Next Article in Journal
Promising Fungicides from Allelochemicals: Synthesis of Umbelliferone Derivatives and Their Structure–Activity Relationships
Next Article in Special Issue
Synthesis of Novel Pyrazole Derivatives and Their Tumor Cell Growth Inhibitory Activity
Previous Article in Journal
Differential Effects of the Flavonolignans Silybin, Silychristin and 2,3-Dehydrosilybin on Mesocestoides vogae Larvae (Cestoda) under Hypoxic and Aerobic In Vitro Conditions
Previous Article in Special Issue
Design, Synthesis and Biological Evaluation of Novel 4-Substituted Coumarin Derivatives as Antitumor Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Evaluation of Anticancer Activities of Novel C-28 Guanidine-Functionalized Triterpene Acid Derivatives

Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russia
*
Authors to whom correspondence should be addressed.
Molecules 2018, 23(11), 3000; https://doi.org/10.3390/molecules23113000
Submission received: 8 October 2018 / Revised: 5 November 2018 / Accepted: 13 November 2018 / Published: 16 November 2018
(This article belongs to the Special Issue Design and Synthesis of Organic Molecules as Antineoplastic Agents)

Abstract

:
Triterpene acids, namely, 20,29-dihydrobetulinic acid (BA), ursolic acid (UA) and oleanolic acid (OA) were converted into C-28-amino-functionalized triterpenoids 47, 8a, 15, 18 and 20. These compounds served as precursors for the synthesis of novel guanidine-functionalized triterpene acid derivatives 9b12b, 15c, 18c and 20c. The influence of the guanidine group on the antitumor properties of triterpenoids was investigated. The cytotoxicity was tested on five human tumor cell lines (Jurkat, K562, U937, HEK, and Hela), and compared with the tests on normal human fibroblasts. The antitumor activities of the most tested guanidine derivatives was lower, than that of corresponding amines, but triterpenoids with the guanidine group were less toxic towards human fibroblasts. The introduction of the tris(hydroxymethyl)aminomethane moiety into the molecules of triterpene acids markedly enhanced the cytotoxic activity of the resulting conjugates 15, 15c, 18b,c and 20b,c irrespective of the triterpene skeleton type. The dihydrobetulinic acid amine 15, its guanidinium derivative 15c and guanidinium derivatives of ursolic and oleanolic acids 18c and 20c were selected for extended biological investigations in Jurkat cells, which demonstrated that the antitumor activity of these compounds is mediated by induction of cell cycle arrest at the S-phase and apoptosis.

Graphical Abstract

1. Introduction

Among natural products of plant origin that are considered as abundant sources of lead structures for the discovery of new drugs, the pentacyclic lupane, ursane, and oleanane triterpenoids occupy a prominent place [1,2]. Triterpene acids (betulinic, ursolic, and oleanolic acids, Figure 1) are of interest for pharmacological research, as they exhibit a variety of biological activities including antimicrobial, antiparasitic, antitumor, and antiviral, in particular, anti-HIV, types of activity [3,4,5,6]. Among these properties of triterpenoids, of special interest is their anticancer activity and the ability to trigger the mitochondrial apoptosis pathway in various types of human cancer cells [5,6,7,8,9]. Thus, betulinic acid is capable of inducing apoptosis in tumor cells, such as melanoma, adenocarcinoma, neuroblastoma, medulloblastoma, glioblastoma and neuroectodermal tumors [7,8,9]. The in vivo anticancer activity of betulinic acid was identified using xenograft models [10,11]. The ursolic acid can also induce apoptosis, autophagy, and cell cycle arrest through various pathways, such as inhibition of DNA replication, stimulation of reactive oxygen species (ROS) production, and affecting the balance between proapoptotic and antiapoptotic proteins [6,12,13].
The useful pharmacological properties of triterpene acids are successfully combined with their acceptable systemic toxicity towards animals. However, the relatively low anticancer potential and high hydrophobicity of these secondary metabolites markedly hamper their advancement as anticancer drug candidates. For this reason, active search is in progress for analogues of natural triterpenoids with a higher biological potential and enhanced pharmacological characteristics (hydrophilicity, bioavailability) [8,14,15]. It has been shown [16,17,18,19,20,21,22,23,24,25] that conversion of triterpene compounds to cationic derivatives such as quaternary ammonium [16,17], pyridinium [18,19] or triphenylphosphonium salts [20,21,22,23,24,25] may serve as an efficient approach to improving bioavailability and selectivity of their biological action. Our recent study has shown that triphenylphosphonium derivatives of betulinic and ursolic acids are substantially superior over their prototypes in the antitumor activity and in the triggering mitochondria-dependent apoptosis of cancer cells [24,25].
However, the cytotoxic activity of the phosphonium salts was comparable with their cytotoxic activity against normal peripheral blood cells. In continuation of the search for efficient and selective antitumor agents, we have investigated novel cationic derivatives of pentacyclic triterpenoids containing guanidine groups, which are readily protonated at a physiological pH level. The introduction of hydrophilic guanidine groups into hydrophobic triterpene acid molecules may enhance their transmembrane transport and physicochemical characteristics. Meanwhile, the new hybrid molecules may preserve the selectivity of cytotoxic action against normal cells inherent in the natural triterpene acids. The guanidine group is a common key unit in various natural and synthetic compounds demonstrating antimicrobial, antiviral, and antitumor activities [26]. High symmetry of the Y-shaped guanidinium group promotes the formation of two parallel hydrogen bonds with the biologically relevant counterparts. Unlike ammonium groups, in which the charge is localized on one nitrogen atom (hard cations), guanidinium groups with a delocalized charge actively interact through hydrogen bonds with soft ions such as phosphates and sulfates. This feature of the guanidinium cation induce the efficient transport of biologically active substances through liposomal and cell membranes [27,28,29]. Furthermore, because of high basicity (pKa 13.5), the guanidinium group is important for selective delivery of cytotoxic molecules to tumor cells. Guanidine derivatives can be accumulated in the mitochondria of tumor cells, thus destroying the mitochondrial potential and inhibiting the mitochondrial respiratory chain [29,30].
Polyamines, which are precursors of aminoalkylguanidines, are also used to develop chemotherapeutic agents, including antibacterial and antitumor compounds [31,32]. Structurally, polyamine molecules contain positively charged nitrogen atoms at physiological pH value and can serve as electrostatic bridges between negatively charged phosphates. They are able to bind to negatively charged DNA macromolecules. However, some of physiological diamines, polyamines, and their synthetic analogues have exhibited high toxicity toward normal cells. A large body of data has now been accumulated on the biological activity of polyaminosterols, among which squalamine, trodusquemine, and their synthetic analogues are best known [33,34,35,36]. The synthesis and biological properties of polyamino triterpene acids are described in several publications [6,37,38,39,40]; the effect of introduction of the guanidine group into triterpenoid molecules has not been studied so far. Here we describe the synthesis and comparative evaluation of the cytotoxic and apoptosis-inducing activities of new guanidine derivatives of pentacyclic lupane, ursane, and oleanane triterpenoids and their precursors—C-28 conjugates of triterpene acids with some linear and branched mono-, di-, and triaminoalkanes.

2. Results and Discussion

2.1. Chemistry

While synthesizing the target compounds, we found that the Boc-deprotection of guanidine derivatives of betulinic and betulonic acids in acid medium (50% TFA in CH2Cl2) is complicated by skeletal rearrangements of the lupane skeleton. It is known from the literature [41,42] that hydrogenation of the C-20 double bond of lupane triterpenoids does not considerably affect their cytotoxic activity and selectivity between normal and tumor cell lines; therefore, in the subsequent experiments, we used 20,29-dihydrobetulinic and 20,29-dihydrobetulonic acids 1 and 2 to prepare target compounds 9b12b and 14 (Scheme 1).
Acetate 3 obtained upon hydroxyl group protection in dihydrobetulinic acid 2, and dihydrobetulonic acid 1 were converted to C-28 amide derivatives 47 and 8a via relatively unstable acid chlorides (Scheme 1).
The reactions were carried out with 1,2-diaminoethane, 1,4-diaminobutane (putrescine), tris(2-aminoethyl)amine, and N-tert-butyloxycarbonyl-1,4-bis(3-aminopropyl)piperazine. The use of branched triamine resulted in two guanidine functions being introduced into the triterpenoid molecule. Amides 47 were synthesized using a 3-fold molar excess of amines over triterpenes in order to avoid the formation of dimeric products. Compound 8a was formed in a good yield only with the use of N-Boc bis-aminopropylpiperazine, which was synthesized as described in reference [43]. The N-Boc protection was then removed by treatment with 10% trifluoroacetic acid in CH2Cl2. Guanylation of amines 47 and 8a was carried out according to the typical procedure [44] by using commercially available N,N′-di-Boc-N″-triflylguanidine. The expected compounds 913 were obtained in 60–88% yields after column chromatography on SiO2. The subsequent treatment of Boc derivatives 913 with CF3COOH and then with HCl/MeOH gave hydrochlorides 9b13b. Dihydrobetulinic acid guanidinium hydrochloride 14 was prepared by saponification of the 3-OAc function in conjugate 11b. A well studied and promising betulinic acid derivative is 2-amino-3-hydroxy-2-(hydroxymethyl)propyl 3-O-acetyl betulinate, known as the anticancer agent NVX-207 [45,46,47,48]. We have synthesized its C-20,29 hydrogenated derivative 15. Ursolic and oleanolic acetates 16 and 17 were converted into NVX-207 analogues 18 and 20 with ursane and oleanane skeletons (Scheme 2).
The reactions under reported conditions [45] afforded the desired esters 15, 18, and 20 containing the terminal NH2 group in the branched C-28 chain and amides 19 and 21 in 15–23% and 24–32% yields, respectively. Guanylation of the free amino groups in the resulting amines 15, 18, and 20 on treatment with N,N′-di-Boc-N″-triflylguanidine gave rise to guanidine derivatives 15a, 18a, and 20a in 63–79% yields. The Boc-deprotection with 50% TFA/CH2Cl2 and the subsequent treatment of salts 15b, 18b, and 20b with 5M HCl in MeOH yielded hydrochlorides 15c, 18c, and 20c.
The structures of all products were confirmed by 1D (1H, 13C, 19F) and 2D (HSQC, HMBC, COSY) NMR experiments and MALDI TOF MS. The 13C-NMR spectra of compounds 47, 8a, 15, 18, and 20 show signals for amide carbon atoms at 176.3–179.3 ppm, while the amide group protons resonate at 5.86–6.79 ppm in the 1H-NMR spectra. In the spectra of guanidine derivatives 913, 15a, 18a, and 20a, the NH-C=N and NH-Boc proton signals occurred at 8.30–9.06 ppm and 11.47–11.53 ppm, respectively. The signals for the guanidine group carbon atoms were observed at 161.8–163.6 ppm.

2.2. Biological Evaluation

The in vitro cytotoxic activity of triterpene acids (dihydrobetulinic, ursolic and oleanolic acids), twelve guanidinium salts, and some of their precursors, primary amines 46, 8a, and 15, was evaluated on five human tumor cell lines: Jurkat (T-lymphoblastic leukemia), K562 (chronic myeloid leukemia), U937 (histiocytic lymphoma), HEK 293 (embryonic kidney), and HeLa (cervical cancer). The possible cell toxicity was assessed against normal human fibroblasts. Most of the tested compounds showed moderate or significant activity against Jurkat, K562, and U937 cells as compared to triterpenoic acids (Table 1).
Amino derivatives 46 and 8a showed cytotoxic activity against all tumor cell cultures with IC50 values of 1.3–8 µM. However, these compounds were also cytotoxic against fibroblasts. Contrary to our expectations, guanylation of the terminal amino groups of lupane triterpenoids with C-28 linear aminoalkane chains (compounds 9b11b, 13a, and 14) or the branched tris-aminoethyl moiety (compounds 12a,b) did not considerably enhance the cytotoxic action. Of the listed guanidinium salts, only compounds 9b, 11b, and 14 showed an approximately 3-fold increase in the cytotoxic activity against Jurkat cells in comparison with dihydrobetulinic acid 2 and also showed selectivity towards Jurkat cells with an SI of 8.5, 7.3, and 5.6 respectively (SI = IC50 fibroblasts/IC50 Jurkat cells). The introduction of the tris(hydroxymethyl)aminomethane moiety into the molecules of triterpene acids 3, 16 and 17 markedly enhanced the cytotoxic activities for the resulting conjugates 15, 15c, 18b,c, and 20b,c, irrespective of the triterpene skeleton type. The C-28 esters of dihydrobetulinic, ursolic, and oleanolic acids with amino and guanidine groups in the ester side chain had from moderate to good activity against Jurkat and K562 cell lines. For example, the IC50 values of compounds 15c and 18c were 3.1 and 3.8 µM for T-lymphoblastic leukemia cells and 2.3 and 11.0 µM for chronic myeloid leukemia cells. The most pronounced differences in the antitumor activity were found for oleanolic acid and its conjugates 20b,c. Indeed, the IC50 values of oleanolic acid, 20b, and 20c for Jurkat cells were 271, 6.7, and 7.6 µM, respectively. It is worth noting that amine 15, its guanidine derivative 15c, and guanidinium salts of ursolic and oleanolic acids 18b,c and 20b,c showed acceptable selectivity towards Jurkat tumor cell with an SI from 6.9 to 13.4.
The identified lead compounds with the highest cytotoxicity characteristics, 15, 15c, 18c, and 20c, were further evaluated for the possible apoptosis induction in tumor cell cultures. The measurements were done by flow cytometry (Figure 2).
The highest percentage of late apoptosis (91.7%) was detected upon the treatment of Jurkat cells with the test compound 15 at IC50 concentration exposure for 48 h as depicted in Figure 2E. Compounds 15c, 18c and 20c also showed apoptotic mode of cell death on Jurkat cells line, but, in this case, the apoptotic effect of these guanidine derivatives was notably weaker. Thus, after treatment of Jurkat cells with compound 15c at IC50 concentration (4 μM) the number of vital cells is decreased from 96.2% (control) to 54.5%. Total number of apoptotic cells population constituted 23.7% (7.2% and 16.5% of early and late apoptotic cells, respectively) and number of necrotic cells was 21.7%. Comparable results were obtained with the guanidine derivative of ursolic acid 18c (6.8, 14.3, and 20.7% of early, late apoptotic cells and necrotic cells, respectively). Treatment of the Jurkat cells with 20c resulted in about 15.5% apoptotic cells and 6.1% necrotic cells, with 78.4% of the cells still being considered vital (Figure 2K). Next, we analyzed the ability of dihydrobetulinic acid to stimulate apoptosis. Our results showed that dihydrobetulinic acid triggers apoptosis in Jurkat cells at higher doses as compared to derivatives 15, 15c, and 18c. The number of apoptotic cells on treatment with dihydrobetulinic acid (59 μM) for 48 h constituted around 24% (4.0% of early-stage and 20.5% of secondary necrotic/late-stage apoptotic), while the number of vital cells was 73.1% (Figure 2C).
In summary, our results indicate that the apoptosis is induced in Jurkat cells by all test compounds. However, we observed a higher rate of necrosis after 15c, 18c and 20c incubation compared to derivative 15.
DNA flow cytometry was also used to analyze the cell cycle kinetics in Jurkat cells pre-incubated with dihydrobetulinic acid and derivatives 15, 15c, 18c, or 20c at their IC50 concentration for 24 and 48 h (Figure 3).
The distribution of DNA content in Jurkat cells reveals whether cell proliferation is arrested at one checkpoint. The results of all experiments showed in significant S-phase arrest in cells after treating with these compounds. Thus, the ratio of cells in the S phase increased from 38.3% (control) to 72.7% in cells treated with 15 (4 μM for 48 h) and increased to 47.2–56.8% in cells treated with dihidrobetulinic acid, 15c, 18c, or 20c. The appropriate decrease of the number of cells in the G2/M phase was observed. For example, the treatment of Jurkat cells with 15 and 15c resulted in a decrease of cells in the G2/M phase from 14.5% (control) to 7.0 and 1.5% (Figure 3D,F). Considering these results, we assume that dihydrobetulinic acid and compounds 15, 15c, 18c, or 20c are able to trigger programmed cell death, including apoptotic mechanisms and cell cycle arrest in the S-phase.

