You are on page 1of 10

Article

pubs.acs.org/jmc

Design, Synthesis, and Biological Evaluation of Hexacyclic


Tetracyclines as Potent, Broad Spectrum Antibacterial Agents
Cuixiang Sun,† Diana K. Hunt,† Chi-Li Chen,† Yonghong Deng,† Minsheng He,† Roger B. Clark,†
Corey Fyfe,‡ Trudy H. Grossman,‡ Joyce A. Sutcliffe,‡ and Xiao-Yi Xiao*,†

Discovery Chemistry, ‡Microbiology, Tetraphase Pharmaceuticals, 480 Arsenal Street, Watertown, Massachusetts 02472, United
States
*
S Supporting Information

ABSTRACT: A series of novel hexacyclic tetracycline analogues (“hexacy-


clines”) was designed, synthesized, and evaluated for antibacterial activity
against a wide range of clinically important bacteria isolates, including
multidrug-resistant, Gram-negative pathogens. Valuable structure−activity
relationships were identified, and several hexacyclines displayed potent, broad
spectrum antibacterial activity, including promising anti-Pseudomonas aeruginosa
activity in vitro and in vivo.

■ INTRODUCTION
New, potent antibacterial agents are in urgent need to combat
positions from C4 to C9, including various substitutions and
additional fused ring systems. These chemical modifications
often lead to modulations of tetracyclines’ chemical−physical
life threatening infections caused by multidrug-resistant
properties, ribosomal binding, as well as the ability to overcome
(MDR), Gram-negative and Gram-positive organisms such as
active drug efflux. We have therefore designed a new hexacyclic
carbapenem-resistant Enterobacteriaceae (CRE)1 and methicil-
tetracycline scaffold (1, Figure 1) that consists of a bicyclic EF-
lin-resistant Staphylococcus aureus (MRSA).2 Tetracyclines are a
proven class of broad spectrum antibiotics first discovered in
the mid-1940s.3 While revolutionary at the time, seven decades
of widespread use has largely rendered legacy tetracyclines
ineffective due to progressing tetracycline resistance. 4a
Renewed interest in the class, stemming from advances in
chemical synthesis, has resulted in marked improvements
against tetracycline-specific resistance, leading to the most
recently approved tetracycline, tigecycline,5 in 2005, as well as
newer generations of tetracycline antibiotics currently in late
stage development such as eravacycline.6,7 Figure 1. A hexacyclic tetracycline scaffold.
Two major bacterial resistance mechanisms4b,c for tetracy-
clines have been identified: (1) tetracycline-specific efflux ring appended to the original tetracycline D-ring. A series of
pumps, including tet(A)−tet(D) and tet(K)−tet(L), which are hexacyclic tetracycline analogues (“hexacyclines”) with struc-
frequently found in both Gram-positive and Gram-negative tural variations at a number of positions was synthesized using
pathogens, and (2) ribosomal protection, including tet(M)− our fully synthetic tetracycline strategy.6,8b,10 These hexacy-
tet(O), which are more commonly seen in Gram-positive clines were evaluated for antibacterial activity against a broad
bacteria. A number of semisynthetic tetracyclines, including range of pathogens, especially activities against tetracycline
resistant, Gram-negative bacteria.


doxycycline, minocycline, and tigecycline, have been developed
to overcome bacterial resistance. However, the exploitation of
the tetracycline scaffold has been impeded by the limited RESULTS AND DISCUSSION
chemical methods available for semisynthetic tetracyclines until Chemistry. Using our tetracycline total synthesis ap-
the recent development and application of the tetracycline total proach,6,8b,10 a series of 7-fluorohexacyclines with a wide
synthesis platform, which enables the creation and study of range of substitutions at the 8-position nitrogen (N8) was
previously difficult-to-access, fully synthetic tetracycline ana- prepared from a tricyclic DEF-ring precursor 6 and an AB-ring
logues.6,8−10 enone 7.10,11 As shown in Scheme 1, aniline 29b was treated
Prior structure−activity relationship (SAR) studies5,6,8a,9
from our laboratory and other laboratories indicated that the Received: February 13, 2015
tetracycline scaffold can tolerate structural modifications on

© XXXX American Chemical Society A DOI: 10.1021/acs.jmedchem.5b00262


J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

Scheme 1. Synthesis of 7-Fluorohexacycline Analoguesa

a
Reagents: (a) Br2, NaOAc, HOAc; (b) LHMDS, THF, then Boc2O, and then allyl bromide; (c) n-BuLi, THF, then DMF; (d) N-allyl glycine,
DMF; (e) LDA, TMEDA, THF, then enone 7; (f) Pd(PPh3)4, N,N′-dimethylbarbituric acid, CH2Cl2; (g) (i) N-substitution (see Experimental
Section), (ii) 48% aq HF, THF, (iii) 10% Pd-C, HCl, CH3OH.

