. The combined organic phases were dried over anhydrous Na 2 SO 4 and concentrated in vacuo to afford the crude product which was purified by column chromatography. Prepared from 3-Bromobenzaldehyde (1.00 g, 5.41 mmol) following General Procedure 1 (Wittig olefination). Purification was accomplished by flash column chromatography (2.5% ethyl acetate in hexanes, R f = 0.25) to give the title compound as a colourless oil (940 mg, 4.41 mmol, 82%, E,Z-isomer ratio 0.92:1, estimated by NMR integrals). NMR data of the major Z-isomer: 1 Prepared from 3-bromo-4-chlorobenzaldehyde (500 mg, 2.28 mmol) following General Procedure 1 (Wittig olefination). Purification was accomplished by flash column chromatography (2.5% ethyl acetate in hexanes, R f = 0.36) to give the title compound as a light yellow oil (490 mg, 1.98 mmol, 87%, E,Z-isomer ratio 1.08:1, estimated by NMR integrals). NMR data of the major E-isomer: 1 Pyridinium chlorochromate (503 mg, 2.33 mmol, 1.3 equiv.) was suspended in 10 mL anhydrous dichloromethane under nitrogen atmosphere. Then 5-bromo-1-pentanol (300 mg, 1.80 mmol) was added and the mixture was stirred for 6 h at ambient temperature. The volatiles were removed in vacuo and purification of the residue by flash column chromatography (25% diethyl ether in hexanes, R f = 0.43) gave the product as a colourless oil (191 mg, 1.16 mmol, 64%). 1 13 13 13 Previously separated diastereomers of bisbenzylisoquinoline 13d (110 mg, 0.133 mmol of each diastereomer) were reacted following General Procedure 3. The reactions were completed after 60 h. Purification was accomplished by flash column chromatography (25% acetone in hexanes, R f = 0.15) and the products obtained as a white solid. Purity (HPLC) = 89% (λ = 210 nm) Br 35 ClN 2 O 9 51 (s, 1H, 5-H), 6.08 (s, 1H, 8'-H), 5.28 (d, J = 8.5 Hz, 1H, 1-H), 4.98 (t, J = 6.7 Hz, 1H, 1'-H), 4.02 (q, J = 6.9 Hz, 3H, 3'-H, 'OCH 2 CH 3 ), 3.88 (s, 3H, 6'-OCH 3 ), 3.87 (ddd OCH 2 CH 3 ), 14.3 (OCH 2 CH 3 ). The resonances of C-8 and C-4a could not be identified. IR (ATR): ṽ [cm -1 ] = 3355 Br 35 ClN 2 O 9 (ethoxycarbonyl)-6-methoxy-1,2,3,4-tetrahydroisoquinolin-7-yl)oxy)-N-ethoxycarbonyl-6 12 (s, 1H, 1-H), 5.02 (t, J = 6.5 Hz, 1H, 1'-H), 4.14 -3.94 (m, 6H, 3-H, 3'-H, 'OCH 2 CH 3 , OCH 2 CH 3 ), 3.86 (s, 3H, 6'-OCH 3 ), 3.82 (s, 3H, 6-OCH 3 ), 3.58 (s, 3H, 7-OCH 3 ), 3.27 (t, J = 6 Precursor of RMS9: yield: 85.5 mg, 0.113 mmol, 23%. mp: 200.5°C. 1 H NMR C-10', C-14'), 128.2 (C-8a'), 122.2 (C-11' or C-13' or C-13, C-14), 121.7 (C-11' or C-13' or C-13), 120.5 (C-8a) 13 (dd, J = 8.2, 2.5 Hz, 1H, 13'-H or 11'-H), 6.70 (d, J = 5.8 Hz, 1H, 13'-H or 11'-H), 6.60 (s, 1H, 5'-H), 6.41 (d, J = 1.7 Hz, 1H, 12-H), 6.34 (s, 1H, 5-H), 6.28 (s, 1H, 10-H), 6.19 (dd, J = 8.3, 2.2 Hz, 1H, 14'-H or 10'-H), 6.01 (s, 1H, 8'-H) 1 (C-14' or C-10'), 130.3 (C-4a'), 129.8 (C-14' or C-10'), 128.6 (C-8a'), 123.5 (C-8a), 121.0 (C-13' and C-11'), 120.2 (C-8'), 118.2 (C-10) m, 3H, 3'-H, OCH 2 CH 3 ), 3.72 (s, 3H, 6-OCH 3 ), 3.58 (d, J = 6.6 Hz, 1H, α'-H), 3.55 (s, 3H, 6'-OCH 3 ), 3.40 (td, J = 11.4, 4.9 Hz, 1H, 3'-H), 3.32 (d, J = 15.0 Hz, 1H, α-H), 3.27 -3.13 (m, 2H, 3-H, 4'-H), 3.12 (s, 3H, 7-OCH 3 ), 2.86 -2.70 (m, 3H, 4-H, 4'-H, α'-H), 2.68 -2.54 (m, 2H, 4-H, α-H), 1.33 (t, J = 7.0 Hz, 3H, 'OCH 2 CH 3 ), 1.09 (br s, 3H, OCH 2 CH 3 ). 