3. Materials and Methods

3.1. Chemistry

IR spectra (thin films or solutions in CHCl3) were obtained with use of a Vertex 70v spectrometer (Bruker, Karlsruhe, Germany). 1H- and 13C-NMR spectra were recorded in CDCl3, in MeOD or in d6-DMSO with Me4Si as the internal standard on an AVANCE–500 instrument (500.13 (1H), 125.78 MHz (13C), 470.59 MHz (19F)) or on an AVANCE-400 (400.13 (1H), 100.62 MHz (13C), 376.50 MHz (19F)) (Bruker). Mass spectra of new compounds were recorded on a Bruker–Autoflex III spectrometer (MALDI TOF, positive ion mode, sinapic acid as the matrices). Optical rotation was determined on a 141 polarimeter (Perkin–Elmer, Beaconsfield, UK). Specific rotation [α]D is expressed in (deg·mL)/(g·dm)−1; the concentration of the solution c is expressed in g/100 mL. Elemental analysis was carried out on a 1106 analyzer (Carlo Erba, Milan, Italy). TLC was carried out on Sorbfil plates (Sorbpolimer, Krasnodar, Russia) in hexane–EtOAc and chloroform–methanol, spots were visualized with anisaldehyde. Silica gel L (KSKG grade, 50–160 μm) was employed for column chromatography. All reagents and solvents were of the purest grade available, and generally were used without further treatment. The starting compounds ursolic, oleanolic acids and reagents: sodium borohydride (NaBH4), acetyl chloride, 10% Pd/C, oxalyl chloride, 1,2-diaminoethane, 1,4-diaminobutane, tris(2-aminoethyl)amine, 1,4-bis(3-aminopropyl)piperazine, tris(hydroxymethyl)aminomethane, 1,3-di-Boc-2-(trifluoromethylsulfonyl)guanidine, triethylamine (Et3N), dimethylaminopyridine (DMAP), trifluoroacetic acid (TFA) were purchased from Acros Organics (Geel, Belgium). Dihydrobetulonic and dihydrobetulinic acids were obtained from betulin according to the known procedures [41]. Acetates of oleanolic, ursolic, dihydrobetulonic and dihydrobetulinic acids were synthesized according to the typical procedures. Mono-Boc-protected bis-aminopropylpiperazine and compounds 1923 were prepared by a reported methods [43,45]. NMR 1H and 13C spectra of all new compounds are in Supplementary Materials.

3.1.1. General Procedure for the Synthesis of Amines 47

Oxalyl chloride (0.13 mL, 1.5 mmol) was added with stirring to a solution of compounds 1 or 3 (0.5 mmol) in dry CH2Cl2 (5 mL) precooled to 0 °C, and stirring of the reaction mixture was continued at room temperature for 2 h. Then the solvent and excess oxalyl chloride were removed under vacuum. The amine (1.5 mmol) was dissolved in dry CH2Cl2 (2 mL) and under vigorous stirring were added Et3N (0.2 mL, 1.5 mmol) and a solution of the acid chloride of 1 or 3 (0.5 mmol) in dry CH2Cl2 (4 mL). The mixture was stirred for 24 h (monitoring by TLC). The mixture was then poured into cold H2O and extracted with CH2Cl2 (2 × 15 mL each). The organic phases washed brine and were dried over Na2SO4 and evaporated under reduced pressure. The residue was chromatographed on silica gel, using CH2Cl2/MeOH 30:1→1:1 (v/v), to obtain pure compounds 47.
N-(4-Aminobuthyl)-3-oxolupane-28-amide (4), White powder, 78% yield; mp 170–172 °C (EtOH); [ α ] D 19 : +4° (c 0.23, CH2Cl2); IR (CHCl3) νmax 1641, 1702 (C=O), 3357 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 5.86 (t, J = 5.5 Hz, 1H, CONH), 3.32–3.20 (m, 2H, H-1′), 2.76 (t, J = 6.5 Hz, 2H, H-4′), 2.55–1.14 (m, 26H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 1.08, 1.03, 0.98, 0.97, 0.94 (all s, 3H each, H-23–H-27), 0.87 (d, J = 6.5 Hz, 3H, H-29), 0.75 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 218.2 (C-3), 176.3 (C-28), 56.0 (C-17), 55.0 (C-5), 49.8 (C-9), 49.5 (C-19), 47.4 (C-4), 44.2 (C-18), 42.7 (C-14), 41.6 (C-4′), 40.7 (C-8), 39.6 (C-1), 39 (C-22), 38.8 (C-1′), 37.7 (C-13), 36.9 (C-10), 34.2 (C-2), 33.8 (C-16), 33.6 (C-7), 30.6 (C-3′), 29.9 (C-20), 29.4 (C-15), 27.2 (C-23), 27.0 (C-2’), 26.6 (C-12), 23.1 (C-29), 23.0 (C-21), 21.5 (C-24), 21.1 (C-11), 19.7 (C-6), 16.0 (C-25), 15.9 (C-26), 14.6 (C-30), 14.5 (C-27); Anal. Calcd. for C34H58N2O2: C, 77.51, H, 11.10. Found: C, 78.03, H, 11.02%. MS: m/z 527.45 [M + H]+ (calcd. for C34H58N2O2, 526.45).
3β-N-(2-Aminoethyl)-3-O-acetyl-lupane-28-amide (5), White powder, 74% yield; mp 120–122 °C (EtOH); [ α ] D 19 −8° (c 0.24, CH2Cl2); IR (CHCl3) νmax 1647, 1735 (C=O), 3367 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 6.14 (t, J = 5.5 Hz, 1H, CONH), 4.48–4.44 (m, 1H, H-3), 3.35–3.21 (m, 2H, H-1′), 2.81 (t, J = 6.0 Hz, 2H, H-2′), 2.43–0.97 (m, 25H, CH, CH2 in pentacyclic skeleton), 2.01 (s, 3H, CH3CO–), 0.94, 0.92, 0.85, 0.84, 0.83, 0.82 (all s, 3H each, H-23–H-27 and H-29), 0.77 (d, J = 10.0 Hz, 1H, H-5), 0.73 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 176.7 (C-28), 171.0 (COCH3), 80.9 (C-3), 56.2 (C-17), 55.4 (C-5), 50.3 (C-9), 49.5 (C-19), 44.3 (C-18), 42.6 (C-14), 41.8 (C-2′), 41.7 (C-1′), 40.8 (C-8), 38.7 (C-1), 38.4 (C-22), 37.8 (C-4), 37.6 (C-13), 37.1 (C-10), 34.4 (C-7), 33.6 (C-16), 29.9 (C-20), 29.5 (C-15), 27.9 (C-23), 27.0 (C-2), 23.7 (C-12), 23.1 (C-21, C-29), 21.3 (COCH3), 21.0 (C-11), 18.2 (C-6), 16.5 (C-25), 16.2 (C-24, C-26), 14.6 (C-30), 14.5 (C-27); Anal. Calcd. for C34H58N2O3: C, 75.23, H, 8.84. Found: C, 75.74, H, 8.79%. MS: m/z 543.40 [M + H]+ (calcd. for C34H58N2O3, 542.44).
3β-N-(4-Aminobuthyl)-3-O-acetyl-lupane-28-amide (6), White powder, 58% yield; mp 104–106 °C (EtOH); [ α ] D 19 −11° (c 0.69, CHCl3); IR (CHCl3) νmax 1646, 1734 (C=O), 3367 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 5.86 (t, J = 5.5 Hz, 1H, CONH), 4.48–4.45 (m, 1H, H-3), 3.30–3.17 (m, 2H, H-1′), 2.73 (t, J = 6.5 Hz, 2H, H-4′), 2.47–0.98 (m, 25H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 2.03 (s, 3H, CH3CO–), 0.95, 0.93, 0.92, 0.84, 0.83, 0.82, (all s, 3H each, H-23–H-27 and H-29), 0.78 (d, J = 10.0 Hz, 1H, H-5), 0.73 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 176.3 (C-28), 171.0 (COCH3), 80.9 (C-3), 56.0 (C-17), 55.4 (C-5), 50.3 (C-9), 49.5 (C-19), 44.3 (C-18), 42.6 (C-14), 41.6 (C-4′), 40.8 (C-8), 39.0 (C-1′), 38.7 (C-22), 38.4 (C-1), 37.8 (C-4), 37.6 (C-13), 37.1 (C-10), 34.4 (C-7), 33.6 (C-16), 30.7 (C-3′), 29.9 (C-20), 29.4 (C-15), 27.9 (C-23), 27.2 (C-2′), 27.0 (C-2), 23.7 (C-12), 23.0 (C-21, C-29), 21.3 (COCH3), 21.0 (C-11), 18.2 (C-6), 16.5 (C-25), 16.2 (C-26, C-24), 14.6 (C-30), 14.5 (C-27); Anal. Calcd. for C36H62N2O3: C, 75.74, H, 10.95. Found: C, 76.01, H, 10.89%. MS: m/z 609.43 [M + K]+ (calcd. for C36H62N2O3, 570.48).
3β-N-[2-(N,N-bis-Aminoethyl)-aminoethyl]-3-O-acetyl-lupane-28-amide (7), White powder, 69% yield; mp 108–110 °C (EtOH); [ α ] D 19 −5° (c 0.16, C2H5OH); IR (CHCl3) νmax 1648, 1735 (C=O), 3441 (NH) cm−1; 1H-NMR (400 MHz, MeOD) δ: 4.49–4.45 (m, 1H, H-3), 3.33–3.27 (m, 2H, H-1′), 2.74–2.72 (m, 4H, H-4′, H-42″), 2.60–2.58 (m, 6H, H-2′, H-3′, H-3″), 2.32–0.84 (m, 26H, CH, CH2 in pentacyclic skeleton), 2.05 (s, 3H, CH3CO–), 1.02, 1.00, 0.93, 0.90, 0.89, 0.88 (all s, 3H each, H-23–H-27 and H-29), 0.79 (d, J = 6.4 Hz, 3H, H-30); 13C-NMR (100 MHz, MeOD) δ: 179.3 (C-28), 172.8 (COCH3), 82.5 (C-3), 57.7 (C-17), 57.4 (C-5), 57.0 (C-3′, C-3″), 55.2 (C-2’), 51.9 (C-9), 51.1 (C-19), 45.5 (C-18), 43.8 (C-14), 42.2 (C-8), 40.2 (C-22), 39.9 (C-1), 39.8 (C-4′, C-4″), 39.0 (C-4), 38.8 (C-1′), 38.4 (C-13), 38.3 (C-10), 35.8 (C-7), 34.1 (C-16), 31.4 (C-20), 30.9 (C-15), 28.7 (C-23), 28.5 (C-2), 24.8 (C-12), 24.2 (C-29), 23.8 (C-21), 22.4 (COCH3), 21.4 (C-11), 19.4 (C-6), 17.2 (C-25), 17.0 (C-24, C-26), 15.3 (C-30, C-27); Anal. Calcd. for C38H68N4O3: C, 72.56, H, 10.90. Found: C, 73.04, H, 10.87%. MS: m/z 629.56 [M + H]+ (calcd. for C38H68N4O3, 628.53).
3β-N-{[3-(3-Aminopropyl)piperazinyl]propyl}-3-O-acetyl-lupane-28-amide (8a), The acid chloride of 3 was synthesized according to the general procedure for synthesis of amines 47. Then the N-tert-butoxycarbonyl-1,4-bis(3-aminopropyl)piperazine (0.45 g, 1.5 mmol) and Et3N (0.25 mL, 1.8 mmol) were added to acid chloride of 3 (0.50 g, 1 mmol) in dry CH2Cl2 (10 mL). The mixture was stirred at room temperature for 16 h (monitoring by TLC), then was poured into cold H2O and extracted with CH2Cl2 (2 × 15 mL). The organic phases washed brine, dried over Na2SO4 and evaporated under reduced pressure to obtain compound 8. Then the compound 8 was dissolved in CH2Cl2 (7 mL), acidified with TFA 10% (v/v) in CH2Cl2 (17 mL) for deprotection reaction and stirred for around 5 h. The reaction was quenched using saturated aqueous potassium carbonate solution (20 mL). The aqueous phase was extracted with CH2Cl2 (2 × 15 mL). The organic phases washed brine, dried over Na2SO4 and evaporated under reduced pressure. The residue was chromatographed on silica gel, using CH2Cl2/MeOH 50:1→5:1, to obtain pure compound 8a. White powder, 78% yield; mp 119–121 °C (EtOH); [ α ] D 21 −7.4° (c 0.5, CHCl3); IR (CHCl3) νmax 1735 (C=O), 3361 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 6.79 (t, J = 5.5 Hz, 1H, CONH), 4.47–4.44 (m, 1H, H-3), 3.31–3.25 (m, 2H, H-1′), 2.78–2.77 (m, 2H, H-1″), 2.50–2.42 (m, 12H, H-3′–H-5′, H-3″–H-5″), 2.41–0.98 (m, 25H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-2″), 2.02 (s, 3H, CH3CO–), 0.93, 0.92, 0.91, 0.83, 0.82, 0.81 (3H each, all s, H-23–H-27 and H-29), 0.77 (d, J = 10.5 Hz, 1H, H-5), 0.72 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 176.3 (C-28), 171.0 (COCH3), 80.9 (C-3), 57.8 (C-3′), 56.7 (C-3″), 55.9 (C-17), 55.4 (C-5), 53.5 (C-5′, C-5″), 53.3 (C-4′), 53.3 (C-4″), 50.3 (C-9), 49.6 (C-19), 44.1 (C-18), 42.6 (C-14), 40.9 (C-1″), 40.8 (C-8), 39.3 (C-1′), 38.8 (C-22), 38.4 (C-1), 37.8 (C-4), 37.4 (C-13), 37.1 (C-10), 34.4 (C-7), 33.7 (C-16), 29.9 (C-20), 29.8 (C-2′), 29.7 (C-15), 27.9 (C-23), 26.9 (C-2), 25.3 (C-2″), 23.7 (C-12), 23.0 (C-21, C-29), 21.3 (COCH3), 21.0 (C-11), 18.2 (C-6), 16.5 (C-25), 16.3 (C-24), 16.2 (C-26), 14.6 (C-30), 14.5 (C-27); Anal. Calcd. for C42H74N4O3: C, 73.85, H, 10.92. Found: C, 74.13, H, 10.84%. MS: m/z 683.52 [M + H]+ (calcd. for C42H74N4O3, 682.58).