with bromine in the presence of sodium acetate to give the and iodine to give compound 13, which was benzylated under
bromide intermediate 3 in 85% yield. Full protection of the standard conditions to afford intermediate 14. Compound 14
aniline group by treatment with lithium bis(trimethylsilyl)- was then reacted at 80 °C with BocNH2 in the presence of
amide (LHMDS) and Boc2O, followed by allyl bromide yielded Pd(OAc)2, Xantphos,14 and cesium carbonate to yield
compound 4 (88% yield). Compound 4 was formylated by intermediate 15 in 28% yield over three steps. Aryl bromide
treatment with n-BuLi at −100 °C in THF followed by DMF 15 was treated with PhLi and n-BuLi followed by DMF to
addition to give aldehyde 5 in 94% isolated yield. Cyclo- afford aldehyde 16 (37% yield), which was allylated with allyl
addition12 of aldehyde 5 with N-allyl glycine in DMF at 80 °C bromide and NaH to give compound 17 in 82% yield.
afforded the desired tricyclic DEF-ring precursor 6 in 71% yield Cycloaddition12 of compound 17 with a range of cyclic and
with a cis-configuration between the fused 5- and 6-membered acyclic N-alkylated amino acids under the conditions described
heterocyclic rings (the corresponding trans isomer was not above gave the desired DEF-ring precursors 18 in good yields.
detected). Michael−Dieckmann annulation10 of 6 with the AB- Michael−Dieckmann annulation10 of 18 with enone 7, followed
ring enone 7 by treatment with lithium diisopropylamide by desilylation and hydrogenation, gave the desired 7-
(LDA) and N,N,N′,N′-tetramethylethylenediamine (TMEDA) trifluoromethoxy hexacycline analogues 20a−20f in yields
at −78 °C in THF gave the fully protected intermediate 8 in similar to those in Scheme 1 after reverse phase HPLC
98% yield as a mixture of two diastereomers. The allyl group on purification (Table 2).
N8 was removed by standard deallylation conditions to afford Biology. The novel hexacyclines were evaluated for in vitro
intermediate 9 (the two diastereomers were separated by antibacterial activity in minimal inhibitory concentration (MIC)
preparative HPLC, 67% combined yield). The two diaster- assays against a panel of tetracycline-susceptible and tetracy-
eomers of secondary amine 9 were then separately alkylated by cline-resistant, Gram-positive and Gram-negative bacterial
reductive alkylation with a variety of aldehydes to yield the N8 strains (Tables 1 and 2). All isolates in the panel were
substituted intermediates, and these were deprotected by multidrug-resistant (MDR, i.e., resistant to ≥3 different classes
aqueous HF treatment followed by hydrogenation as previously of antibiotics) except S. aureus SA101, Escherichia coli EC107,
described6 to afford the desired hexacycline analogues 10a− and S. aureus SA158, a tet (K) tetracycline-resistant strain and
10h in reasonable yields (20−60%) after reverse phase HPLC otherwise drug susceptible. MIC data of representative
purification (Table 1). hexacycline analogues are listed in Tables 1 and 2. As
Similarly, several analogues with a methyl substituent at the mentioned above, most of the final compounds were isolated
bridgehead carbon C10a were also prepared using 3-bromo-2- as two separate cis-diastereomers. Although it is known that the
methylpropene instead of ally bromide in Scheme 1, step b “north-west” portion of tetracyclines is not involved in
(11a, 11b, and 11c, Table 1). ribosomal binding, the two diastereomers displayed consid-
Subsequently, to explore the effect of other C7 substitutions, erably different MIC values across the entire panel of bacterial
a series of 7-trifluoromethoxy hexacycline analogues was strains, as exemplified by the diastereomer pair 10b and 10c in
prepared according to Scheme 2. In addition, a range of cyclic Table 1. For brevity, only the relatively more active
and acyclic amino acids was also incorporated in the diastereomers from each pair are listed in Tables 1 and 2,
cycloaddition step to explore structural variations on the F- except for compounds 10a, 10h, and 20a, which were isolated
ring. Thus, phenol 1213 was iodinated by treatment with NaH as mixtures of the two diastereomers (∼1:1).
B DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

Table 1. In Vitro Antibacterial Activity of 7-Fluorohexacycline Analogues

a
Single diastereomer unless otherwise noted. bStrains were obtained from the American Type Culture Collection (ATCC, Manassas, VA) unless
otherwise noted. The first six strains from the left are Gram-positive strains. The last nine strains are Gram-negative strains. Strains with “tet(A)”,
“tet(B)”, “tet(K)”, or “tet(M)” genes noted underneath are tetracycline-resistant strains. SA, Staphylococcus aureus; EFs, Enterococcus faecalis; EFm,
Enterococcus faecium; SP, Streptococcus pneumoniae; EC, Escherichia coli; KP, Klebsiella pneumoniae (KP457 contains a blacTX‑M‑15 extended spectrum
β-lactamase gene); PM, Proteus mirabilis; PA, Pseudomonas aeruginosa; ECl, Enterobacter cloacae; AB, Acinetobacter baumannii; SM, Stenotrophomonas
maltophilia; BC, Burkholderia cenocepacia. cObtained from Micromyx (Kalamazoo, MI). dObtained from Marilyn Roberts’ laboratory at the
University of Washington. eObtained from Eurofins-Medinet, Chantilly, VA. fObtained from R. K. Ernst’s Laboratories at University of Maryland. gA
mixture of two diastereomers (∼1:1).

When the pyrrolidine nitrogen (N8) was not substituted, the amount of increase in lipophilicity (10d and 11c). The
hexacycline analogues had only modest antibacterial activity, as presence of additional polar groups, such as hydroxyl, amino,
shown by MIC data of compounds 10a, 11a, and 20a. and amide groups, on the nitrogen substituent drastically
However, when the nitrogen was substituted with a methyl decreased the compounds’ antibacterial activities (10e−10h).
group, the potency was dramatically improved by 2- to >32-fold Interestingly, the potency of the difluoroethylaminoethyl
against most strains in the panel except PM385 (10c vs 10a, substituted analogue 10g was decreased by a much lesser
20b vs 20a). Further increases in lipophilicity of the nitrogen degree, especially against Gram-positive pathogens, probably
substituent tended to decrease potency, especially against due to the decreased basicity and/or polarity of the
Gram-negative strains, by varying degrees depending on the difluoroethylamino group. Alkyl substitution at the C10a
C DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

Scheme 2. Synthesis of 7-Trifluoromethoxyhexacycline Analoguesa

a
Reagents: (a) NaH, toluene, then I2; (b) BnBr, K2CO3, DMF; (c) BocNH2, Pd(OAc)2, Xantphos, Cs2CO3, 1,4-dioxane; (d) PhLi, n-BuLi, THF,
then DMF; (e) NaH, DMF, then allyl bromide; (f) N-alkylated amino acid, DMF; (g) LDA, TMEDA, THF, then enone 7; (h) (i) 48% aq HF, THF,
(ii) 10% Pd-C, HCl, CH3OH.