13 C NMR, HSQC, HMBC (101 MHz, Tcl 2 , 100 °C) δ [ppm] = 156.8 (C-12'), 155.8 (C=O), 152.6 (C-6 C-13' and C-11'), 120.2 (C-8a and C-12), 119.6 (C-8'), 117.3 (C-10) HRMS (ESI) ClN 2 O 9 ethoxycarbonyl) ring C-propylidene analogues of bisnortetrandrine and -isotetrandrine (14e 86 (br s, 1H, 13'-H or 11'-H), 6.65 (br s, 1H, 13'-H or 11'-H), 6.60 (s, 1H, 5'-H), 6.28 (s, 1H, 5-H), 6.20 (br s, 1H, 14'-H or 10'-H), 5.66 (s, 1H, 8'-H) (m, 1H, 3'-H), 3.72 (s, 3H, 6-OCH 3 ), 3.58 (s, 3H, 6'-OCH 3 ), 3.55 -3.46 (m, 1H, 3'-H), 3.43 (dd, J = 12.7, 6.0 Hz, 1H, α'-H), 3.30 -3.20 (m 53 (s, 1H, 5'-H), 6.45 (br s, 1H, 10'-H or 14 4 (C-10' or C-14'), 130.5 (C-10' or C-14'), 127.8 (C-8a'), 123.3 (C-14), 122.5 (C-13), 121.8 (C-11' or C-13'), 121.5 (C-11' or C-13'), 120.4 (C-8a) 2 mg, 0.0298 mmol, 74%). mp: 174.0 -176.0 °C. 1 H NMR, COSY (400 MHz 18 (s, 3H, 7-OCH 3 ), 3.00 -2.85 (m, 4H, 3-H, 3'-H, 4-H, 4'-H), 2.83 (d, J = 11.7 Hz, 1H, α'-H), 2.79 -2.70 (m, 2H, α-H, 4'-H), 2.63 (s, 3H, 2'-NCH 3 ), 2.54 (d, J = 13.6 Hz, 1H, α-H), 2.41 (dd, J = 11.3, 5.2 Hz, 1H, 4-H), 2.34 (s, 3H, 2-NCH 3 ) 2833, found: 677.2839. 5.2.20. (±)-Ring C thiophene analogues of tetrandrine and -isotetrandrine 1'R) isomers of carbamate 14c (10.0 mg, 0beige solid (5.2 mg, 0.00868 mmol, 62%) 64 (dd, J = 8.3, 2.6 Hz, 1H, 13'-H or 11'-H), 6.48 (s, 1H, 5'-H), 6.48 -6.43 (m, 1H, 14'-H or 10'-H), 6.35 (d, J = 1.7 Hz, 1H, 12-H), 6.26 (s, 1H, 5-H), 6.02 (s, 2H, 10-H, 8'-H), 3.94 (d, J = 7.6 Hz 1 (C-14' or C-10'), 130.1 (C-14' or C-10'), 129.4 (C-4a), 129.1 (C-4a'), 127.5 (C-8a'), 121.9 (C-13' or C-11'), 121.0 (C-13' or C-11') 90 (s, 1H, 8'-H), 3.93 (d, J = 7.2 Hz, 1H, 1-H), 3.79 (dd, J = 11.2, 5.5 Hz, 1H, 1'-H), 3.74 (s, 3H, 6-OCH 3 ), 3.52 -3.43 (m, 1H, 3'-H), 3.36 (s, 3H, 6'-OCH 3 ), 3.31 -3.21 (m CDCl 3 ): δ [ppm] = 7.30 (d, J = 6.3 Hz, 1H, 14'-H or 10'-H), 7.25 -7.22 (m, 1H, 13-H), 7.09 (d, J = 7.0 Hz, 1H, 13'-H or 11'-H), 6.79 (d, J = 8.2 Hz, 1H, 14-H), 6.62 (s, 1H, 13'-H or 11'-H), 6.54 (s, 2H, 10-H, 5'-H), 6.44 (s, 1H, 14'-H or 10 C-14' or C-10'), 130.5 (C-14' or C-10'), 129.5 (C-13), 129.0 (C-4a'), 127.6 (C-8a'), 124.1 (C-14), 122.1 (C-13' and C-11'), 120.6 (C-8a) 1H, 13'-H or 11'-H), 6.90 (dd, J = 8.1, 1.9 Hz, 1H, 14-H), 6.76 (dd, J = 8.3, 2.6 Hz, 1H, 13'-H or 11'-H), 6.59 (d, J = 1.8 Hz, 1H, 10-H), 6.51 (s, 1H, 5'-H), 6.30 (s, 1H, 5-H), 6.30 (dd, J = 8.6, 2.1 Hz, 2H, 14'-H or 10'-H), 5.98 (s, 1H, 8'-H), 3.88 (dd, J = 10.9, 5.6 Hz, 1H, 1'-H), 3.75 (s, 3H, 6-OCH 3 ), 3.71 (d, J = 9 51 (s, 3H, 2'-NCH 3 ), 2.45 (s, 3H, 2-NCH 3 ), 2.44 -2.38 (m, 1H, 4-H), 1.78 -1.68 (m, 2H, 3''-H, 2''-H), 1.66 -1.53 (m, 2H, 3''-H, 1''-H), 1.48 -1.36 (m, 2H, 2''-H, 1''-H). 