3.1.2. General Procedure for the Synthesis of Compounds 15, 1821

Acid chlorides of 3, 16, 17 was synthesized according to the general procedure for synthesis of amines 47. Then the acid chlorides of 3, 16, 17 (1 mmol) was dissolved in a mixture of pyridine (4 mL), CH2Cl2 (1 mL) and DMAP (0.09 g, 0.7 mmol) was added. After complete dissolution of DMAP, a solution containing of TRIS (tris(hydroxymethyl)aminomethane) (0.24 g, 2 mmol) in pyridine (0,5 mL) was added. The mixture was stirred for 10 h at room temperature and the solvent was removed rapidly under vacuum. The residue was chromatographed on silica gel, using CH2Cl2/MeOH 30:1→1:1, to obtain pure compounds 15, 1821.
3β-[2-Amino-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-lupane-28-oate (15), White powder, 16% yield; mp 134–136 °C (EtOH); [ α ] D 21 −13.1° (c 0.51, CHCl3); IR (CHCl3) νmax 1731 (C=O), 3366 (OH), 3446 (NH) cm−1; 1H-NMR (400 MHz, CDCl3) δ: 4.49–4.45 (m, 1H, H-3), 4.09, 4.03 (both d, J = 11.6 Hz, 1H each, H-1′), 3.52 (br s, 4H, H-3′, H-4′), 2.70 (br s, 4H, NH2, OH), 2.05 (s, 3H, CH3CO–), 2.24–0.81 (m, 26H, CH, CH2 in pentacyclic skeleton), 0.95, 0.91, 0.85, 0.84, 0.83, 0.82 (all s, 3H each, H-23–H-27 and H-29), 0.76 (d, J = 6.8 Hz, 3H, H-30); 13C-NMR (100 MHz, CDCl3) δ: 177.0 (C-28), 171.1 (COCH3), 80.9 (C-3), 64.9 (C-1′), 64.2 (C-3′, C-4′), 57.3 (C-17, C-2′), 55.4 (C-5), 50.2 (C-9), 48.8 (C-19), 44.2 (C-18), 42.6 (C-14), 40.7 (C-8), 38.4 (C-22), 38.1 (C-1), 37.8 (C-4), 37.5 (C-13), 37.1 (C-10), 34.3 (C-7), 32.0 (C-16), 29.7 (C-15, C-20), 27.9 (C-23), 26.9 (C-2), 23.7 (C-12), 23.0 (C-29), 22.8 (C-21), 21.3 (COCH3), 20.9 (C-11), 18.2 (C-6), 16.5 (C-25), 16.2 (C-24), 16.1 (C-26), 14.7 (C-30), 14.6 (C-27); Anal. Calcd. for C36H61NO6: C, 71.60, H, 10.18. Found: C, 72.09, H, 10.11%. MS: m/z 604.42 [M + H]+ (calcd. for C36H61NO6, 603.45).
3β-[2-Amino-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetylurs-12-en-28-oate (18), White powder, 15% yield; mp 129–131 °C (EtOH); [ α ] D 18 +40° (c 0.74, CHCl3); IR (CHCl3) νmax 1721 (C=O), 3444 (NH), 3468 (OH) cm−1; 1H-NMR (400 MHz, CDCl3) δ: 5.25 (br s, 1H, H-12), 4.51–4.47 (m, 1H, H-3), 4.02, 3.93 (both d, J = 12 Hz, 1H each, H-1′), 3.48 (br s, 4H, H-3′, H-4′), 2.68 (br s, 4H, NH2, OH), 2.19 (d, J = 11.2 Hz, 1H, H-18), 2.05 (s, 3H, CH3CO–), 1.99–0.81 (m, 22H, CH, CH2 in pentacyclic skeleton), 1.08, 0.96, 0.94, 0.88, 0.87, 0.86, 0.74 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (100 MHz, CDCl3) δ: 178.1 (C-28), 171.1 (COCH3), 138.3 (C-13), 125.6 (C-12), 80.9 (C-3), 65.7 (C-1′), 64.4 (C-3′, C-4′), 56.6 (C-2′), 55.3 (C-5), 53.0 (C-18), 48.6 (C-17), 47.4 (C-9), 42.1 (C-14), 39.5 (C-4, C-8), 38.9 (C-19), 38.3 (C-20), 37.7 (C-1), 37.1 (C-22), 36.8 (C-10), 32.9 (C-7), 30.6 (C-21), 28.1 (C-23), 27.9 (C-15), 24.3 (C-16, C-27), 23.5 (C-2, C-11), 21.3 (COCH3), 21.1 (C-30), 18.2 (C-6), 17.2 (C-29), 17.0 (C-26), 16.7 (C-24), 15.5 (C-25); Anal. Calcd. for C36H59NO6: C, 71.84, H, 9.88. Found: C, 72.13, H, 9.83%. MS: m/z 602.40 [M + H]+ (calcd. for C36H59NO6, 601.43).
3β-N-[(1,1′,1-tris-Hidroxymethyl)methyl]-3-O-acetyl-ursolamide (19), White powder, 24% yield; mp 203–205 °C (EtOH); [ α ] D 18 +26.9° (c 0.74, CH3OH); IR (CH3OH) νmax 1734 (C=O), 2923, 2871, 2852 (OH), 3352 (NH) cm−1; 1H-NMR (500 MHz, MeOD) δ: 5.38 (br s, 1H, H-12), 4.50–4.47 (m, 1H, H-3), 3.66, 3.58 (both d, J = 10 Hz, 3H each, H-2′–H-4′), 2.12–0.88 (m, 23H, CH, CH2 in pentacyclic skeleton), 2.05 (s, 3H, CH3CO–), 1.18, 1.04, 0.99, 0.95, 0.93, 0.92, 0.91 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, CDCl3) δ: 178.5 (C-28), 170.7 (COCH3), 138.1 (C-13), 125.8 (C-12), 80.4 (C-3), 61.8 (C-1′), 60.8 (C-2′–C-4′), 55.1 (C-5), 53.6 (C-18), 48.3 (C-17), 47.4 (C-9), 42.3 (C-14), 39.5 (C-4, C-8), 38.9 (C-19), 38.4 (C-20), 37.7 (C-22, C-1), 36.8 (C-10), 33 (C-7), 30.9 (C-21), 28.3 (C-23), 27.8 (C-15), 24.5 (C-16), 23.7 (C-2), 23.4 (C-11, C-27), 21.5 (COCH3), 21.4 (C-30), 18.2 (C-6), 17.5 (C-29, C-26), 17.1 (C-24), 15.7 (C-25); Anal. Calcd. for C36H59NO6: C, 71.84, H, 9.88. Found: C, 72.09, H, 9.79%. MS: m/z 602.41 [M + H]+ (calcd. for C36H59NO6, 601.43).
3β-[2-Amino-3-hydroxy-2(hydroxymethyl)propyl]-3-O-acetylolean-12-en-28-oate (20), White powder, 23% yield; mp 141–143 °C (EtOH); [ α ] D 18 +49° (c 0.75, CHCl3); IR (CHCl3) νmax 1618, 1727 (C=O), 3447 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 5.30 (br s, 1H, H-12), 4.51–4.48 (m, 1H, H-3), 4.04, 3.99 (both d, J = 11.5 Hz, 1H each, H-1′), 3.50 (br s, 4H, H-3′, H-4′), 2.84 (d, J = 10.0 Hz, 1H, H-18), 2.68–2.60 (m, 4H, NH2, OH), 2.05 (s, 3H, CH3CO–), 2.01–0.83 (m, 22H, CH, CH2 in pentacyclic skeleton), 1.14, 0.94, 0.93, 0.92, 0.87, 0.86, 0.73 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, CDCl3) δ: 178.2 (C-28), 171.1 (COCH3), 143.7 (C-13), 122.6 (C-12), 80.9 (C-3), 65.7 (C-1′), 64.6 (C-3′, C-4′), 56.6 (C-2′), 55.3 (C-5), 47.5 (C-9), 47.2 (C-17), 45.7 (C-19), 41.8 (C-14), 41.5 (C-18), 39.3 (C-8), 38.1 (C-1), 37.7 (C-4), 36.9 (C-10), 33.8 (C-22), 33.0 (C-30), 32.8 (C-7), 32.7 (C-21), 30.7 (C-20), 28.0 (C-23), 27.6 (C-15), 25.8 (C-27), 23.6 (C-29), 23.5 (C-11), 23.4 (C-2), 23.1 (C-16), 21.3 (COCH3), 18.2 (C-6), 17.1 (C-26), 16.7 (C-24), 15.4 (C-25); Anal. Calcd. for C36H59NO6: C, 71.84, H, 9.88. Found: C, 72.04, H, 9.82%. MS: m/z 602.40 [M + H]+ (calcd. for C36H59NO6, 601.43).
3β-N-[(1,1,1-tris-Hydroxymethyl)methyl]-3-O-acetyl-oleanolamide (21), White powder, 32% yield; mp 249–251 °C (EtOH); [ α ] D 18 +33.9° (c 0.61, CH3OH); IR (CH3OH) νmax 1732(C=O), 2945, 2927 (OH), 3353 (NH) cm−1; 1H-NMR (500 MHz, d6-DMSO) δ: 6.62 (s, 1H, NH), 5.24 (br s, 1H, H-12), 4.97 (t, J = 5 Hz, 3H, –OH), 4.42–4.38 (m, 1H, H-3), 3.49–3.42 (m, 6H, H-2′–H-4′), 2.61 (d, J = 10.0 Hz, 1H, H-18), 2.00 (s, 3H, CH3CO–), 1.97–0.84 (m, 22H, CH, CH2 in pentacyclic skeleton), 1.91, 0.90, 0.88, 0.87, 0.82, 0.81, 0.78 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, d6-DMSO) δ: 178.3 (C-28), 170.6 (COCH3), 143.7 (C-13), 122.6 (C-12), 80.4 (C-3), 61.8 (C-1′), 60.8 (C-2′–C-4′), 55 (C-5), 47.4 (C-9), 46.8 (C-17), 46.6 (C-19), 42.0 (C-14), 41.9 (C-18), 39.5 (C-8), 38.2 (C-1), 37.7 (C-4), 36.9 (C-10), 34.1 (C-22), 33.3 (C-7), 33.2 (C-30), 32.7 (C-21), 30.8 (C-20), 28.2 (C-23), 27.4 (C-15), 25.8 (C-27), 23.8 (C-29), 23.7 (C-2), 23.4 (C-11, C-16), 21.4 (COCH3), 18.2 (C-6), 17.4 (C-26), 17.1 (C-24), 15.6 (C-25); Anal. Calcd. for C36H59NO6: C, 71.84, H, 9.88. Found: C, 72.11, H, 9.85%. MS: m/z 602.39 [M + H]+ (calcd. for C36H59NO6, 601.43).