Table 2. In Vitro Antibacterial Activity of 7-Trifluoromethoxyhexacycline Analogues

a
Single diastereomers unless otherwise noted. bSee footnotes b-f for Table 1. cA mixture of two diastereomers (∼1:1).

position decreased potency against most strains in the panel, as (20f) or lipophilicity (either by alkyl substitution on the F-
shown by the 10a-methyl substituted analogues 11a, 11b, and ring (20c) or by incorporation of a seventh ring (G-ring, 20d,
11c. 20e)) decreased potency. Interestingly, compared to the 7-
Similar general SAR trends were also observed for the 7- fluoro analogue 10c, the corresponding 7-trifluoromethoxy
trifluoromethoxy series (Table 2). Thus, a methyl group on the analogue 20b was more potent against most Gram-positive
pyrrolidine nitrogen (N8) was the optimum substitution for strains (≥4-fold) as well as some of the Gram-negative strains
antibacterial activity (compound 20b). Additional polarity (Escherichia coli strains EC107 and EC155, and Stenotropho-
D DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

Figure 2. In vivo efficacy of compound 10c in (A), mouse lung infection model challenged with P. aeruginosa PA1145; (B) mouse thigh infection
model challenged with P. aeruginosa PA694.

monas maltophilia SM256) but 2−4-fold less active against mouse infection models challenged with P. aeruginosa
Proteus mirabilis PM385, Pseudomonas aeruginosa PA555, and demonstrated that compound 10c was equally or more
Acinetobacter baumannii AB250. efficacious than various comparator compounds at comparable
Overall, compounds 10c and 20b displayed high in vitro dose levels. These data support the further optimization of the
potency against a broad range of MDR Gram-positive and hexacycline scaffold for the discovery and development of new
Gram-negative pathogens. Particularly, compound 10c also tetracycline antibiotics against a broad range of pathogens,
demonstrated promising in vitro activity against P. aeruginosa including MDR Gram-negative bacteria.


PA555 with an MIC of 4 μg/mL, which is among the lowest
MIC values in the tetracycline class. Ribosomal inhibition was EXPERIMENTAL SECTION
confirmed by an in vitro P. aeruginosa coupled transcription/ Chemistry. All commercially available reagents and solvents,
translation assay,15 in which compound 10c was 10-fold more including anhydrous solvents, were used without further purification.
potent than tetracycline with an IC50 value of 0.21 μM. All reactions under dry conditions were performed under nitrogen
Compound 10c was further tested for in vivo efficacy in two atmospheres. 1H and 13C NMR (nuclear magnetic resonance) spectra
mouse infection models challenged with P. aeruginosa. In a were recorded at room temperature (25 °C) on a 400 MHz JEOL
mouse lung infection model challenged with P. aeruginosa ECX-400 spectrometer. CDCl3 (for intermediates) or CD3OD (for
PA1145 (Figure 2A), compound 10c (MIC = 4 μg/mL) final compounds) were used as the solvents. Chemical shifts (δ) are
demonstrated a 4-log colony-forming unit (CFU) reduction in expressed in parts per million (ppm). The signals of the deuterated
bacterial burden when dosed intravenously (IV) at 40 mg/kg, solvents were used as the internal standards. Thin-layer chromatog-
raphy (TLC) analysis was performed on Merck Silica Gel 60 F254 and
twice daily. Comparator compounds meropenem (MIC = 4 visualized under UV light. Flash chromatography was performed on a
μg/mL) and amikacin (MIC = 1 μg/mL) showed bacterial Biotage Isolera One using SNAP cartridges (KP-Sil). Purity of tested
burden reductions of about 2−2.5 log CFU when administered compounds was determined to be ≥95% by reverse phase analytical
at the same doses. In the mouse thigh infection model HPLC/MS analysis (high-performance liquid chromatography/mass
challenged with P. aeruginosa PA694 (Figure 2B), compound spectrometry) performed on a Waters Alliance system (column,
10c (MIC = 8 μg/mL) showed promising dose-proportional SunFire C18, 5 μm, 4.6 mm × 50 mm; solvent A, water with 0.1%
efficacy when dosed at 1.5, 5, 15, and 40 mg/kg, IV, twice daily formic acid; solvent B, acetonitrile with 0.1% formic acid; MS detector,
and was equally efficacious at 40 mg/kg as comparator Waters 3100) unless otherwise noted. Reverse phase preparative
levofloxacin (MIC = 1 μg/mL) dosed IV at 5 mg/kg twice HPLC was performed on a Waters Autopurification system with mass-
directed fraction collection (for final compounds: column, Polymerx
daily (1.8 log CFU reduction from T = 0 and 4.5 log CFU RP-1 100A, 10 μm, 150 mm × 21.20 mm; flow rate, 20 mL/min;
reduction from T = 24 h). solvent A, water with 0.05 N HCl; solvent B, acetonitrile; for