13 C NMR, DEPT, HMQC, HMBC (126 MHz HRMS (ESI): m/z calcd for Alkaloids of Sinomenium and Cocculus. XIX. Alkaloids of Stephania terandra S. Moore Die Konstitution des Tetrandrins. Die Alkaloide von Stephania tetrandra Tetrandrine -A molecule of wide bioactivity Herbal alkaloid tetrandrine: from an ion channel blocker to inhibitor of tumor proliferation Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV Two-pore channels control Ebola virus host cell entry and are drug targets for disease treatment NAADP-dependent Ca2+ signaling regulates Middle East respiratory syndrome-coronavirus pseudovirus translocation through the endolysosomal system Bis-benzyltetrahydroisoquinoline derivatives as therapeutics for filovirus Tetrandrine and cancer -An overview on the molecular approach Tetrandrine, a Chinese plant-derived alkaloid, is a potential candidate for cancer chemotherapy Tetrandrine Interaction with ABCB1 Reverses Multidrug Resistance in Cancer Cells Through Competition with Anti-Cancer Drugs Followed by Downregulation of ABCB1 Expression The bis benzylisoquinoline alkaloids, tetrandine and fangchinoline, enhance the cytotoxicity of multidrug resistance-related drugs via modulation of P-glycoprotein Tetrandrine enhances cytotoxicity of cisplatin in human drug-resistant esophageal squamous carcinoma cells by inhibition of multidrug resistance-associated protein 1 Characterization of tetrandrine, a potent inhibitor of P-glycoprotein-mediated multidrug resistance Tetrandrine and fangchinoline, bisbenzylisoquinoline alkaloids from Stephania tetrandra can reverse multidrug resistance by inhibiting P-glycoprotein activity in multidrug resistant human cancer cells Studies on the Calcium Antagonistic Action of Tetrandrine: III. Effect of Tetrandrine on Positive Inotropic Action of Isoproterenol and Ca ++ and on Excitation-Contraction Coupling in Isolated Cat Papillary Muscles Interaction of tetrandrine with slowly inactivating calcium channels. Characterization of calcium channel modulation by an alkaloid of Chinese medicinal herb origin Tetrandrine Inhibits Both T and L Calcium Channel Currents in Ventricular Cells The isoquinoline alkaloids: chemistry and pharmacology Tetrandrine Tablets Used in the Treatment of COVID-19 (TT-NPC) Tetrandrine-induced cell cycle arrest and apoptosis in A549 human lung carcinoma cells Inhibition of proliferation and induction of apoptosis by tetrandrine in HepG2 cells Two-Pore Channel Function Is Crucial for the Migration of Invasive Cancer Cells Molecular mechanisms of drug resistance Metabolic Activation and Toxicities of bis-Benzylisoquinoline Alkaloids Pulmonary Toxicity and Metabolic Activation of Tetrandrine in CD-1 Mice CYP3A5 mediates bioactivation and cytotoxicity of tetrandrine Studies of the chronic toxicity of tetrandrine in dogs: An inhibitor of silicosis Acute and sub-chronic toxicity of tetrandrine in intravenously exposed female BALB/c mice Tetrandrine-induced apoptosis in rat primary hepatocytes is initiated from mitochondria: Caspases and Endonuclease G (Endo G) pathway Molecular mechanisms involved in drug-induced liver injury caused by urate-lowering Chinese herbs: A network pharmacology study and biology experiments ROS generated by CYP450, especially CYP2E1, mediate mitochondrial dysfunction induced by tetrandrine in rat hepatocytes Pulmonary toxicity and metabolic activation of dauricine in CD-1 mice Identification of Quinone Methide Metabolites of Dauricine in Human Liver Microsomes and in Rat Bile Antioxidative and antiapoptosis: Neuroprotective effects of dauricine in Alzheimer's disease models Dauricine induces apoptosis, inhibits proliferation and invasion through inhibiting