3.1.3. General Procedure for the Guanilation of Amines 47, 8a, 15, 18 and 20

The amine (0.5 mmol) is added neat to a solution of 1,3-di-Boc-2-(trifluoromethyl-sulfonyl)guanidine (0.18 g, 0.45 mmol) or (0.60 g, 0.9 mmol) for amine 7 and Et3N (0.07 mL, 0.5 mmol) in CH2Cl2 (at r.t.) of compounds 47, 8a or in CHCl3 (at reflux) of compounds 15, 18, 20 (10 mL). The mixture was stirred for 2–12 h (TLC monitoring CH2Cl2/MeOH 20:1). After completion of the reaction, the mixture is diluted with CH2Cl2 and washed witH-NH4Cl, NaHCO3 and brine. After drying with sodium sulfate and filtering the solvent is removed under reduced pressure. The residue was chromatographed on silica gel, using hexane/EtOAc 10:1→1:1, to obtain pure compounds 913, 15a, 18a and 20a.
N-[4-tert-Butyloxycarbonyl buthylguanidine]-3-oxo-lupane-28-amide (9), White powder, 88% yield; mp 158–160 °C (EtOH); [ α ] D 23 +3.4° (c 0.59, CH2Cl2); IR (CHCl3) νmax 1637, 1718 (C=O), 3337 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 11.52 (s, 1H, NH in Boc), 8.30 (br s, 1H, NH–C=N), 5.70 (br s, 1H, CONH), 3.48–3.41 (m, 2H, H-2′), 3.37–3.20 (m, 2H, H-1′), 1.51, 1.48 (both s, 9H each, CH3 in Boc), 2.54–1.15 (m, 26H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 1.08, 1.03, 0.98, 0.97, 0.95 (all s, 3H each, H-23–H-27), 0.87 (d, J = 6.5 Hz, 3H, H-29), 0.75 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 218.1 (C-3), 176.3 (C-28), 163.6 (C=N), 156.1, 153.3 (CONH-Boc), 83.0, 79.2 (C in Boc), 56.0 (C-17), 55.0 (C-5), 49.8 (C-9), 49.5 (C-19), 47.3 (C-4), 44.1 (C-18), 42.6 (C-14), 40.7 (C-8), 40.5 (C-4′), 39.6 (C-1), 38.7 (C-22, C-1′), 37.6 (C-13), 36.9 (C-10), 34.1 (C-2), 33.8 (C-16), 33.6 (C-7), 29.9 (C-20), 29.4 (C-15), 28.4, 28.0 (CH3 in Boc), 27.7 (C-23), 27.0 (C-2′, C-3′), 26.6 (C-12), 23.0 (C-21, C-29), 21.5 (C-24), 21.0 (C-11), 19.6 (C-6), 16.0 (C-25), 15.9 (C-26), 14.6 (C-30), 14.5 (C-27); Anal. Calcd. for C45H76N4O6: C, 70.27, H, 9.96. Found: C, 70.62, H, 9.91%. MS: m/z 791.56 [M + Na]+ (calcd. for C45H76N4O6, 768.58).
3β-N-(2-tert-Butyloxycarbonylethylguanidine)-3-O-acetyl-lupane-28-amide (10), White powder, 86% yield; mp 176–177 °C (EtOH); [ α ] D 23 +3.8° (c 0.56, CHCl3); IR (CHCl3) νmax 1724 (C=O), 3329 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 11.53 (s, 1H, NH in Boc), 8.65 (br s, 1H, NH–C=N), 6.88 (br s, 1H, CONH), 4.50–4.47 (m, 1H, H-3), 3.69–3.54 (m, 2H, H-2′), 3.47–3.35 (m, 2H, H-1′), 2.05 (s, 3H, CH3CO–), 1.51, 1.50 (both s, 9H each, CH3 in Boc), 2.46–0.96 (m, 25H, CH, CH2 in pentacyclic skeleton), 0.94, 0.90, 0.87, 0.86, 0.85, 0.84 (all s, 3H each, H-23–H-27 and H-29), 0.79 (d, J = 9.5 Hz, 1H, H-5), 0.75 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 176.9 (C-28), 171.0 (COCH3), 163.1 (C=N), 153.5, 153.0 (CONH-Boc), 83.5 (C in Boc), 80.9 (C-3), 77.3 (C in Boc), 56.0 (C-17), 55.4 (C-5), 50.3 (C-9), 49.7 (C-19), 44.2 (C-18), 42.5 (C-14), 41.2 (C-2′), 40.7 (C-8), 39.7 (C-1′), 38.6 (C-22), 38.4 (C-1), 37.8 (C-4), 37.5 (C-13), 37.1 (C-10), 34.3 (C-7), 33.4 (C-16), 29.8 (C-20), 29.4 (C-15), 28.3, 28.0 (CH3 in Boc), 27.9 (C-23), 26.9 (C-2), 23.7 (C-12), 23.1 (C-29), 23.0 (C-21), 21.3 (COCH3), 21.0 (C-11), 18.3 (C-6), 16.5 (C-25), 16.2 (C-24), 16.1 (C-26), 14.7 (C-30), 14.6 (C-27); Anal. Calcd. for C45H76N4O7: C, 68.84, H, 9.76. Found: C, 69.23, H, 9.69%. MS: m/z 807.54 [M + Na]+ (calcd. for C45H76N4O7, 784.57).
3β-N-(4-tert-Butyloxycarbonylbutylguanidine)-3-O-acetyl-lupane-28-amide (11), White powder, 82% yield; mp 148–150 °C (EtOH); [ α ] D 17 −4° (c 0.52, CHCl3); IR (CHCl3) νmax 1640, 1720 (C=O), 3288, 3337, 3410 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 11.48 (s, 1H, NH in Boc), 8.30 (t, J = 5.0 Hz, 1H, NH–C=N), 5.73 (t, J = 5.5 Hz, 1H, CONH), 4.46–4.43 (m, 1H, H-3), 3.42–3.37 (m, 2H, H-2′), 3.31–3.16 (m, 2H, H-1′), 2.01 (s, 3H, CH3CO–), 1.47, 1.46 (both s, 9H each, CH3 in Boc), 2.45–0.94 (m, 25H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 0.91, 0.90, 0.83, 0.82, 0.81, 0.80 (all s, 3H each, H-23–H-27 and H-29), 0.75 (d, J = 9.5 Hz, 1H, H-5), 0.71 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 176.2 (C-28), 170.9 (COCH3), 163.6 (C=N), 156.1, 153.3 (CONH-Boc), 83.0 (C in Boc), 81.0 (C-3), 79.2 (C in Boc), 56.0 (C-17), 55.4 (C-5), 50.3 (C-9), 49.5 (C-19), 44.2 (C-18), 42.6 (C-14), 40.8 (C-8), 40.5 (C-4′), 38.7 (C-1′, C-22), 38.4 (C-1), 37.8 (C-4), 37.5 (C-13), 37.1 (C-10), 34.4 (C-7), 33.6 (C-16), 29.9 (C-3′), 29.4 (C-20), 28.3, 28.0 (CH3 in Boc), 27.8 (C-15), 27.3 (C-23), 26.9 (C-2′), 26.5 (C-2), 23.7 (C-12), 23.0 (C-21, C-29), 21.3 (COCH3), 21.0 (C-11), 18.2 (C-6), 16.5 (C-25), 16.2 (C-24, C-26), 14.6 (C-30), 14.5 (C-27); Anal. Calcd. for C47H80N4O7: C, 69.42, H, 9.92. Found: C, 69.74, H, 9.85%. MS: m/z 835.51 [M + Na]+ (calcd. for C47H80N4O7, 812.60).
3β-N-[2-(N,N′-bis-tert-Butyloxycarbonylethylgyanidine)-aminoethyl]-3-O-acetyl-lupane-28-amide (12), White powder, 87% yield; mp 188–190 °C (EtOH); [ α ] D 19 +1.02° (c 0.96, CHCl3); IR (CHCl3) νmax 1641, 1722 (C=O), 3335, 3442 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 11.52 (s, 2H, NH in Boc), 8.52 (br s, 2H, NH–C=N), 6.35 (t, 1H, J = 5.5 Hz, CONH), 4.49–4.46 (m, 1H, H-3), 3.53–3.43 (m, 4H, H-4′, H-4″), 3.41–3.22 (m, 2H, H-1′), 2.65–2.54 (m, 6H, H-2′, H-3′, H-3″), 2.04 (s, 3H, CH3CO–), 1.50, 1.48 (both br s, 18H each, CH3 in Boc), 2.32–0.98 (m, 25H, CH, CH2 in pentacyclic skeleton), 0.93, 0.92, 0.85, 0.84, 0.83, 0.82 (all s, 3H each, H-23–H-27 and H-29), 0.78 (d, J = 9.0 Hz, 1H, H-5), 0.73 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 176.7 (C-28), 171.0 (COCH3), 163.5 (C=N), 155.9, 153.2 (CONH-Boc), 82.9 (C in Boc), 81.0 (C-3), 79.2 (C in Boc), 55.9 (C-17), 55.5 (C-5), 54.8 (C-2′), 53.8 (C-3′, C-3″), 50.4 (C-9), 49.6 (C-19), 43.8 (C-18), 42.5 (C-14), 40.8 (C-8), 39.0 (C-4′, C-4″), 38.9 (C-22), 38.4 (C-1), 38.0 (C-13), 37.8 (C-1′), 37.2 (C-4), 37.1 (C-10), 34.4 (C-7), 33.4 (C-16), 29.8 (C-20), 29.5 (C-15), 28.3, 28.1 (CH3 in Boc), 28.0 (C-23), 27.0 (C-2), 23.7 (C-12), 23.1 (C-29), 23.0 (C-21), 21.3 (COCH3), 21.0 (C-11), 18.3 (C-6), 16.5 (C-25), 16.4 (C-24), 16.2 (C-26), 14.7 (C-30), 14.4 (C-27); Anal. Calcd. for C60H104N8O11: C, 64.72; H, 9.41. Found: C, 65.02; H, 9.34%. MS: m/z 1135.71 [M + Na]+ (calcd. for C60H104N8O11, 1112.78).
3β-N-{[3-(3-tert-Butyloxycarbonylpropylgyanidine)piperazinyl]propyl}-3-O-acetyl-lupane-28-amide (13), White powder, 60% yield; mp 131–134 °C (EtOH); [ α ] D 21 −7.9° (c 0.57, CHCl3); IR (CHCl3) νmax 1640, 1722 (C=O), 3289, 3333 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 11.50 (br s, 1H, NH in Boc), 8.53 (br s, 1H, NH–C=N), 6.86 (br s, 1H, CONH), 4.47–4.45 (m, 1H, H-3), 3.51–3.42 (m, 2H, H-1′′), 3.32–3.27 (m, 2H, H-1′), 2.48–2.41 (m, 14H, H-3′–H-5′, H-2″–H-5″), 2.06 (s, 3H, CH3CO–), 1.49 (br s, 18H, CH3 in Boc), 2.33–0.99 (m, 25H, CH, CH2 in pentacyclic skeleton, 2H, H-2′), 0.93, 0.92, 0.85, 0.84, 0.83, 0.81 (all s, 3H each, H-23–H-27 and H-29), 0.78 (d, J = 10.5 Hz, 1H, H-5), 0.73 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 176.4 (C-28), 171.0 (COCH3), 163.7 (C=N), 156.1, 153.0 (CONH-Boc), 82.8 (C in Boc), 80.9 (C-3), 79.2 (C in Boc), 56.4 (C-3′), 55.9 (C-17), 55.9 (C-3″), 55.4 (C-5), 53.1 (C-4′, C-4″, C-5′, C-5″), 50.3 (C-9), 49.6 (C-19), 44.1 (C-18), 42.6 (C-14), 40.8 (C-8), 39.8 (C-22, C-1″), 39.2 (C-1′), 38.8 (C-1), 38.4 (C-4), 37.8 (C-10), 37.4 (C-13), 34.5 (C-7), 33.7 (C-16), 29.8 (C-20), 29.4 (C-15), 28.3, 28.1 (CH3 in Boc), 27.9 (C-23), 26.9 (C-2), 26.9 (C-2′), 25.9 (C-2″), 23.7 (C-12), 23.1 (C-21), 23.0 (C-29), 21.3 (COCH3), 21.0 (C-11), 18.2 (C-6), 16.5 (C-25), 16.3 (C-24), 16.2 (C-26), 14.6 (C-30), 14.5 (C-27); Anal. Calcd. for C53H92N6O7: C, 68.79, H, 10.02. Found: C, 69.04, H, 9.96%. MS: m/z 947.58 [M + Na]+ (calcd. for C53H92N6O7, 924.70).
3β-[2-tert-Butyloxycarbonylguanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-lupane-28-oate (15a), White powder, 63% yield; mp 106–108 °C (EtOH); [ α ] D 21 −5.6° (c 0.48, CHCl3); IR (CHCl3) νmax 1655, 1714 (C=O), 3271, 3437 (NH) cm−1; 1H-NMR (400 MHz, CDCl3) δ: 11.47 (s, 1H, NH in Boc), 9.05 (s, 1H, NH–C=N), 4.49–4.45 (m, 1H, H-3), 4.25 (br s, 2H, H-1′), 3.82-3.77, 3.57–3.53 (both m, 2H each, H-3′, H-4′), 2.05 (s, 3H, CH3CO–), 1.49, 1.47 (both br s, 9H each, CH3 in Boc), 2.30–0.98 (m, 25H, CH, CH2 in pentacyclic skeleton), 0.94, 0.90, 0.88, 0.86, 0.85, 0.83 (all s, 3H each, H-23–H-27 and H-29), 0.79 (d, J = 9.6 Hz, 1H, H-5), 0.75 (d, J = 6.4 Hz, 3H, H-30); 13C-NMR (100 MHz, CDCl3) δ: 176.3 (C-28), 171.1 (COCH3), 161.8 (C=N), 155.7, 152.8 (CONH-Boc), 83.8 (C in Boc), 80.9 (C-3), 80.1 (C in Boc), 62.9 (C-1′), 62.8 (C-4′), 62.8 (C-3′), 61.9 (C-2′), 57.3 (C-17), 55.4 (C-5), 50.2 (C-9), 49.0 (C-19), 44.0 (C-18), 42.5 (C-14), 40.7 (C-8), 38.4 (C-22), 38.0 (C-1), 37.8 (C-4), 37.1 (C-13, C-10), 34.3 (C-7), 31.8 (C-16), 29.8 (C-20), 29.7 (C-15), 28.1 (CH3 in Boc), 28.0 (CH3 in Boc, C-23), 26.9 (C-2), 23.7 (C-12), 23.0 (C-29), 22.7 (C-21), 21.3 (COCH3), 20.9 (C-11), 18.2 (C-6), 16.5 (C-25), 16.2 (C-24), 15.9 (C-26), 14.7 (C-30), 14.6 (C-27); Anal. Calcd. for C47H79N3O10: C, 66.72, H, 9.41. Found: C, 67.13, H, 9.36%. MS: m/z 868.45 [M + Na]+ (calcd. for C47H79N3O10, 845.58).
3β-[2-tert-Butyloxycarbonylguanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-urs-12-en-28-oate (18a), White powder, 75% yield; mp 118–120 °C (EtOH); [ α ] D 21 +31.3° (c 0.53, CHCl3); IR (CHCl3) νmax 1653, 1729 (C=O), 3271, 3443 (NH) cm−1; 1H-NMR (500 MHz, CDCl3) δ: 11.50 (s, 1H, NH in Boc), 9.06 (s, 1H, NH–C=N), 5.26 (br s, 1H, H-12), 4.51–4.48 (m, 1H, H-3), 4.13 (br s, 2H each, H-1′), 3.82–3.76 (m, 2H, H-3′), 3.54–3.52 (m, 2H, H-4′), 2.26 (d, J = 11.0 Hz, 1H, H-18), 2.04 (s, 3H, CH3CO–), 1.99–0.81 (m, 22H, CH, CH2 in pentacyclic skeleton), 1.50, 1.47 (both br s, 9H each, CH3 in Boc), 1.07, 0.94, 0.93, 0.86, 0.85, 0.83, 0.75 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, CDCl3) δ: 177.4 (C-28), 171.0 (COCH3), 161.8 (C=N), 155.6, 152.8 (CONH-Boc), 138.1 (C-13), 125.8 (C-12), 83.7 (C in Boc), 80.9 (C-3), 79.9 (C in Boc), 63.4 (C-1′), 62.9 (C-3′), 62.6 (C-4′), 61.6 (C-2′), 55.3 (C-5), 52.8 (C-18), 48.5 (C-17), 47.5 (C-9), 42.0 (C-14), 39.5 (C-8), 39.0 (C-4), 38.7 (C-19), 38.3 (C-20), 37.7 (C-1), 36.8 (C-22), 36.5 (C-10), 32.9 (C-7), 30.6 (C-21), 28.1 (CH3 in Boc), 28.0 (C-15), 28 (C-23), 24.1 (C-16), 23.6 (C-2), 23.5 (C-27), 23.3 (C-11), 21.3 (C-30), 21.0 (COCH3), 18.2 (C-6), 17.0 (C-29), 16.9 (C-26), 16.7 (C-24), 15.5 (C-25); Anal. Calcd. for C47H77N3O10: C, 66.87, H, 9.19. Found: C, 67.27, H, 9.14%. MS: m/z 866.43 [M + Na]+ (calcd. for C47H77N3O10, 843.56).
3β-[2-tert-Butyloxycarbonylguanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-olean-12-en-28-oate (20a), White powder, 79% yield; mp 140–142 °C (EtOH); [ α ] D 19 +30.4° (c 0.56, CHCl3); IR (CHCl3) νmax 1618, 1727 (C=O), 3434 (NH) cm−1; 1H-NMR (400 MHz, CDCl3) δ: 11.49 (s, 1H, NH in Boc), 9.03 (s, 1H, NH–C=N), 5.30 (br s, 1H, H-12), 5.10 (br s, 1H, OH), 4.49–4.45 (m, 1H, H-3), 4.26, 4.15 (both d, J = 11.6 Hz, 1H each, H-1′), 3.77, 3.54 (both d, J = 12.0 Hz, J = 11.6 Hz, 2H each, H-3′, H-4′), 2.85 (d, J = 10.0 Hz, 1H, H-18), 2.03 (s, 3H, CH3CO–), 1.99–0.80 (m, 22H, CH, CH2 in pentacyclic skeleton), 1.51, 1.48 (both br s, 9H each, CH3 in Boc), 1.12, 0.91, 0.90, 0.89, 0.85, 0.84, 0.71 (3H each, all s, H-23–H-27, H-29 and H-30); 13C-NMR (100 MHz, CDCl3) δ: 177.4 (C-28), 171.0 (COCH3), 161.8 (C=N), 155.6, 152.8 (CONH-Boc), 138.1 (C-13), 125.8 (C-12), 83.7 (C in Boc), 80.9 (C-3), 79.9 (C in Boc), 63.4 (C-1′), 62.9 (C-3′), 62.6 (C-4′), 61.6 (C-2′), 55.3 (C-5), 52.8 (C-18), 48.5 (C-17), 47.5 (C-9), 42.0 (C-14), 39.5 (C-8), 39.0 (C-4), 38.7 (C-19), 38.3 (C-20), 37.7 (C-1), 36.8 (C-22), 36.5 (C-10), 32.9 (C-7), 30.6 (C-21), 28.1 (CH3 in Boc), 28.0 (C-15, C-23), 24.1 (C-16), 23.6 (C-2), 23.5 (C-27), 23.3 (C-11), 21.3 (C-30), 21.0 (COCH3), 18.2 (C-6), 17.0 (C-29), 16.9 (C-26), 16.7 (C-24), 15.5 (C-25); Anal. Calcd. for C47H77N3O10: C, 66.87, H, 9.19. Found: C, 67.24, H, 9.14%. MS: m/z 866.47 [M + Na]+ (calcd. for C47H77N3O10, 843.56).