■ CONCLUSIONS
A series of hexacycline analogues based on scaffold 1 was
intermediates: column, SunFire Prep C18 OBD, 5 μm, 19 mm × 50
mm; flow rate, 20 mL/min; solvent A, water with 0.1% formic acid;
solvent B, acetonitrile with 0.1% formic acid) unless otherwise noted.
Phenyl 3-Amino-2-(benzyloxy)-4-bromo-5-fluoro-6-methyl-
designed and synthesized with a range of substitutions at C7, benzoate (3). To a solution of phenyl 3-amino-2-(benzyloxy)-5-
N8, C9, and C10a, and their antibacterial activity was evaluated fluoro-6-methylbenzoate (2,9b 15.00 g, 42.69 mmol) and NaOAc
against a broad range of Gram-negative and Gram-positive (10.50 g, 128.07 mmol, 3 equiv) in HOAc (100 mL) was added a
pathogens, including multidrug-resistant and tetracycline- solution of Br2 (2.20 mL, 42.69 mmol, 1 equiv) in HOAc (10 mL)
resistant bacterial strains. Analogues 10c, a 7-fluorohexacycline dropwise via a syringe at 17 → 19 °C while cooled in a cold water
bath. After stirring at 20 °C for 20 min, more Br2 (66 μL) in HOAc (1
analogue, and 20b, a 7-trifluoromethoxyhexacycline analogue, mL) was added. After stirring for 5 min, the reaction was poured into
demonstrated potent, broad spectrum in vitro antibacterial ice/water. The resulting mixture was extracted with EtOAc (600 mL).
activity. Compound 10c also displayed promising anti-P. The organic phase was separated, washed with 10% aqueous Na2S2O3
aeruginosa potency with MIC values ranging from 4 to 8 μg/ solution, water, saturated aqueous sodium bicarbonate, and brine. The
mL in this study. In vivo efficacy studies in thigh and lung organic phase was dried over sodium sulfate, filtered, and concentrated

E DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

under reduced pressure. Flash chromatography on silica gel using 5% (m, 1 H), 3.09−2.99 (m, 1 H), 2.85−2.54 (m, 2 H), 2.34 (s, 3 H),
→ 6% EtOAc/hexanes yielded the desired product 3 as a thick pale- 2.20−2.09 (m, 2 H), 2.06−1.94 (m, 1 H), 2.45−2.22 (m, 10 H). MS
yellow oil (15.59 g, 85%). 1H NMR (400 MHz, CDCl3) δ 7.44−7.35 (ESI) m/z calcd for C34H36FN2O5 [M − H]− 571.26; found 571.14.
(m, 7 H), 7.28−7.25 (m, 1 H), 7.15−7.13 (m, 2 H), 5.01 (s, 2 H), 4.27 tert-Butyl (1R,19S,26S,27S)-14,22-Bis(benzyloxy)-19-[(tert-
(br s, 2 H), 2.32 (d, J = 2.4 Hz, 3 H). MS (ESI) m/z calcd for butyldimethylsilyl)oxy]-26-(dimethylamino)-4-fluoro-18-hy-
C21H18BrFNO3 [M + H]+ 430.05, 432.05; found 429.94, 431.92. droxy-16,20-dioxo-7-(prop-2-en-1-yl)-24-oxa-7,12,23-
Phenyl 2-(Benzyloxy)-4-bromo-3-{[(tert-butoxy)carbonyl]- triazaheptacyclo[15.11.0.03,15.05,13.06,10.019,27.021,25]octacosa-
(prop-2-en-1-yl)amino}-5-fluoro-6-methylbenzoate (4). To a 3,5(13),14,17,21(25),22-hexaene-12-carboxylate (8). LDA (1.3
solution of aniline 3 (908 mg, 2.11 mmol) in anhydrous THF (8 equiv) was prepared at −40 °C from n-butyllithium (1.6 M solution in
mL) was added a solution of LHMDS in THF (4.43 mL, 1.0 M, 4.43 hexanes, 845 μL, 1.35 mmol) and diisopropylamine (216 μL, 1.50
mmol, 2.1 equiv) dropwise over 7 min while maintaining the internal mmol) in THF (10 mL). The LDA solution was cooled to −78 °C
temperature below −70 °C (dry ice−acetone bath). The reaction (dry ice−acetone bath) and TMEDA (217 μL, 1.45 mmol, 1.4 equiv)
solution was stirred at −78 °C for 15 min. A solution of Boc2O (484 was added, followed by dropwise addition of a solution of compound 6
mg, 2.22 mmol, 1.05 equiv) in THF (1 mL) was added dropwise while (655 mg, 1.14 mmol, 1.1 equiv) in THF (4.5 mL) with a 1.5 mL rinse.
maintaining the internal temperature below −71 °C. The reaction was This resulted in a deep-red solution. After 30 min, the reaction was
stirred at −78 °C for 30 min, and then the dry ice was removed from cooled to −100 °C (liquid nitrogen−ethanol bath), and a solution of
the cold bath. The reaction was then warmed up to −50 °C, and allyl enone 7 (503 mg, 1.04 mmol) in THF (3.5 mL) was added dropwise,
bromide (0.20 mL, 2.32 mmol, 1.1 equiv) was added. The reaction was maintaining the internal temperature below −95 °C. After complete
warmed up to rt in 20 min and heated at 50 °C for 3 h. More allyl addition, the reaction was allowed to warm to −70 °C over 40 min,
bromide (0.20 mL, 2.32 mmol, 1.1 equiv) was added. The reaction was and a solution of LHMDS (1 M in THF, 1.04 mL, 1.04 mmol, 1
heated at 50 °C for 2 h, cooled to rt, and diluted with EtOAc (40 mL). equiv) was added. The reaction was allowed to warm to −20 °C over 1
The reaction solution was washed with saturated aqueous NH4Cl (2 × h and quenched by the addition of NH4Cl (saturated aqueous
30 mL) and brine (30 mL), dried over sodium sulfate, filtered, and solution, 30 mL). The reaction mixture was extracted with EtOAc (2 ×
concentrated under reduced pressure. Flash chromatography on silica 100 mL). The combined extracts were dried over Na2SO4, filtered, and
gel using 2% → 5% EtOAc/hexanes yielded the desired product 4 concentrated under reduced pressure. The resulting foam was purified
(1.06 g, 88%, ∼3:1 rotamers). 1H NMR (400 MHz, CDCl3) δ 7.39− via column chromatography (Biotage 100 g column, 4−60% EtOAc in
7.34 (m, 7 H), 7.29−7.25 (m, 1 H), 7.04−7.00 (m, 2 H), 6.00−5.90 hexanes), yielding 983 mg (98%, mixture of diastereomers) of the
(m, 1 H), 5.09−5.04 (m, 1 H), 5.03−5.00 (m, 2.25 H), 4.92 (d, J = desired product 8 as a yellow solid. MS (ESI) m/z calcd for
10.4 Hz, 0.75 H), 4.50 (dd, J = 6.1, 14.6 Hz, 0.75 H), 4.24 (dd, J = 6.1, C54H64FN4O9 [M − H]− 959.44; found 959.28.
15.3 Hz, 0.25 H), 4.04−3.97 (m, 1 H), 2.42 (d, J = 2.4 Hz, 2.25 H), tert-Butyl (1R,19S,26S,27S)-14,22-Bis(benzyloxy)-19-[(tert-
butyldimethylsilyl)oxy]-26-(dimethylamino)-4-fluoro-18-hy-
2.40 (d, J = 2.4 Hz, 0.75 H), 1.54 (s, 2.25 H), 1.44 (s, 6.75 H). MS droxy-16,20-dioxo-24-oxa-7,12,23-triazaheptacyclo-
(ESI) m/z calcd for C29H29BrFNO5Na [M + Na]+ 592.11, 594.11; [15.11.0.03,15.05,1306,10.019,27.021,25]octacosa-3,5(13),14,17,21-
found 591.99, 593.98. (25),22-hexaene-12-carboxylate (9a and 9b). A solution of
Phenyl 2-(Benzyloxy)-3-{[(tert-butoxy)carbonyl](prop-2-en- compound 8 (945 mg, 0.983 mmol), N,N′-dimethylbarbituric acid
1-yl)amino}-5-fluoro-4-formyl-6-methylbenzoate (5). To a (1.53 g, 9.80 mmol, 10 equiv), and tetrakis(triphenylphosphine)-