NF-κB signaling pathway in colon cancer cells In vitro and in vivo metabolic activation of berbamine to quinone methide intermediate Berbamine Inhibits the Growth of Liver Cancer Cells and Cancer-Initiating Cells by Targeting Ca 2+ /Calmodulin-Dependent Protein Kinase II Berbamine inhibited the growth of prostate cancer cells in vivo and in vitro via triggering intrinsic pathway of apoptosis Cepharanthine: An update of its mode of action, pharmacological properties and medical applications Isoquinoline Alkaloids from Berberis vulgaris as Potential Lead Compounds for the Treatment of Alzheimer's Disease Racemic total synthesis and evaluation of the biological activities of the isoquinoline-benzylisoquinoline alkaloid muraricine Cytochrome P-450 3A4: regulation and role in drug metabolism A modular approach to the bisbenzylisoquinoline alkaloids tetrandrine and isotetrandrine Synthesis of Hydroxy-α-sanshool Spectral characteristics of the bisbenzylisoquinoline alkaloids Long-term functional stability of human HepaRG hepatocytes and use for chronic toxicity and genotoxicity studies Automated detection of hepatotoxic compounds in human hepatocytes using HepaRG cells and image-based analysis of mitochondrial dysfunction with JC-1 dye Establishment of the transformants expressing human cytochrome P450 subtypes in HepG2, and their applications on drug metabolism and toxicology In vitro micronucleus test in HepG2 transformants expressing a series of human cytochrome P450 isoforms with chemicals requiring metabolic activation Advantages of human hepatocyte-derived transformants expressing a series of human cytochrome p450 isoforms for genotoxicity examination o-Methoxy-4-alkylphenols That Form Quinone Methides of Intermediate Reactivity Are the Most Toxic in Rat Liver Slices Hepatotoxicity of eugenol and related compounds in mice depleted of glutathione: structural requirements for toxic potency The influence of 4-alkyl substituents on the formation and reactivity of 2-methoxy-quinone methides: evidence that extended πconjugation dramatically stabilizes the quinone methide formed from eugenol Atypical multidrug resistance in a therapy-induced drug-resistant human leukemia cell line (LALW-2): resistance to Vinca alkaloids independent of P-glycoprotein Synergistic antitumour activity of sorafenib in combination with tetrandrine is mediated by reactive oxygen species (ROS)/Akt signaling Mitochondrial membrane potential Inhibition of Cyclin-dependent Kinase 5 -A Strategy to Improve Sorafenib Response in Hepatocellular Carcinoma Therapy RNA Editing Enzymes Modulate the Expression of Hepatic CYP2B6, CYP2C8, and Other Cytochrome Analysis of apoptosis by propidium iodide staining and flow cytometry Comparison of Stain-Free gels with traditional immunoblot loading control methodology Total Synthesis of (−)-Melanthioidine by Copper-Mediated Cyclodimerization This work was funded by the German Research Foundation (DFG VO 376/19-1 and BR 1034/7-1). We thank Anna Niedrig for performing the HPLC measurements as well as Bernadette Grohs and Julian Frädrich for technical assistance to conduct cell-based experiments. Supplementary data related to this article can be found at xxx