3.1.4. General Procedure for the Synthesis of Compounds 9a13a, 15b, 18b and 20b

Compounds 913 and 15a, 18a, 20a (0.2 mmol) in 1 mL of dry CH2Cl2 were treated with TFA (1 mL) and the mixture was stirred for 4–6 h at room temperature (TLC control, hexane:EtOAc, 1:1, v/v). The solution was evaporated to dryness to obtain pure compounds 9a13a, 15b, 18b and 20b.
N-(4-Butylgyanidine)-3-oxolupane-28-amide trifluoroacetate (9a), White powder, 96% yield; mp 142–144 °C (EtOH); [ α ] D 19 −0.6° (c 0.34, CHCl3); IR (CHCl3) νmax 1669 (C=O), 3206, 3437 (NH) cm−1; 19F-NMR (376.50 MHz, CDCl3) δ: −75.91; 1H-NMR (400 MHz, CDCl3) δ: 11.13 (br s, 1H, NH in Boc), 7.50 (br s, 1H, NH–C=N), 6.82 (br s, 2H, NH2), 6.39 (br s, 1H, CONH), 3.26 (m, 4H, H-1′, H-4′), 2.45–1.19 (m, 26H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 1.07, 1.01, 0.96, 0.92, 0.90 (all s, 3H each, H-23–H-27), 0.85 (d, J = 6.0 Hz, 3H, H-29), 0.75 (d, J = 6.0 Hz, 3H, H-30); 13C-NMR (100 MHz, CDCl3) δ: 220.4 (C-3), 178.8 (C-28), 161.4 (q, JC,F = 37 Hz), 157.2 (C=N), 117.5 (q, JC,F = 288 Hz), 56.4 (C-17), 54.8 (C-5), 49.6 (C-9), 49.2 (C-19), 47.4 (C-4), 44.4 (C-18), 42.7 (C-14), 40.9 (C-1, C-4′), 40.7 (C-8), 38.8 (C-22), 38.1 (C-1′), 38.0 (C-13), 36.8 (C-10), 34.2 (C-2), 33.6 (C-16), 33.3 (C-7), 29.9 (C-20), 29.7 (C-3′), 29.4 (C-15), 27.0 (C-2′), 26.7 (C-23), 25.5 (C-12), 23.0 (C-21), 22.9 (C-29), 20.9 (C-11, C-24), 19.6 (C-6), 15.8 (C-25), 15.6 (C-26), 14.5 (C-30), 14.4 (C-27); Anal. Calcd. for C37H61F3N4O4: C, 65.08, H, 9.00. Found: C, 65.52, H, 8.94%. MS: m/z 569.43 [M + H]+ (calcd. for C35H60N4O2, 568.47).
3β-N-(2-Ethylgyanidine)-3-O-acetyl-lupane-28-amide trifluoroacetate (10a), White powder, 93% yield; mp 132–134 °C (EtOH); [ α ] D 19 −5.6° (c 0.31, CHCl3); IR (CHCl3) νmax 1671 (C=O), 3351 (NH) cm−1; 19F-NMR (470.59 MHz, CDCl3) δ: −75.80; 1H-NMR (500 MHz, CDCl3) δ: 10.63 (br s, 1H, NH in Boc), 8.05 (br s, 1H, NH–C=N), 7.00 (br s, 2H, NH2), 6.79 (br s, 1H, CONH), 4.48–4.45 (m, 1H, H-3), 3.39–3.28 (m, 4H, H-1′, H-2′), 2.09 (s, 3H, CH3CO–), 2.31–1.01 (m, 25H, CH, CH2 in pentacyclic skeleton), 0.95, 0.92, 0.89, 0.86, 0.85, 0.83 (all s, 3H each, H-23–H-27 and H-29), 0.79 (d, J = 11.0 Hz, 1H, H-5), 0.75 (d, J = 6.0 Hz, 3H, H-30); 13C-NMR (125 MHz, CDCl3) δ: 179.6 (C-28), 171.6 (COCH3), 161.2 (q, JC,F = 37 Hz), 157.9 (C=N), 117.2 (q, JC,F = 287 Hz), 81.2 (C-3), 56.4 (C-17), 55.4 (C-5), 50.2 (C-9), 49.3 (C-19), 44.3 (C-18), 42.6 (C-14), 40.7 (C-1, C-2′), 40.6 (C-8), 38.6 (C-1′), 38.4 (C-22), 37.8 (C-4, C-13), 37.0 (C-10), 34.2 (C-7), 33.1 (C-16), 29.9 (C-20), 29.3 (C-15), 27.9 (C-23), 26.9 (C-2), 23.6 (C-12), 23.0 (C-21, C-29), 21.3 (COCH3), 21.0 (C-11), 18.1 (C-6), 16.4 (C-25), 16.0 (C-24), 15.9 (C-26), 14.5 (C-27, C-30); Anal. Calcd. for C37H61F3N4O5: C, 63.59, H, 8.80. Found: C, 63.88, H, 8.73%. MS: m/z 585.62 [M + H]+ (calcd. for C35H60N4O3, 584.47).
3β-N-(4-Butylgyanidine)-3-O-acetyl-lupane-28-amide trifluoroacetate (11a), White powder, 95% yield; mp 148–150 °C (EtOH); [ α ] D 17 −12° (c 0.49, CHCl3); IR (CHCl3) νmax 1672 (C=O), 3207, 3354 (NH) cm−1; 19F-NMR (470.59 MHz, CDCl3) δ: −77.19; 1H-NMR (500 MHz, MeOD) δ: 4.48–4.45 (m, 1H, H-3), 3.28–3.15 (m, 4H, H-1′, H-4′), 2.04 (s, 3H, CH3CO–), 2.61–1.04 (m, 25H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 1.02, 0.98, 0.92, 0.90, 0.89, 0.88 (3H each, all s, H-23–H-27 and H-29), 0.84 (d, J = 11.0 Hz, 1H, H-5), 0.79 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 179.6 (C-28), 172.9 (COCH3), 161.2 (q, JC,F = 37 Hz), 158.8 (C=N), 117.2 (q, JC,F = 287 Hz), 82.6 (C-3), 57.5 (C-17), 57.0 (C-5), 51.9 (C-9), 51.1 (C-19), 45.5 (C-18), 43.8 (C-14), 42.2 (C-8, C-4′), 40.0 (C-22), 39.8 (C-1), 39.4 (C-1′), 39.0 (C-4), 38.9 (C-13), 38.4 (C-10), 35.8 (C-7), 34.1 (C-16), 31.3 (C-20), 30.7 (C-3′), 28.7 (C-15), 28.5 (C-23), 28.1 (C-2′), 27.4 (C-2), 24.8 (C-12), 24.2 (C-29), 23.7 (C-21), 22.5 (C-11), 21.3 (COCH3), 19.4 (C-6), 17.2 (C-25), 17.0 (C-24, C-26), 15.3 (C-30), 15.2 (C-27); Anal. Calcd. for C39H65F3N4O5: C, 64.44, H, 9.01. Found: C, 64.87, H, 8.94%. MS: m/z 613.48 [M + H]+ (calcd. for C37H64N4O3, 612.50).
3β-N-[2-(N,N-bis-Ethylgyanidine)-aminoethyl]-3-O-acetyl-lupane-28-amide trifluoroacetate (12a), White powder, 96% yield; mp 116–118 °C (EtOH); [ α ] D 17 −10.5° (c 0.2, C2H5OH); IR (CHCl3) νmax 1681 (C=O), 3199, 3362 (NH) cm−1; 19F-NMR (470.59 MHz, MeOD) δ: −76.98; 1H-NMR (500 MHz, MeOD) δ: 4.48–4.44 (m, 1H, H-3), 3.73–3.70 (m, 4H, H-4′, H-4″), 3.56–3.53 (m, 2H, H-1′), 3.46 (t, J = 6.5 Hz, 4H, H-3′, H-3″), 3.29–3.26 (m, 2H, H-2′), 2.04 (s, 3H, CH3CO–), 2.52–1.06 (m, 25H, CH, CH2 in pentacyclic skeleton), 1.02, 0.97, 0.92, 0.90, 0.88, 0.87 (all s, 3H each, H-23–H-27 and H-29), 0.85 (d, J = 11.5 Hz, 1H, H-5), 0.79 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 181.2 (C-28), 173.0 (COCH3), 161.7 (q, JC,F = 37 Hz), 159.1 (C=N), 117.5 (q, JC,F = 288 Hz), 35.6 (C-4″), 53.4 (C-3″), 82.6 (C-3), 57.6 (C-17), 56.9 (C-5), 53.9 (C-2′), 53.4 (C-3′), 51.8 (C-9), 51.0 (C-19), 45.5 (C-18), 43.8 (C-14), 42.2 (C-8), 39.7 (C-1), 39.7 (C-22), 39.0 (C-13, C-4), 38.4 (C-1′), 37.8 (C-10), 35.6 (C-4′, C-7), 33.8 (C-16), 31.3 (C-20), 30.7 (C-15), 28.6 (C-23), 28.4 (C-2), 24.8 (C-12), 24.1 (C-21), 23.5 (C-29), 22.3 (C-11), 21.3 (COCH3), 19.4 (C-6), 17.1 (C-25), 17.0 (C-24), 16.9 (C-26), 15.2 (C-30), 15.1 (C-27); Anal. Calcd. for C44H74F6N8O7: C, 56.16, H, 7.93. Found: C, 56.63, H, 7.86%. MS: m/z 713.59 [M + H]+ (calcd. for C40H72N8O3, 712.57).
3β-N-{[3-(3-Propylguanidine)piperazinyl]propyl}-3-O-acetyl-lupane-28-amide trifluoroacetate (13a), White powder, 96% yield; mp 92–94 °C (EtOH); [ α ] D 19 +8° (c 0.54, CHCl3); IR (CHCl3) νmax 1673, 1773 (C=O), 3190, 3367 (NH) cm−1; 19F-NMR (470.59 MHz, MeOD) δ: −77.25; 1H-NMR (500 MHz, MeOD) δ: 4.48–4.45 (m, 1H, H-3), 3.49–3.40 (m, 4H, H-2″, H-3″), 3.31–3.22 (m, 8H, H-4′, H-4″, H-5′, H-5″), 3.13–3.10 (m, 2H, H-1″), 3.00–2.98 (m, 2H, H-1′), 2.58–0.84 (m, 26H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 2.08 (s, 3H, CH3CO–), 1.02, 0.99, 0.92, 0.89, 0.86, 0.84 (all s, 3H each, H-23–H-27 and H-29), 0.80 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 180.2 (C-28), 173.0 (COCH3), 161.7 (q, JC,F = 37 Hz), 158.9 (C=N), 117.5 (q, JC,F = 288 Hz), 82.6 (C-3), 57.6 (C-17), 57.0 (C-5), 56.0 (C-3″), 55.2 (C-3′), 51.9 (C-9), 51.0 (C-19), 50.1 (C-4′, C-4″, C-5′, C-5″), 45.5 (C-18), 43.8 (C-14), 42.2 (C-8), 39.9 (C-1′), 39.8 (C-1″), 39.6 (C-1, C-22), 39.0 (C-4), 38.9 (C-13), 38.4 (C-10), 35.7 (C-7), 34.0 (C-16), 31.3 (C-20), 30.7 (C-15), 28.6 (C-23), 28.5 (C-2″), 25.6 (C-2′), 24.9 (C-2), 24.2 (C-12), 23.6 (C-21, C-29), 22.4 (C-11), 21.3 (COCH3), 19.4 (C-6), 17.1 (C-25), 17.0 (C-24), 16.9 (C-26), 15.2 (C-30), 15.1 (C-27); Anal. Calcd. for C45H77F3N6O5: C, 64.41, H, 9.25. Found: C, 64.88, H, 9.19. MS: m/z 747.58 [M + Na]+ (calcd. for C43H76N6O3, 724.60).
3β-[2-Guanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-lupane-28-oate trifluoroacetate (15b), White powder, 95% yield; mp 126–129 °C (EtOH); [ α ] D 19 −9.5° (c 0.34, CHCl3); IR (CHCl3) νmax 1620, 1698 (C=O), 3437 (NH) cm−1; 19F-NMR (470.59 MHz, CDCl3) δ: −75.97; 1H-NMR (500 MHz, MeOD) δ: 4.50–4.47 (m, 1H, H-3), 4.31 (2H, m, H-1′), 3.75 (m, 4H, H-3′, H-4′), 2.18–0.99 (m, 26H, CH, CH2 in pentacyclic skeleton), 2.08 (s, 3H, CH3CO–), 0.98, 0.97, 0.96, 0.87, 0.86 (all s, 3H each, H-23–H-27), 0.89 (d, J = 6.0 Hz, 3H, H-29), 0.76 (d, J = 6.0 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 177.4 (C-28), 172.3 (COCH3), 161.4 (q, JC,F = 37 Hz), 157.5 (C=N), 117.5 (q, JC,F = 288 Hz), 81.7 (C-3), 63.1 (C-3′, C-4′), 62.0 (C-2′), 60.2 (C-1′), 57.5 (C-17), 55.4 (C-5), 50.2 (C-9), 48.8 (C-19), 44.1 (C-18), 42.6 (C-14), 40.7 (C-8), 38.4 (C-22), 38.2 (C-1), 37.8 (C-4, C-13), 37.0 (C-10), 34.2 (C-7), 31.9 (C-16), 29.7 (C-20), 29.0 (C-15), 27.9 (C-23), 26.8 (C-2), 23.6 (C-12), 22.9 (C-29, C-21), 21.3 (COCH3), 20.9 (C-11), 18.1 (C-6), 16.4 (C-25), 16.1 (C-24), 15.8 (C-26), 14.6 (C-27, C-30); Anal. Calcd. for C39H64F3N3O8: C, 61.64; H, 8.49. Found: C, 62.34; H, 8.43%. MS: m/z 646.39 [M + H]+ (calcd. for C37H63N3O6, 645.47).
3β-[2-Guanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-urs-12-en-28-oate trifluoroacetate (18b), White powder, 98% yield; mp 124–126 °C (EtOH); [ α ] D 19 +30.0° (c 0.72, CH2Cl2); IR (CHCl3) νmax 1619, 1682 (C=O), 3438 (NH) cm−1; 19F-NMR (470.59 MHz, MeOD) δ: −77.17; 1H-NMR (500 MHz, MeOD) δ: 5.29 (br s, 1H, H-12), 4.49–4.46 (m, 1H, H-3), 4.31, 4.17 (both d, J = 11.5 Hz, 1H each, H-1′), 3.75 (m, 4H, H-3′, H-4′), 2.24 (d, J = 11.0 Hz, 1H, H-18), 2.04 (s, 3H, CH3CO–), 2.13–0.87 (m, 22H, CH, CH2 in pentacyclic skeleton), 1.15, 1.01, 0.97, 0.98, 0.92, 0.90, 0.81 (3H each, all s, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, MeOD) δ: 179.2 (C-28), 173.0 (COCH3), 161.4 (q, JC,F = 37 Hz), 159.2 (C=N), 140.1 (C-13), 127.2 (C-12), 117.5 (q, JC,F = 288 Hz), 82.5 (C-3), 64.3 (C-3′), 64.2 (C-4′), 62.7 (C-2′), 62.2 (C-1′), 56.8 (C-5), 54.4 (C-18), 49.5 (C-17), 48.6 (C-9), 43.3 (C-14), 40.9 (C-8), 40.4 (C-4, C-19), 39.6 (C-20), 38.8 (C-1), 38.1 (C-22), 38.0 (C-10), 34.2 (C-7), 31.7 (C-21), 29.2 (C-15), 28.7 (C-23), 25.5 (C-16), 24.6 (C-2), 24.5 (C-27), 24.4 (C-11), 21.6 (C-30), 21.3 (COCH3), 19.4 (C-6), 17.9 (C-29), 17.8 (C-26), 17.3 (C-24), 16.2 (C-25); Anal. Calcd. for C39H62F3N3O8: C, 61.80, H, 8.25. Found: C, 62.33, H, 8.19%. MS: m/z 644.47 [M + H]+ (calcd. for C37H61N3O6, 643.46).
3β-[2-Guanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-olean-12-en-28-oate trifluoroacetate (20b), White powder, 97% yield; mp 130–132 °C (EtOH); [ α ] D 17 +26.6° (c 0.53, C2H5OH); IR (CHCl3) νmax 1619, 1682 (C=O), 3438 (NH) cm−1; 19F-NMR (470.59 MHz, CDCl3) δ: −77.24; 1H-NMR (500 MHz, MeOD) δ: 5.31 (br s, 1H, H-12), 4.49–4.46 (m, 1H, H-3), 4.33, 4.20 (d, J = 11.5 Hz, 1H each, H-1′), 3.75 (m, 4H, H-3′, H-4′), 2.89 (d, 1H, J = 9.5 Hz, H-18), 2.09–0.88 (m, 22H, CH, CH2 in pentacyclic skeleton), 2.04 (s, 3H, CH3CO–), 1.19, 1.00, 0.97, 0.95, 0.90, 0.89, 0.79 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, MeOD) δ: 178.9 (C-28), 173.0 (COCH3), 161.4 (q, JC,F = 37 Hz), 159.2 (C=N), 145.0 (C-13), 124.1 (C-12), 117.5 (q, JC,F = 288 Hz), 82.6 (C-3), 64.3 (C-4′), 64.2 (C-3′), 62.7 (C-2′), 62.3 (C-1′), 56.9 (C-5), 48.5 (C-9, C-17), 47.1 (C-19), 43.0 (C-14), 42.9 (C-18), 40.8 (C-8), 39.5 (C-1), 38.9 (C-4), 38.3 (C-10), 34.9 (C-22), 33.9 (C-30), 33.7 (C-7), 33.6 (C-21), 31.7 (C-20), 28.9 (C-15), 28.7 (C-23), 26.6 (C-27), 24.7 (C-11, C-29), 24.3 (C-2), 24.2 (C-16), 21.3 (COCH3), 19.5 (C-6), 17.9 (C-26), 17.3 (C-24), 16.1 (C-25); Anal. Calcd. for C39H62F3N3O8: C, 61.80, F, 7.52, H, 8.25. Found: C, 62.12, H, 8.21%. MS: m/z 644.57 [M + H]+ (calcd. for C37H61N3O6, 643.46).