solution of bromide 4 (1.06 g, 1.86 mmol) in anhydrous THF (30 palladium(0) (77.5 mg, 0.067 mmol, 0.068 equiv) in CH2Cl2 (10 mL)
mL) was added a solution of n-BuLi in hexanes (1.16 mL, 1.6 M, 1.86 was degassed by bubbling nitrogen for 5 min. The reaction flask was
mmol, 1 equiv) dropwise while maintaining the internal temperature fitted with a nitrogen-flushed water-cooled reflux condenser, and the
below −100 °C (liquid nitrogen−ethanol bath). After stirring for 3 reaction was heated to 35 °C in an oil bath. After 4.5 h, the reaction
min, a solution of DMF (0.22 mL, 2.79 mmol, 1.5 equiv) in THF (1 was cooled to rt and saturated aqueous NaHCO3 (20 mL) was added
mL) was added dropwise while maintaining the internal temperature slowly over 5 min, followed by the addition of pH 7 phosphate buffer
below −100 °C. The reaction solution was then allowed to warm up to (30 mL). The aqueous layer was extracted with CH2Cl2 (2 × 75 mL),
−78 °C (dry ice−acetone bath) and stirred at that temperature for 35 and the combined organic layers were dried over Na2SO4, filtered, and
min. Saturated aqueous NH4Cl was added. The resulting mixture was concentrated under reduced pressure. The crude material was purified
allowed to warm up to rt and extracted with EtOAc (40 mL). The on a Waters Autopurification system equipped with a Sunfire Prep
organic phase was washed with brine, dried over sodium sulfate, C18 OBD column [5 μm, 19 mm × 50 mm; flow rate, 20 mL/min;
filtered, and concentrated under reduced pressure. Flash chromatog- solvent A, H2O with 0.1% HCO2H; solvent B, CH3CN with 0.1%
raphy on silica gel using 3% → 12% EtOAc/hexanes yielded the HCO2H; gradient, 30 → 80% B; mass-directed fraction collection] to
desired product 5 (0.91 g, 94%, ∼2:1 rotamers). 1H NMR (400 MHz, provide the two diastereomers as yellow solids (9a, 291 mg; 9b, 284
CDCl3) δ 10.22 (s, 1 H), 7.38−7.33 (m, 7 H), 7.28−7.24 (m, 1 H), mg; 9a + 9b, 31 mg; 67% total yield). 9a: 1H NMR (400 MHz,
7.02−6.99 (m, 2 H), 5.93−5.79 (m, 1 H), 5.04−4.96 (m, 3.35 H), 4.89 CDCl3) δ 16.11−15.68 (br m, 1 H), 7.52−7.45 (m, 2 H), 7.41−7.25
(d, J = 9.8 Hz, 0.65 H), 4.64 (dd, J = 5.5, 14.6 Hz, 0.65 H), 4.32 (dd, J (m, 8 H), 5.36 (s, 2 H), 4.90−4.65 (br m, 1 H), 4.58−4.38 (br m, 1
= 5.5, 14.6 Hz, 0.35 H), 3.97 (dd, J = 7.9, 14.6 Hz, 0.35 H), 3.90 (dd, J H), 4.28−4.04 (br m, 2 H), 3.94 (d, J = 10.38 Hz, 1 H), 3.25−3.14 (m,
= 8.5, 14.6 Hz, 0.65 H), 2.40 (d, J = 1.8 Hz, 2 H), 2.37 (d, J = 1.8 Hz, 1 1 H), 3.09−2.88 (m, 3 H), 2.62−2.29 (m, 10 H), 2.19−2.03 (m, 2 H),
H), 1.51 (s, 3 H), 1.36 (s, 6 H). MS (ESI) m/z calcd for 1.65−1.43 (br m, 1 H), 1.42−1.35 (m, 10 H), 0.82 (s, 9 H), 0.28 (s, 3
C30H30FNO6Na [M + Na]+ 542.20; found 542.1. H), 0.12 (s, 3 H); MS (ESI) m/z calcd for C51H62FN4O9Si [M + H]+
5-tert-Butyl 7-Phenyl 6-(Benzyloxy)-9-fluoro-8-methyl-1- 921.43, found 921.48. 9b: 1H NMR (400 MHz, CDCl3) δ 16.30−
(prop-2-en-1-yl)-1H,2H,3H,3aH,4H,5H,9bH-pyrrolo[3,2-c]- 15.79 (br m, 1 H), 7.52−7.44 (m, 2 H), 7.41−7.24 (m, 8 H), 5.36 (s, 2
quinoline-5,7-dicarboxylate (6). A solution of aldehyde 5 (570 mg, H), 5.06−4.51 (br m, 2 H), 4.38−4.06 (br m, 2 H), 3.96 (d, J = 10.38
1.09 mmol) and N-allylglycine (338 mg, 2.23 mmol, 2 equiv) in DMF Hz, 1 H), 3.26−3.14 (m, 1 H), 3.12−2.86 (m, 3 H), 2.60−2.32 (m, 10
(11 mL) was heated at 90 °C for 17 h. The reaction mixture was H), 2.18−2.02 (m, 2 H), 1.62−1.50 (br m, 1 H), 1.50−1.02 (br m, 10
cooled to rt, poured into water (125 mL), and extracted with EtOAc H), 0.81 (s, 9 H), 0.27 (s, 3 H), 0.12 (s, 3 H); MS (ESI) m/z calcd for
(100 mL, 2 × 50 mL). The combined organic layers were washed with C51H62FN4O9Si [M + H]+ 921.43, found 921.47.
water and brine, dried over anhydrous Na2SO4, filtered, and (5S,9S,10S,12R)-9-(Dimethylamino)-15-fluoro-4,5,8,25-tetra-
concentrated under reduced pressure. The resulting oil was purified hydroxy-18-(2-hydroxyethyl)-2,6-dioxo-18,23-diazahexacyclo-
via column chromatography (Biotage 100 g column, 4−40% EtOAc in [12.11.0.03,12.05,10.016,24,.017,21]pentacosa-1(25),3,7,14,16(24)-
hexanes gradient), yielding 444 mg (71%) of the desired product 6 as a pentaene-7-carboxamide (10e). To a solution of 9a (12.5 mg,
clear oil (cis). 1H NMR (400 MHz, CDCl3) δ 7.40−7.21 (m, 8 H), 0.0135 mmol) in 1,2-dichloroethane (300 μL) was added (tert-
7.08−7.01 (m, 2 H), 5.79−5.64 (m, 1 H), 5.16−5.08 (m, 1 H), 5.01− butyldimethylsilyloxy)acetaldehyde (90%, 14 μL, 0.068 mmol, 5
4.92 (m, 2 H), 4.85−4.75 (m, 1 H), 4.30−4.20 (m, 1 H), 3.80−3.77 equiv), acetic acid (3.9 μL, 0.068 mmol, 5 equiv), and sodium

F DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

triacetoxyborohydride (13 mg, 0.61 mmol, 5 equiv). After stirring at rt Hz, 1 H), 4.98−4.92 (m, 1 H), 4.11−3.92 (m, 2 H), 3.91−3.79 (m, 1
for 18 h, the reaction was quenched with saturated aqueous NaHCO3 H), 3.80−3.53 (m, 5 H), 3.51−3.39 (m, 2 H), 3.27−2.85 (m, 10 H),
(10 mL). The reaction mixture was extracted with EtOAc (2 × 20 2.83−2.67 (m, 1 H), 2.63−2.49 (m, 1 H), 2.26−2.04 (m, 3 H), 1.68−
mL). The combined organic layers were dried over Na2SO4, filtered, 1.53 (m, 1 H). MS (ESI) m/z calcd for C30H37F3N5O7 [M + H]+
and concentrated under reduced pressure to provide the N8 alkylated 636.27; found 636.19.
intermediate. MS (ESI) m/z calcd for C59H80FN4O10Si [M + H]+ (5S,9S,10S,12R)-9-(Dimethylamino)-15-fluoro-4,5,8,25-tetra-
1079.54; found 1079.23. Final deprotections using previously hydroxy-18-[2-(N-methylacetamido)ethyl]-2,6-dioxo-18,23-
described desilylation and hydrogenation procedures6 provided the diazahexacyclo[12.11.0.0 3,12 .0 5,10 .0 16,24 .0 17,21 ]pentacosa-1-
desired compound 10e as a yellow solid after reverse phase HPLC (25),3,7,14,16(24)-pentaene-7-carboxamide (10h). 1H NMR
purification (2.8 mg, HCl salt, 27% over three steps). 1H NMR (400 (400 MHz, CD3OD, HCl salt) δ 4.07 (s, 1 H), 3.92−3.69 (m, 3
MHz, CD3OD) δ 4.98−4.95 (m, 1 H), 4.07 (s, 1 H), 3.93−3.84 (m, 1 H), 3.68−3.62 (m, 1 H), 3.59−3.39 (m, 4 H), 3.14−2.91 (m, 13 H),
H), 3.80−3.36 (m, 6 H), 3.20−2.64 (m, 11 H), 2.54−2.40 (m, 1 H), 2.88−2.61 (m, 1 H), 2.56−2.43 (m, 1 H), 2.29−2.02 (m, 6 H), 1.67−
2.30−2.08 (m, 3 H), 1.68−1.55 (m, 1 H). MS (ESI) m/z calcd for 1.55 (m, 1 H). MS (ESI) m/z calcd for C31H39FN5O8 [M + H]+
C28H34FN4O8 [M + H]+ 573.24; found 573.12. 628.27; found 628.11.
(5S,9S,10S,12R)-9-(Dimethylamino)-18-ethyl-15-fluoro- (5S,9S,10S,12R)-9-(Dimethylamino)-15-fluoro-4,5,8,25-tetra-
4,5,8,25-tetrahydroxy-2,6-dioxo-18,23-diazahexacyclo- hydroxy-21-methyl-2,6-dioxo-18,23-diazahexacyclo-
[12.11.0.03,12.05,10.016,24.017,21]pentacosa-1(25),3,7,14,16(24)- [12.11.0.03,12.05,10.016,24.017,21]pentacosa-1(25),3,7,14,16(24)-
pentaene-7-carboxamide (10d). By similar procedures, compound pentaene-7-carboxamide (11a). 1H NMR (400 MHz, CD3OD,
10d was prepared from 9a using acetaldehyde in the reductive HCl salt) δ 4.31 (s, 1 H), 4.08 (s, 1 H), 3.47−3.40 (m, 2 H), 3.18−
alkylation step. 1H NMR (400 MHz, CD3OD, HCl salt) δ 4.85 (d, J = 3.11 (m, 2 H), 3.08−2.95 (m, 9 H), 2.23−2.13 (m, 4 H), 1.65−1.55
6.4 Hz, 1 H), 4.11 (s, 1 H), 3.72−3.66 (m, 1 H), 3.63−3.58 (m, 1 H), (m, 1 H), 1.15 (s, 3 H). MS (ESI) m/z calcd for C27H32FN4O7 [M +
3.46 (dd, J = 5.0, 12.8 Hz, 1 H), 3.42−3.33 (m, 2 H), 3.19−2.97 (m, H]+ 543.23; found 543.14.
10 H), 2.80 (br s, 1 H), 2.54−2.45 (m, 1 H), 2.25−2.03 (m, 3 H), (5S,9S,10S,12R)-9-(Dimethylamino)-15-fluoro-4,5,8,25-tetra-
1.64−1.54 (m, 1 H), 1.38 (t, J = 6.9 Hz, 3 H). MS (ESI) m/z calcd for hydroxy-18,21-dimethyl-2,6-dioxo-18,23-diazahexacyclo-
[12.11.0.03,12.05,10.016,24.017,21]pentacosa-1(25),3,7,14,16(24)-
C28H34FN4O7 [M + H]+ 557.24; found 557.15. pentaene-7-carboxamide (11b). 1H NMR (400 MHz, CD3OD,
(5S,9S,10S,12R)-9-(Dimethylamino)-15-fluoro-4,5,8,25-tetra- HCl salt) δ 4.36 (s, 1 H), 4.08 (s, 1 H), 3.82−3.75 (m, 1 H), 3.41−
hydroxy-2,6-dioxo-18,23-diazahexacyclo-
[12.11.0.03,12.05,10.016,24.017,21]pentacosa-1(25),3,7,14,16(24)- 3.33 (m, 1 H), 3.22−3.11 (m, 3 H), 3.08−2.96 (m, 11 H), 2.34−2.18
pentaene-7-carboxamide (10a). Compound 9a was deprotected (m, 3 H), 2.10−2.04 (m, 1 H), 1.65−1.56 (m, 1 H), 1.09 (s, 3 H). MS
using previously described procedures6 to provide compound 10a. 1H (ESI) m/z calcd for C28H34FN4O7 [M + H]+ 557.24; found 557.13.