3.1.5. General Procedure for the Synthesis of Compounds 9b12b, 15c, 18c and 20c

The compounds 9a12a, 15b, 18b, 20b (0.2 g) were dissolved in 2 mL MeOH and 5M HCl was added dropwise until the precipiate formed. The solution was evaporated to dryness and this procedure was repeated three times. The precipitate which formed was filtered off and washed with water to pH = 7. The salts 9b12b and 15c, 18c, 20c were obtained as white solids with a quantitative yield.
N-(4-Butylgyanidine)-3-oxolupane-28-amide dihydrochloride (9b), White powder, 87% yield; mp 176–178 °C (EtOH); [ α ] D 21 −11° (c 0.29, C2H5OH); IR (CHCl3) νmax 1721 (C=O), 3342 (NH) cm−1; 1H-NMR (500 MHz, d6-DMSO) δ: 7.71 (br s, 1H, NH), 7.59 (br s, 1H, CONH), 3.10–3.01 (m, 4H, H-1′, H-4′), 2.61–1.04 (m, 26H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 0.99, 0.93, 0.91, 0.87, 0.86 (3H each, all s, H-23–H-27), 0.81 (d, J = 6.5 Hz, 3H, H-29), 0.71 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, d6-DMSO) δ: 217.1 (C-3), 176.1 (C-28), 157.4 (C=N), 55.7 (C-17), 54.3 (C-5), 49.6 (C-9), 49.5 (C-19), 47.0 (C-4), 43.8 (C-18), 42.6 (C-14), 40.9 (C-4′), 40.3 (C-8), 39.7 (C-1), 38.1 (C-22, C-1′), 37.0 (C-13), 36.8 (C-10), 34.1 (C-2), 33.8 (C-16), 32.7 (C-7), 30.0 (C-20), 29.4 (C-15), 27.2 (C-3′), 27.0 (C-2′), 26.9 (C-23), 26.4 (C-12), 23.6 (C-29), 23.1 (C-21), 21.7 (C-11), 21.2 (C-24), 19.7 (C-6), 16.2 (C-25), 16.1 (C-26), 15.0 (C-30), 14.6 (C-27); Anal. Calcd. for C35H62Cl2N4O2: C, 65.50, Cl, 11.05, H, 9.74. Found: C, 65.97, Cl, 11.78, H, 9.68%. MS: m/z 569.49 [M + H]+ (calcd. for C35H60N4O2, 568.47).
3β-N-(2-Ethylgyanidine)-3-O-acetyl-lupane-28-amide hydrochloride (10b), White powder, 82% yield; mp 192–194 °C (EtOH); [ α ] D 17 −16° (c 0.23, C2H5OH); IR (CHCl3) νmax 1652, 1716 (C=O), 3155, 3327 (NH) cm−1; 1H-NMR (500 MHz, MeOD) δ: 4.48–4.45 (m, 1H, H-3), 3.43–3.16 (m, 4H, H-1′, H-2′), 2.04 (s, 3H, CH3CO–), 2.60–0.81 (m, 25H, CH, CH2 in pentacyclic skeleton), 1.02, 0.98, 0.92, 0.90, 0.88, 0.87 (all s, 3H each, H-23–H-27 and H-29), 0.80 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 180.8 (C-28), 173.0 (COCH3), 159.0 (C=N), 82.6 (C-3), 57.6 (C-17), 57.0 (C-5), 51.1 (C-9, C-19), 45.5 (C-18), 42.7 (C-14, C-1′), 42.2 (C-8), 39.8 (C-1), 39.7 (C-2′), 39.4 (C-22), 39.0 (C-4), 38.9 (C-13), 38.4 (C-10), 35.7 (C-7), 34.0 (C-16), 31.4 (C-20), 30.7 (C-15), 28.6 (C-23), 28.5 (C-2), 24.8 (C-12), 24.1 (C-21), 23.6 (C-29), 22.4 (C-11), 21.3 (COCH3), 19.4 (C-6), 17.1 (C-25), 16.9 (C-24, C-26), 15.2 (C-30), 15.1 (C-27); Anal. Calcd. for C35H61ClN4O3: C, 67.66, Cl, 5.71, H, 9.90. Found: C, 67.99, Cl, 5.40, H, 9.84%. MS: m/z 585.54 [M + H]+ (calcd. for C35H60N4O3, 584.47).
3β-N-(4-Butylgyanidine)-3-O-acetyl-lupane-28-amide hydrochloride (11b), White powder, 79% yield; mp 156–158 °C (EtOH); [ α ] D 22 −14.5° (c 0.53, C2H5OH); IR (CHCl3) νmax 1645, 1716 (C=O), 3168, 3338 (NH) cm−1; 1H-NMR (500 MHz, MeOD) δ: 7.45 (br s, 1H, NH), 4.48–4.45 (m, 1H, H-3), 3.23–3.18 (m, 4H, H-1′, H-4′), 2.04 (s, 3H, CH3CO–), 2.61–0.84 (m, 26H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 1.01, 0.98, 0.92, 0.89, 0.88, 0.86 (all s, 3H each, H-23–H-27 and H-29), 0.79 (d, J = 7.0 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 179.6 (C-28), 172.9 (COCH3), 158.8 (C=N), 82.6 (C-3), 57.5 (C-17), 57.0 (C-5), 51.9 (C-9), 51.1 (C-19), 45.5 (C-18), 43.8 (C-14), 42.4 (C-8), 42.2 (C-4′), 40.0 (C-22, C-1′), 39.8 (C-1), 39.0 (C-4), 38.9 (C-13), 38.4 (C-10), 35.8 (C-7), 34.1 (C-16), 31.4 (C-20), 30.8 (C-3′), 28.6 (C-15), 28.2 (C-23, C-2′), 27.4 (C-2), 24.8 (C-12), 24.4 (C-21), 24.2 (C-29), 22.5 (C-11), 21.3 (COCH3), 19.4 (C-6), 17.1 (C-25), 17 (C-24), 16.9 (C-26), 15.2 (C-30), 15.1 (C-27); Anal. Calcd. for C37H65ClN4O3: C, 68.43, Cl, 5.46, H, 10.09. Found: C, 68.88; Cl, 5.12; H, 10.02%. MS: m/z 614.51 [M + 2H]+ (calcd. for C37H64N4O3, 612.50).
3β-N-[2-(N,N-bis-Ethylgyanidine)-aminoethyl]-3-O-acetyl-lupane-28-amide dihydrochloride (12b), White powder, 88% yield; mp 198–200 °C (EtOH); [ α ] D 17 −16° (c 0.29, C2H5OH); IR (CHCl3) νmax 1637, 1672, 1734 (C=O), 3162, 3271, 3324 (NH) cm−1; 1H-NMR (500 MHz, MeOD) δ: 4.48–4.45 (m, 1H, H-3), 3.90–3.83 (m, 4H, H-4′, H-4″), 3.66–3.59 (m, 6H, H-2′, H-3′, H-3″), 3.38–3.33 (m, 2H, H-1′), 2.04 (s, 3H, CH3CO–), 2.51–0.85 (m, 26H, CH, CH2 in pentacyclic skeleton), 1.03, 0.99, 0.93, 0.90, 0.88, 0.87 (all s, 3H each, H-23–H-27 and H-29), 0.80 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 181.0 (C-28), 172.9 (COCH3), 158.9 (C=N), 82.5 (C-3), 57.7 (C-17), 56.9 (C-5), 53.7 (C-3′, C-3″), 53.6 (C-2′), 51.8 (C-9), 51.0 (C-19), 45.5 (C-18), 43.8 (C-14), 42.2 (C-8), 39.7 (C-22), 39.0 (C-1, C-13), 38.4 (C-4), 37.7 (C-10, C-1′), 35.7 (C-7), 35.0 (C-4′, C-4″), 33.9 (C-16), 31.3 (C-20), 30.8 (C-15), 28.6 (C-23), 28.4 (C-2), 24.8 (C-12), 24.2 (C-21), 23.6 (C-29), 22.4 (C-11), 21.3 (COCH3), 19.4 (C-6), 17.1 (C-25, C-24), 16.9 (C-26), 15.2 (C-30), 15.1 (C-27); Anal. Calcd. for C40H74Cl2N8O3: C, 61.13, Cl, 9.02, H, 9.49. Found: C, 61.55, Cl, 11.40, H, 9.42%. MS: m/z 713.51 [M + H]+ (calcd. for C40H72N8O3, 712.57).
3β-[2-Guanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-lupane-28-oate hydrochloride (15c), White powder, 68% yield; mp 136–138 °C (EtOH); [ α ] D 22 −14.5° (c 0.53, C2H5OH); IR (CHCl3) νmax 1673, 1733 (C=O), 3325 (NH) cm−1; 1H-NMR (500 MHz, MeOD) δ: 4.48–4.45 (m, 1H, H-3), 4.31 (2H, m, H-1′), 3.75 (m, 4H, H-3′, H-4′), 2.04 (s, 3H, CH3CO–), 2.29–0.85 (m, 26H, CH, CH2 in pentacyclic skeleton), 1.03, 1.00, 0.98, 0.91, 0.88, 0.87 (all s, 3H each, H-23–H-27 and H-29), 0.81 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, MeOD) δ: 177.1 (C-28, COCH3), 159.1 (C=N), 82.6 (C-3), 64.1 (C-3′), 64.1 (C-4′), 62.8 (C-2′), 61.8 (C-1′), 58.7 (C-17), 56.9 (C-5), 51.8 (C-9), 50.4 (C-19), 45.8 (C-18), 43.9 (C-14), 42.1 (C-8), 39.7 (C-22), 39.6 (C-1), 39.0 (C-4), 38.4 (C-10, C-13), 35.6 (C-7), 33.2 (C-16), 31.2 (C-20), 30.9 (C-15), 28.6 (C-23), 28.4 (C-2), 24.8 (C-12), 23.9 (C-21), 23.5 (C-29), 22.3 (C-11), 21.3 (COCH3), 19.4 (C-6), 17.1 (C-25), 16.9 (C-24), 16.8 (C-26), 15.2 (C-30), 15.1 (C-27); Anal. Calcd. for C37H64ClN3O6: C, 65.13, Cl, 5.20, H, 9.45. Found: C, 65.6 4, Cl, 4.50; H, 9.39%. MS: m/z 647.47 [M + 2H]+ (calcd. for C37H63N3O6, 645.47).
3β-[2-Guanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-urs-12-en-28-oate hydrochloride (18c), White powder, 62% yield; mp 142–144 °C (EtOH); [ α ] D 19 +37° (c 0.51, C2H5OH); IR (CHCl3) νmax 1675, 1733 (C=O), 3186, 3335 (NH) cm−1; 1H-NMR (500 MHz, MeOD) δ: 7.07 (s, 1H, NH), 5.30 (br s, 1H, H-12), 4.50–4.47 (m, 1H, H-3), 4.29, 4.23 (both d, J = 11.5 Hz, 1H each, H-1′), 3.75 (m, 4H, H-3′, H-4′), 2.25 (d, J = 11.5 Hz, 1H, H-18), 2.05 (s, 3H, CH3CO–), 2.14–0.88 (m, 22H, CH, CH2 in pentacyclic skeleton), 1.16, 1.02, 1.00, 0.92, 0.91, 0.90, 0.81 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, MeOD) δ: 178.6 (C-28), 173.0 (COCH3), 159.2 (C=N), 139.6 (C-13), 127.3 (C-12), 82.6 (C-3), 64.2 (C-3′, C-4′), 62.9 (C-2′), 62.3 (C-1′), 56.8 (C-5), 54.5 (C-18), 49.3 (C-17), 49.0 (C-9), 43.4 (C-14), 41.0 (C-8), 40.5 (C-4, C-19), 39.6 (C-20), 38.9 (C-1), 38.2 (C-22), 38.0 (C-10), 34.2 (C-7), 31.8 (C-21), 29.3 (C-15), 28.8 (C-23), 25.5 (C-16), 24.7 (C-2), 24.5 (C-27), 24.4 (C-11), 21.7 (C-30), 21.3 (COCH3), 19.5 (C-6), 17.9 (C-29), 17.8 (C-26), 17.4 (C-24), 16.2 (C-25); Anal. Calcd. for C37H62ClN3O6: C, 65.32, Cl, 5.21, H, 9.19. Found: C, 65.74, Cl, 4.51, H, 9.14%. MS: m/z 644.34 [M + H]+ (calcd. for C37H61N3O6, 643.46).
3β-[2-Guanidine-3-hydroxy-2-(hydroxymethyl)propyl]-3-O-acetyl-olean-12-en-28-oate hydrochloride (20c), White powder, 53% yield; mp 124–128 °C (EtOH); [ α ] D 19 +34° (c 0.34, C2H5OH); IR (CHCl3) νmax 1662, 1733 (C=O), 3344 (NH) cm−1; 1H-NMR (500 MHz, MeOD) δ: 5.32 (br s, 1H, H-12), 4.49–4.46 (m, 1H, H-3), 4.33, 4.20 (both d, J = 11.5 Hz, 1H each, H-1′), 3.76 (m, 4H, H-3′, H-4′), 2.89 (d, J = 9.5 Hz, 1H, H-18), 2.13–0.90 (m, 22H, CH, CH2 in pentacyclic skeleton), 2.05 (s, 3H, CH3CO–), 1.20, 1.00, 0.98, 0.96, 0.94, 0.91, 0.88 (all s, 3H each, H-23–H-27, H-29 and H-30); 13C-NMR (125 MHz, MeOD) δ: 178.8 (C-28), 172.9 (COCH3), 159.1 (C=N), 145.0 (C-13), 124.1 (C-12), 82.6 (C-3), 64.2 (C-4′), 64.1 (C-3′), 62.8 (C-2′), 62.3 (C-1′), 56.8 (C-5), 49.0 (C-9), 48.5 (C-17), 47.1 (C-19), 43.0 (C-14), 42.9 (C-18), 40.7 (C-8), 39.5 (C-1), 38.9 (C-4), 38.2 (C-10), 34.9 (C-22), 33.9 (C-30), 33.7 (C-7, C-21), 31.7 (C-20), 28.9 (C-15), 28.7 (C-23), 26.6 (C-27), 24.7 (C-11, C-29), 24.3 (C-2), 24.2 (C-16), 21.3 (COCH3), 19.5 (C-6), 17.9 (C-26), 17.3 (C-24), 15.6 (C-25); Anal. Calcd. for C37H62ClN3O6: C, 65.32, Cl, 5.21, H, 9.19. Found: C, 65.74, Cl, 4.80, H, 9.12%. MS: m/z 644.44 [M + H]+ (calcd. for C37H61N3O6, 643.46).
3β-N-(4-Butylgyanidine)-3-hydroxy-lupane-28-amide (14), To a solution of the compound 11b (0.33 g, 0.5 mmol) in MeOH (4 mL) and THF (4 mL) was added 4 N NaOH (4 mL). The reaction mixture was stirred at room temperature for 24 h (monitoring by TLC) and then neutralized with 20% HCl. The solution was dried under vacuum and reconstituted with CH2Cl2. The organic layer was washed with brine and dried over anhydrous MgSO4 and concentrated under reduced pressure to obtain pure compound 14. White powder, 79% yield; mp 260–262 °C (EtOH); [ α ] D 19 −10° (c 0.24, DMSO); IR (CHCl3) νmax 1731 (C=O), 3366 (NH) cm−1; 1H-NMR (500 MHz, DMSO-d5) δ: 7.73 (br s, 1H, NH), 7.58 (br s, 1H, CONH), 4.29 (br s, 1H, OH), 3.09–2.98 (m, 5H, H-3, H-1′, H-4′), 2.19–1.02 (m, 26H, CH, CH2 in pentacyclic skeleton, 4H, H-2′, H-3′), 0.89, 0.87, 0.84, 0.78, 0.66 (all s, 3H each, H-23–H-27), 0.81 (d, J = 6.5 Hz, 3H, H-29), 0.72 (d, J = 6.5 Hz, 3H, H-30); 13C-NMR (125 MHz, DMSO-d5) δ: 176.1 (C-28), 157.4 (C=N), 77.2 (C-3), 55.7 (C-17), 55.4 (C-5), 50.3 (C-9), 49.6 (C-19), 43.8 (C-18), 42.5 (C-14), 40.9 (C-8), 40.8 (C-4′), 39.0 (C-22), 38.8 (C-1), 38.6 (C-4), 38.1 (C-1′), 37.2 (C-13), 36.9 (C-10), 34.6 (C-7), 32.8 (C-16), 30.0 (C-20), 29.4 (C-15), 28.6 (C-23), 27.4 (C-2), 27.2 (C-3′), 27.0 (C-2′), 26.4 (C-12), 23.6 (C-29), 23.2 (C-21), 21.1 (C-11), 18.5 (C-6), 16.4 (C-25), 16.3 (C-24, C-26), 15.0 (C-30), 14.7 (C-27); Anal. Calcd. for C35H62N4O2: C, 73.63, H, 10.95. Found: C, 73.74, H, 10.88%. MS: m/z 593.31 [M + Na]+ (calcd. for C35H62N4O2, 570.49).

3.2. Biology

3.2.1. Cell Culturing

Cells (Jurkat, K562, U937, HeLa, HEK293 and normal Fibroblasts) were purchased from Russian Cell Culture Collection (Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia) and cultured according to standard mammalian tissue culture protocols and sterile technique. Human cell lines HEK293 and HeLa were obtained from the HPA Culture Collections (Salisbury, UK). All cell lines used in the study were tested and shown to be free of mycoplasma and viral contamination.
HEK293, HeLa cell lines and fibroblasts were cultured as monolayers and maintained in Dulbecco’s modified Eagle’s medium (DMEM, Gibco BRL, Waltham, MA, USA) supplemented with 10% foetal bovine serum and 1% penicillin-streptomycin solution at 37 °C in a humidified incubator under a 5% CO2 atmosphere.
Cells were maintained in RPMI 1640 (Jurkat, K562, U937) (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 4 mM glutamine, 10% FBS (Sigma, Burlington, MA, USA) and 100 units/mL penicillin-streptomycin (Sigma). All types of cells were grown in an atmosphere of 5% CO2 at 37 °C. The cells were subcultures at 2–3 days intervals. Adherent cells (HEK293, HeLa, fibroblasts) were suspended using trypsin/EDTA and counted after they have reached 80% confluency. Cells were then seeded in 24 well plates at 5 × 104 cells per well and incubated overnight. Jurkat, K562, U937 cells were subcultured at 2 day intervals with a seeding density of 1 × 105 cells per 24 well plates in RPMI with 10% FBS.

3.2.2. Cytotoxicity Assay

Viability (Live/dead) assessment was performed by staining cells with 7-AAD (7-Aminoactinomycin D) (Biolegend, San Diego, CA, USA). Cells were treated of test compounds with six different concentrations (1, 5, 10, 15, 30 and 60 μM). After treatment, cells were harvested, washed 1–2 times with phosphate-buffered saline (PBS) and centrifuged at 400× g for 5 min. Cell pellets were resuspended in 200 μL of flow cytometry staining buffer (PBS without Ca2+ and Mg2+, 2.5% FBS) and stained with 5 μL of 7-AAD staining solutionfor 15 min at room temperature in the dark. Samples were acquired on NovoCyteTM 2000 FlowCytometry System (ACEA, San Diego, CA, USA) equipped with 488 nm argon laser. Detection of 7-AAD emission was collected through a 675/30 nm filter in FL4 channel.

3.2.3. Viability and Apoptosis

Apoptosis was determined by flow cytometric analysis of Annexin V and 7-aminoactinomycin D staining. Briefly, 200 μL of Guava Nexin reagent (Millipore, Bedford, MA, USA) was added to 5 × 105 cells in 200 μL, and the cells were incubated with the reagent for 20 min at room temperature in the dark. The plates were treated with compounds 15, 15c, 18c, 20c and dihydrobetulinic acid at IC50 concentration (4, 8 and 59 μM) for 24 h and 48 h. At the end of incubation, the cells were analyzed on NovoCyteTM 2000 FlowCytometry System (ACEA). Different states of cell death were defined as follows: normal cells are localized in the lower-left quadrant (Annexin V/PI); early apoptotic cells are in the lower-right quadrant (Annexin V+/PI); late apoptotic cells and necrotic cells are in the upper-right quadrant (Annexin V+/PI+); and necrotic cells are in the upper-left quadrant (Annexin V/PI+).

3.2.4. Cell Cycle Analysis

Cell cycle was analyzed using the method of propidium iodide staining. Briefly, cells were plated in 24-well round bottom plates at density 10 × 105 cells per well, centrifuged at 450× g for 5 min, and fixed with ice-cold 70% ethanol for 24 h at 0 °C. Cells were then washed with PBS and incubated with 250 μL of Guava Cell Cycle Reagent (Millipore, Burlington, MA, USA) for 30 min at room temperature in the dark. Samples were analyzed on NovoCyteTM 2000 FlowCytometry System (ACEA, San Diego, CA, USA).

4. Conclusions

Novel betulinic, ursolic, and oleanolic acid derivatives, containing a guanidine moiety have been designed and synthesized in an attempt to develop potent antitumor agents. These compounds and their precursors, monoamine, diamine and triamine derivatives, were tested for cytotoxic activity on various human tumor cell lines. Guanidine-functionalized triterpenoids demonstrated higher cytotoxicity in Jurkat cells, compared with original triterpenoic acids. Most of the tested guanidine derivatives showed higher IC50 values than amines, but were less toxic to human fibroblasts. The lead molecules—dihydrobetulinic acid amine 15, its guanidine derivative 15c, and guanidinium salts of ursolic and oleanolic acids 18c and 20c were selected for extended biological testing by using flow cytometry analysis. Our results showed that the antitumor activity of compounds 15, 15c, and 18c is caused by apoptotic processes and induction of cell cycle arrest in the S-phase. Nevertheless, addition information concerning the molecular mechanisms and targets of these triterpene acid derivatives is needed.

Supplementary Materials

The following are available online: 1H-NMR and 13C-NMR spectra of all new compounds.

Author Contributions

Supervision, U.D. and V.O.; validation and writing-review & editing, A.S. and L.D.; performing the chemistry experiments R.K. and D.N.; performing the biology experiments M.Y., L.D. and V.D.; The manuscript was prepared through the contributions L.D., V.D. and D.N.

Funding

This work was performed under financial support from the Russian Science Foundation (Grant 16-13-10051).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hill, R.A.; Connolly, J.D. Triterpenoids. Nat. Prod. Rep. 2017, 34, 90–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Dzubak, P.; Hajduch, M.; Vydra, D.; Hustova, A.; Kvasnica, M.; Biedermann, D.; Markova, L.; Urban, M.; Sarek, J. Pharmacological activities of natural triterpenoids and their therapeutic implications. Nat. Prod. Rep. 2006, 23, 394–411. [Google Scholar] [CrossRef] [PubMed]
  3. Sarek, J.; Kvasnica, M.; Vlk, M.; Urban, M.; Dzubak, P.; Hajduch, M. The potential of triterpenoids in the treatment of melanoma. In Research on Melanoma: A Glimpse into Current Directions and Future Trends; Murph, M., Ed.; IntechOpen: Rijeka, Croatia, 2011; pp. 125–158. [Google Scholar]
  4. Sheng, H.; Sun, H. Synthesis, biology and clinical significance of pentacyclic triterpenes: A multi-target approach to prevention and treatment of metabolic and vascular diseases. Nat. Prod. Rep. 2011, 28, 543–593. [Google Scholar] [CrossRef] [PubMed]
  5. Salvador, J.A.R.; Moreira, V.M.; Goncalves, B.M.F.; Lealab, A.S.; Jing, Y. Ursane-type pentacyclic triterpenoids as useful platforms to discover anticancer drugs. Nat. Prod. Rep. 2012, 29, 1463–1479. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, H.; Gao, Y.; Wang, A.; Zhou, X.; Zheng, Y.; Zhou, J. Evolution in medicinal chemistry of ursolic acid derivatives as anticancer agents. Eur. J. Med. Chem. 2015, 92, 648–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Cichewicz, R.H.; Kouzi, S.A. Chemistry, biological activity, and chemotherapeutic potential of betulinic acid for the prevention and treatment of cancer and HIV infection. Med. Res. Rev. 2004, 24, 90–114. [Google Scholar] [CrossRef] [PubMed]
  8. Csuk, R. Betulinic acid and its derivatives: A patent review (2008–2013). Expert Opin. Ther. Pat. 2014, 24, 913–923. [Google Scholar] [CrossRef] [PubMed]
  9. Fulda, S.; Galluzzi, L.; Kroemer, G. Targeting mitochondria for cancer therapy. Nat. Rev. Drug Discov. 2010, 9, 447–464. [Google Scholar] [CrossRef] [PubMed]
  10. Damle, A.A.; Pawar, Y.P.; Narkar, A.A. Anticancer activity of betulinic acid on MCF-7 tumors in nude mice. Indian J. Exp. Biol. 2013, 51, 485–491. [Google Scholar] [PubMed]
  11. Mullauer, F.B.; Bloois, L.; Daalhuisen, J.B.; Brink, M.S.T.; Storm, G.; Medema, J.P.; Schiffelers, R.M.; Kessler, J.H. Betulinic acid delivered in liposomes reduces growth of human lung and colon cancers in mice without causing systemic toxicity. Anticancer Drugs. 2011, 22, 223–233. [Google Scholar] [CrossRef] [PubMed]
  12. Pathak, A.K.; Bhutani, M.; Nair, A.S.; Ahn, K.S.; Chakraborty, A.; Kadara, H.; Guha, S.; Sethi, G.; Aggarwal, B.B. Ursolic acid inhibits STAT3 activation pathway leading to suppression of proliferation and chemosensitization of human multiple myeloma cells. Clin. Cancer Res. 2007, 5, 943–955. [Google Scholar] [CrossRef] [PubMed]
  13. Villar, V.H.; Vögler, O.; Barceló, F.; Broto, J.M.; Serra, J.M.; Gutiérrez, V.R.; Alemany, R. Down-regulation of AKT signalling by ursolic acid induces intrinsic apoptosis and sensitization to doxorubicin in soft tissue sarcoma. PLoS ONE 2016, 11, e0155946. [Google Scholar] [CrossRef] [PubMed]
  14. Lin, C.; Wen, X.; Sun, H. Oleanolic acid derivatives for pharmaceutical use: A patent review. Expert Opin. Ther. Pat. 2016, 26, 643–655. [Google Scholar] [CrossRef] [PubMed]
  15. Hussain, H.; Green, I.R.; Ali, I.; Khan, I.A.; Ali, Z.; Al-Sadi, A.M.; Ahmed, I. Ursolic acid derivatives for pharmaceutical use: A patent review (2012–2016). Expert Opin. Ther. Pat. 2017, 27, 1061–1072. [Google Scholar] [CrossRef] [PubMed]
  16. Biedermann, D.; Eignerova, B.; Hajduch, M.; Sarek, J. Synthesis and evaluation of biological activity of the quaternary ammonium salts of lupane-, oleanane-, and ursane-type acids. Synthesis 2010, 22, 2839–3848. [Google Scholar] [CrossRef]
  17. Kataev, V.E.; Strobykina, Y.; Zakharova, L.Y. Quaternary ammonium derivatives of natural terpenoids. Synthesis and properties. Russ. Chem. Bull. 2014, 63, 1884–1900. [Google Scholar] [CrossRef]
  18. Serafim, T.L.; Carvalho, F.S.; Bernardo, T.C.; Pereira, G.C.; Perkins, E.; Holy, J.; Krasutsky, D.A.; Kolomitsyna, O.N.; Krasutsky, P.A.; Oliveira, P.J. New derivatives of lupine triterpenoids disturb breast cancer mitochondria and induce cell death. Bioorg. Med. Chem. 2014, 22, 6270–6287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Bernardo, T.C.; Cunha-Oliveira, T.; Serafim, T.L.; Holy, J.; Krasutsky, D.; Kolomitsyna, O.; Krasutsky, P.; Moreno, A.M.; Oliveira, P.J. Dimethylaminopyridine derivatives of lupine triterpenoids cause mitochondrial disruption and induce the permeability transition. Bioorg. Med. Chem. 2013, 21, 7239–7249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Tsepaeva, O.V.; Nemtarev, A.V.; Abdullin, T.I.; Grigor’eva, L.R.; Kuznetsova, E.V.; Akhmadishina, R.A.; Ziganshina, L.E.; Cong, H.H.; Mironov, V.F. Design, synthesis, and cancer cell growth inhibitory activity of triphenylphosphonium derivatives of the triterpenoid betulin. J. Nat. Prod. 2017, 80, 2232–2239. [Google Scholar] [CrossRef] [PubMed]
  21. Ye, Y.; Zhang, T.; Yuan, H.; Li, D.; Lou, H.; Fan, P. Mitochondria-targeted lupanetriterpenoid derivatives and their selective apoptosis-Inducing anticancer mechanisms. J. Med. Chem. 2017, 60, 6353–6363. [Google Scholar] [CrossRef] [PubMed]
  22. Strobykina, I.Y.; Belenok, M.G.; Semenova, M.N.; Semenov, V.V.; Babaev, V.M.; Rizvanov, I.K.; Mironov, V.F.; Kataev, V.E. Triphenylphosphonium cations of the diterpenoid isosteviol: Synthesis and antimitotic activity in a sea urchin embryo model. J. Nat. Prod. 2015, 78, 1300–1308. [Google Scholar] [CrossRef] [PubMed]
  23. Spivak, A.Y.; Nedopekina, D.A.; Shakurova, E.R.; Khalitova, R.R.; Gubaidullin, R.R.; Odinokov, V.N.; Dzhemilev, U.M.; Bel’skii, Y.P.; Bel’skaya, N.V.; Stankevich, S.A.; et al. Synthesis of lupine triterpenoids with triphenylphosphonium substituents and studies of their antitumor activity. Russ. Chem. Bull. 2013, 62, 188–198. [Google Scholar] [CrossRef]
  24. Spivak, A.Y.; Nedopekina, D.A.; Khalitova, R.R.; Gubaidullin, R.R.; Odinokov, V.N.; Bel’skii, Y.P.; Bel’skaya, N.V.; Khazanov, V.A. Triphenylphosphonium cations of betulinic acid derivatives: Synthesis and antitumor activity. Med. Chem. Res. 2017, 26, 518–531. [Google Scholar] [CrossRef]
  25. Nedopekina, D.A.; Gubaidullin, R.R.; Odinokov, V.N.; Maximchik, P.V.; Zhivotovsky, B.; Bel’skii, Y.P.; Khazanov, V.A.; Manuylova, A.V.; Gogvadze, V.; Spivak, A.Y. Mitohondria-targeted betulinic and ursolic acid derivatives: Synthesis and anticancer activity. Med. Chem. Commun. 2017, 8, 1934–1945. [Google Scholar] [CrossRef] [PubMed]
  26. Saczewski, F.; Balewski, L. Biological activities of guanidine compounds. Expert Opin. Ther. Pat. 2009, 19, 1417–1448. [Google Scholar] [CrossRef] [PubMed]
  27. Wexselblatt, E.; Esko, J.D.; Tor, Y. On guanidinium and cellular uptake. J. Org. Chem. 2014, 79, 6766–6774. [Google Scholar] [CrossRef] [PubMed]
  28. Pantos, A.; Tsogas, I.; Paleos, C.M. Guanidinium group: A versatile moiety inducing transport and multicompartmentalization in complementary membranes. Biochim. Biophys. Acta 2008, 1778, 811–823. [Google Scholar] [CrossRef] [PubMed]
  29. Castagnolo, D.; Schenone, S.; Botta, M. Guanylated diamines, triamines, and polyamines: Chemistry and biological properties. Chem. Rev. 2011, 111, 5247–5300. [Google Scholar] [CrossRef] [PubMed]
  30. Sibrian-Vazquez, M.; Nesterova, I.V.; Jensen, T.J.; Vicente, M.G. Mitochondria targeting by guanidine- and biguanidine-porphyrin photosensitizers. Bioconjug. Chem. 2008, 19, 705–713. [Google Scholar] [CrossRef] [PubMed]
  31. Blanchet, M.; Borselli, D.; Brunel, J.M. Polyamine derivatives: A revival of an old neglected scaffold to fight resistant Gram-negative bacteria? Future Med. Chem. 2016, 8, 963–973. [Google Scholar] [CrossRef] [PubMed]
  32. Nowotarski, S.L.; Woster, P.M.; Casero, R.A. Polyamines and cancer: Implications for chemotherapy and chemoprevention. Expert Rev. Mol. Med. 2013, 15, e3. [Google Scholar] [CrossRef] [PubMed]
  33. Fujiwara, T.; Hasegawa, S.; Hirashima, N.; Nakanishi, M.; Ohwada, T. Gene transfection activities of amphiphilic steroid-polyamine conjugates. Biochim. Biophys. Acta 2000, 1468, 396–402. [Google Scholar] [CrossRef]
  34. Brycki, B.; Koenig, H.; Kowalczyk, I.; Pospieszny, T. Synthesis, spectroscopic and theoretical studies of new dimeric quaternary alkylammonium conjugates of sterols. Molecules 2014, 19, 9419–9434. [Google Scholar] [CrossRef] [PubMed]
  35. Vida, N.; Svobodova, H.; Rarova, L.; Drasar, P.; Saman, D.; Cvacka, J.; Wimmer, Z. Polyamine conjugates of stigmasterol. Steroids 2012, 77, 1212–1218. [Google Scholar] [CrossRef] [PubMed]
  36. Brycki, B.; Koenig, H.; Pospieszny, T. Quaternary alkylammonium conjugates of steroids: Synthesis, molecular structure, and biological studies. Molecules 2015, 20, 20887–20900. [Google Scholar] [CrossRef] [PubMed]
  37. Giniyatullina, G.V.; Flekhter, O.B.; Tolstikov, G.A. Synthesis of squalamine analogues on the basis of lupine triterpenoids. Mendeleev Commun. 2009, 19, 32–33. [Google Scholar] [CrossRef]
  38. Kazakova, O.B.; Giniyatullina, G.V.; Medvedeva, N.I.; Tolstikov, G.A. Synthesis of a triterpene–spermidine conjugate. Russ. J. Org. Chem. 2012, 48, 1370–1373. [Google Scholar] [CrossRef]
  39. Bildziukevich, U.; Vida, N.; Rárová, L.; Kolář, M.; Šaman, D.; Havlíček, L.; Drašar, P.; Wimmer, Z. Polyamine derivatives of betulinic acid and β-sitosterol: A comparative investigation. Steroids 2015, 100, 27–35. [Google Scholar] [CrossRef] [PubMed]
  40. Wang, J.; Jiang, Z.; Xiang, L.; Li, Y.; Ou, M.; Yang, X.; Shao, J.; Lu, Y.; Lin, L.; Chen, J.; et al. Synergism of ursolic acid derivative US597 with 2-deoxy-D-glucose to preferentially induce tumor cell death by dual-targeting of apoptosis and glycolysis. Sci. Rep. 2014, 4, 5006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Kim, D.S.; Pezzuto, J.M.; Pisha, E. Synthesis of betulinic acid derivatives with activity against human melanoma. Bioorg. Med. Chem. Lett. 1998, 8, 1707–1712. [Google Scholar] [CrossRef]
  42. You, Y.J.; Kim, Y.; Nam, N.H.; Ahn, B.Z. Synthesis and cytotoxic activity of A-ring modified betulinic acid derivatives. Bioorg. Med. Chem. Lett. 2003, 13, 3137–3140. [Google Scholar] [CrossRef]
  43. Coulibaly, W.K.; Paquin, L.; Benie, A.; Bekro, Y.A.; Guevel, R.L.; Ravache, M.; Corlu, A.; Bazureau, J.P. Prospective study directed to the synthesis of unsymmetrical linked bis-5-arylidene rhodanine derivatives via “one-pot two steps” reactions under microwave irradiation with their antitumor activity. Med. Chem. Res. 2015, 24, 1653–1661. [Google Scholar] [CrossRef]
  44. Feichtinger, K.; Zapf, C.; Sings, H.L.; Goodman, M. Diprotected triflyl-guanidines: A new class of guanidinylation reagents. J. Org. Chem. 1998, 63, 3804–3805. [Google Scholar] [CrossRef]
  45. Kommera, H.; Kaluderovic, G.N.; Kalbitz, J.; Dräger, B.; Paschke, R. Small structural changes of pentacycliclupane type triterpenoid derivatives lead to significant differences in their anticancer properties. Eur. J. Med. Chem. 2010, 45, 3346–3353. [Google Scholar] [CrossRef] [PubMed]
  46. Willmann, M.; Wacheck, V.; Buckley, J.; Nagy, K.; Thalhammer, J.; Paschke, R.; Triche, T.; Jansen, B.; Selzer, E. Characterization of NVX-207, a novel betulinic acid-derived anti-cancer compound. Eur. J. Clin. Investig. 2009, 39, 384–394. [Google Scholar] [CrossRef] [PubMed]
  47. Bache, M.; Bernhardt, S.; Passin, S.; Wichmann, H.; Hein, A.; Zschornak, M.; Kappler, M.; Taubert, H.; Paschke, R.; Vordermark, D. Betulinic acid derivatives NVX-207 and B10 for treatment of glioblastoma—An in vitro study of cytotoxicity and radiosensitization. Int. J. Mol. Sci. 2014, 15, 19777–19790. [Google Scholar] [CrossRef] [PubMed]
  48. Liebscher, G.; Vanchangiri, K.; Mueller, T.; Feige, K.; Cavalleri, J.M.; Paschke, R. In vitro anticancer activity of betulinic acid and derivatives thereof on equine melanoma cell lines from grey horses and in vivo safety assessment of the compound NVX-207 in two horses. Chem. Biol. Interact. 2016, 246, 20–29. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of all compounds are available from the authors.
Figure 1. Betulinic, ursolic and oleanolic acids.
Figure 1. Betulinic, ursolic and oleanolic acids.
Molecules 23 03000 g001
Scheme 1. Synthesis of target compounds 9b12b and 14. Reagents and conditions: (a) 1. (COCl)2, CH2Cl2; 2. 1,2-diaminoethane or 1,4-diaminobutane or tris(2-aminoethyl)amine or N-tert-butoxy-carbonyl-1,4-bis(3-aminopropyl)piperazine, Et3N, CH2Cl2, r.t.; (b) AcCl, THF, Py, DMAP, r.t.; (c) 10% TFA, CH2Cl2, 3 h, r.t.; (d) 1,3-di-Boc-2-(trifluoromethylsulfonyl)guanidine, Et3N, CH2Cl2; (e) 50% TFA, CH2Cl2, 2–4 h, r.t.; (f) 5 M HCl, MeOH; (g) 4 N NaOH, MeOH, THF.
Scheme 1. Synthesis of target compounds 9b12b and 14. Reagents and conditions: (a) 1. (COCl)2, CH2Cl2; 2. 1,2-diaminoethane or 1,4-diaminobutane or tris(2-aminoethyl)amine or N-tert-butoxy-carbonyl-1,4-bis(3-aminopropyl)piperazine, Et3N, CH2Cl2, r.t.; (b) AcCl, THF, Py, DMAP, r.t.; (c) 10% TFA, CH2Cl2, 3 h, r.t.; (d) 1,3-di-Boc-2-(trifluoromethylsulfonyl)guanidine, Et3N, CH2Cl2; (e) 50% TFA, CH2Cl2, 2–4 h, r.t.; (f) 5 M HCl, MeOH; (g) 4 N NaOH, MeOH, THF.
Molecules 23 03000 sch001
Scheme 2. Synthesis of compounds 15ac, 18ac, 20ac. Reagents and conditions: (a) 1. (COCl)2, CH2Cl2; 2. tris(hydroxymethyl)aminomethane, DMAP, Py, CH2Cl2; (b) 1,3-di-Boc-2-(trifluoromethyl-sulfonyl)guanidine, Et3N, CHCl3, reflux; (c) 50% TFA, CH2Cl2, 2–4 h, r.t.; (d) 5M HCl, MeOH.
Scheme 2. Synthesis of compounds 15ac, 18ac, 20ac. Reagents and conditions: (a) 1. (COCl)2, CH2Cl2; 2. tris(hydroxymethyl)aminomethane, DMAP, Py, CH2Cl2; (b) 1,3-di-Boc-2-(trifluoromethyl-sulfonyl)guanidine, Et3N, CHCl3, reflux; (c) 50% TFA, CH2Cl2, 2–4 h, r.t.; (d) 5M HCl, MeOH.
Molecules 23 03000 sch002
Figure 2. AnnexinV/7-AAD staining upon induction of apoptosis in Jurkat cells. Cells were treated with compounds at their IC50 concentration for 24 and 48 h. Then, the cells were harvested, stained with Annexin V/7-AAD and analyzed by flow cytometry. The experiments were performed three times, and the results of the representative experiments are shown. The first cytogram represents an untreated cell sample (A); after incubation with dihydrobetulinic acid for 24 h (B) and for 48 h (C); after incubation with compound 15 for 24 h (D) and for 48 h (E); after incubation with compound 15c for 24 h (F) and for 48 h (G); after incubation with compound 18c for 24 h (H) and for 48 h (I); after incubation with compound 20c for 24 h (J) and for 48 h (K). Q7-1, necrotic cells; Q7-2, late apoptotic cells; Q7-3, living cells; Q7-4, early apoptotic cells.
Figure 2. AnnexinV/7-AAD staining upon induction of apoptosis in Jurkat cells. Cells were treated with compounds at their IC50 concentration for 24 and 48 h. Then, the cells were harvested, stained with Annexin V/7-AAD and analyzed by flow cytometry. The experiments were performed three times, and the results of the representative experiments are shown. The first cytogram represents an untreated cell sample (A); after incubation with dihydrobetulinic acid for 24 h (B) and for 48 h (C); after incubation with compound 15 for 24 h (D) and for 48 h (E); after incubation with compound 15c for 24 h (F) and for 48 h (G); after incubation with compound 18c for 24 h (H) and for 48 h (I); after incubation with compound 20c for 24 h (J) and for 48 h (K). Q7-1, necrotic cells; Q7-2, late apoptotic cells; Q7-3, living cells; Q7-4, early apoptotic cells.
Molecules 23 03000 g002
Figure 3. Cell cycle analysis in Jurkat cells. Jurkat cells treated with tested compounds at the IC50 concentration for 24 and 48 h. The cells were trypsinized, harvested and washed three times with ice-PBS for PI-stained DNA content detected by flow cytometry. The experiments were performed three times, and the results of the representative experiments are shown. The first cytogram represents an untreated cell sample (A); after incubation with dihydrobetulinic acid for 48 h (B); after incubation with compound 15 for 24 h (C) and for 48 h (D); after incubation with compound 15c for 24 h (E) and for 48 h (F); after incubation with compound 18c for 24 h (G) and for 48 h (H); after incubation with compound 20c for 24 h (I) and for 48 h (J).
Figure 3. Cell cycle analysis in Jurkat cells. Jurkat cells treated with tested compounds at the IC50 concentration for 24 and 48 h. The cells were trypsinized, harvested and washed three times with ice-PBS for PI-stained DNA content detected by flow cytometry. The experiments were performed three times, and the results of the representative experiments are shown. The first cytogram represents an untreated cell sample (A); after incubation with dihydrobetulinic acid for 48 h (B); after incubation with compound 15 for 24 h (C) and for 48 h (D); after incubation with compound 15c for 24 h (E) and for 48 h (F); after incubation with compound 18c for 24 h (G) and for 48 h (H); after incubation with compound 20c for 24 h (I) and for 48 h (J).
Molecules 23 03000 g003
Table 1. Cytotoxicity of dihydrobetulinic-, ursolic- and oleanolic acids and compounds 46, 8a, 9b12b, 12a, 13a, 14, 15, 15c, 18b,c, and 20b,c against Jurkat, K562, Hek293, HeLa, U937, and normal fibroblast cells.
Table 1. Cytotoxicity of dihydrobetulinic-, ursolic- and oleanolic acids and compounds 46, 8a, 9b12b, 12a, 13a, 14, 15, 15c, 18b,c, and 20b,c against Jurkat, K562, Hek293, HeLa, U937, and normal fibroblast cells.
CompoundIC50 (µmol/L) a
JurkatK562U937HEK293HeLaFibroblasts
Dihydro BA59 ± 0.3144 ± 0.2439 ± 0.38128 ± 0.32132 ± 0.47517 ± 0.26
UA23 ± 0.3468 ± 0.1117 ± 0.1296 ± 0.2288 ± 0.35324 ± 0.16
OA271 ± 0.19235 ± 0.24186 ± 0.18247 ± 0.27258 ± 0.31694 ± 0.17
41.4 ± 0.411.3 ± 0.491.7 ± 0.362.4 ± 0.352.8 ± 0.283.4 ± 0.19
52.3 ± 0.112.8 ± 0.231.9 ± 0.344.8 ± 0.253.9 ± 0.415.3 ± 0.27
67.7 ± 0.311.8 ± 0.124.5 ± 0.436.7 ± 0.217.1 ± 0.188.3 ± 0.29
8a4.5 ± 0.293.0 ± 0.198.2 ± 0.135.9 ± 0.146.1 ± 0.387.3 ± 0.43
9b21 ± 0.2457 ± 0.3614 ± 0.1692 ± 0.3481 ± 0.19179 ± 0.43
10b67 ± 0.2198 ± 0.3582 ± 0.14128 ± 0.35152 ± 0.49354 ± 0.46
11b16 ± 0.1855 ± 0.3863 ± 0.2998 ± 0.27106 ± 0.51117 ± 0.38
12a22 ± 0.2189 ± 0.1828 ± 0.25112 ± 0.19101 ± 0.22149 ± 0.43
12b14 ± 0.3752 ± 0.2613 ± 0.0668 ± 0.2972 ± 0.34106 ± 0.23
13a24 ± 0.2711 ± 0.3318 ± 0.2174 ± 0.1579 ± 0.29104 ± 0.26
1417 ± 0.3436 ± 0.4220 ± 0.1259 ± 0.2162 ± 0.2496 ± 0.17
153.3 ± 0.112.1 ± 0.136.6 ± 0.2317 ± 0.2519 ± 0.4931 ± 0.29
15c3.1 ± 0.412.3 ± 0.3415 ± 0.2316 ± 0.2714 ± 0.3127 ± 0.14
18b6.8 ± 0.3240 ± 0.292.1 ± 0.4113 ± 0.1911 ± 0.2747 ± 0.15
18c3.8 ± 0.2311 ± 0.185.3 ± 0.2914 ± 0.3412 ± 0.2951 ± 0.26
20b6.7 ± 0.1520 ± 0.146.1 ± 0.4312 ± 0.2313 ± 0.4149 ± 0.25
20c7.6 ± 0.3313 ± 0.456.8 ± 0.1119 ± 0.1717 ± 0.3254 ± 0.12
a IC50 (µM) is the half maximal inhibitory concentration for viable cells. Each IC50 (mean ± SE) has been derived from at least three experiments in duplicate.

Share and Cite

MDPI and ACS Style

Spivak, A.; Khalitova, R.; Nedopekina, D.; Dzhemileva, L.; Yunusbaeva, M.; Odinokov, V.; D’yakonov, V.; Dzhemilev, U. Synthesis and Evaluation of Anticancer Activities of Novel C-28 Guanidine-Functionalized Triterpene Acid Derivatives. Molecules 2018, 23, 3000. https://doi.org/10.3390/molecules23113000

AMA Style

Spivak A, Khalitova R, Nedopekina D, Dzhemileva L, Yunusbaeva M, Odinokov V, D’yakonov V, Dzhemilev U. Synthesis and Evaluation of Anticancer Activities of Novel C-28 Guanidine-Functionalized Triterpene Acid Derivatives. Molecules. 2018; 23(11):3000. https://doi.org/10.3390/molecules23113000

Chicago/Turabian Style

Spivak, Anna, Rezeda Khalitova, Darya Nedopekina, Lilya Dzhemileva, Milyausha Yunusbaeva, Victor Odinokov, Vladimir D’yakonov, and Usein Dzhemilev. 2018. "Synthesis and Evaluation of Anticancer Activities of Novel C-28 Guanidine-Functionalized Triterpene Acid Derivatives" Molecules 23, no. 11: 3000. https://doi.org/10.3390/molecules23113000

Article Metrics

Back to TopTop