NMR (400 MHz, CD3OD, HCl salt) δ 4.80 (d, J = 6.4 Hz, 1 H), 4.10 (5S,9S,10S,12R)-9-(Dimethylamino)-15-fluoro-4,5,8,25-tetra-
(s, 1 H), 3.49−3.40 (m, 3 H), 3.12−2.96 (m, 9 H), 2.89 (t, J = 11.9 hydroxy-21-methyl-2,6-dioxo-18-propyl-18,23-
diazahexacyclo[12.11.0.0 3,12 .0 5,10 .0 16,24 .0 17,21 ]pentacosa-1-
Hz, 1 H), 2.79−2.71 (m, 1 H), 2.49−2.39 (m, 1 H), 2.24−2.16 (m, 2 (25),3,7,14,16(24)-pentaene-7-carboxamide (11c). 1H NMR
H), 2.12−2.02 (m, 1 H), 1.65−1.56 (m, 1 H). MS (ESI) m/z calcd for (400 MHz, CD3OD, HCl salt) δ 4.40 (s, 1 H), 4.10 (s, 1 H),
C26H30FN4O7 [M + H]+ 529.21; found 529.07. 3.75−3.68 (m, 1 H), 3.49−3.42 (m, 1 H), 3.38−3.31 (m, 1 H), 3.24−
The following analogues (10b, 10c, 10f−10h, and 11a−11c) were 3.15 (m, 3 H), 3.12−3.07 (m, 1 H), 3.04−2.96 (m, 8 H), 2.30−2.18
prepared using similar procedures. See details in Supporting (m, 3 H), 2.13−2.07 (m, 1 H), 1.83−1.75 (m, 1 H), 1.72−1.56 (m, 2
Information. H), 1.08 (s, 3 H), 0.98 (t, J = 7.3 Hz, 3 H). MS (ESI) m/z calcd for
(5S,9S,10S,12R)-9-(Dimethylamino)-15-fluoro-4,5,8,25-tetra- C30H38FN4O7 [M + H]+ 585.27; found 585.26.
hydroxy-18-methyl-2,6-dioxo-18,23-diazahexacyclo-
[12.11.0.03,12.05,10.016,24.017,21]pentacosa-1(25),3,7,14,16(24)- Phenyl 4-Bromo-2-hydroxy-3-iodo-6-methyl-5-
pentaene-7-carboxamide. Diastereomer A, 10b: 1H NMR (400 (trifluoromethoxy)benzoate (13). To phenol 1213 (5.20 mmol,
MHz, CD3OD, HCl salt) δ 4.77 (d, J = 6.4 Hz, 1 H), 4.12 (s, 1 H), obtained from treatment of 2.50 g of the corresponding benzyl ether
3.79−3.74 (m, 1 H), 3.45 (dd, J = 5.5, 12.8 Hz, 1 H), 3.37−3.31 (m, 1 with TFA/anisole, containing inseparable impurities, ∼75% pure) in
H), 3.14−2.97 (m, 13 H), 2.76−2.75 (m, 1 H), 2.58−2.49 (m, 1 H), toluene (20 mL) at rt was added NaH (0.83 g, 20.80 mmol, 60% in
2.50−2.18 (m, 2 H), 2.07−2.00 (m, 1 H), 1.65−1.55 (m, 1 H); MS mineral oil, 4 equiv) in small portions. The reaction mixture was
(ESI) m/z calcd for C27H32FN4O7 [M + H]+ 543.23, found 543.07. stirred at rt for 20 min. Iodine (5.28 g, 20.80 mmol, 4 equiv) was
Diastereomer B, 10c: 1H NMR (400 MHz, CD3OD, HCl salt) δ 4.77 added. The reaction mixture was stirred at rt overnight, diluted with
(d, J = 6.0 Hz, 1 H), 4.10 (s, 1 H), 3.79−3.73 (m, 1 H), 3.47 (dd, J = EtOAc (200 mL), washed with 1 N aqueous HCl (100 mL × 1), 5%
5.5, 12.8 Hz, 1 H), 3.37−3.32 (m, 1 H), 3.12−2.97 (m, 13 H), 2.78 (br aqueous Na2S2O3 (100 mL × 2), and brine (100 mL × 1), dried over
s, 1 H), 2.56−2.51 (m, 1 H), 2.23 (br d, J = 10.1 Hz, 1 H), 2.14 (t, J = sodium sulfate, and concentrated under reduced pressure to yield the
14.2 Hz, 1 H), 2.05−1.98 (m, 1 H), 1.63−1.54 (m, 1 H); 13C NMR crude product 13 as a pale oil. MS (ESI) m/z calcd for C15H8BrF3IO4
(400 MHz, CD3OD, HCl salt) δ 194.96, 187.82, 174.60, 174.26, [M − H]− 514.86; found 514.81.
153.09, 150.77, 147.47, 137.16, 115.57, 112.40, 109.68, 105.77, 96.12, Phenyl 2-(Benzyloxy)-4-bromo-3-iodo-6-methyl-5-
(trifluoromethoxy)benzoate (14). To the above crude phenol 13
75.47, 70.75, 63.21, 54.89, 43.07, 42.03, 41.81, 40.97, 35.91, 35.27,
(5.20 mmol) in DMF (10 mL) at rt was added potassium carbonate
35.12, 33.88, 31.12, 27.20; MS (ESI) m/z calcd for C27H32FN4O7 [M
(1.44 g, 10.44 mmol, 2 equiv) and benzyl bromide (0.74 mL, 6.23
+ H]+ 543.23, found 543.07.
mmol, 1.2 equiv). The reaction mixture was stirred at rt for 3 h, diluted
(5S,9S,10S,12R)-9-(Dimethylamino)-18-[2-(dimethylamino)-
ethyl]-15-fluoro-4,5,8,25-tetrahydroxy-2,6-dioxo-18,23- with EtOAc (200 mL), washed with water (200 mL × 1, 100 mL × 1)
diazahexacyclo[12.11.0.0 3,12 .0 5,10 .0 16,24 .0 17,21 ]pentacosa-1- and brine (50 mL × 1), dried over sodium sulfate, and concentrated
(25),3,7,14,16(24)-pentaene-7-carboxamide (10f). 1H NMR under reduced pressure. Flash column chromatography on silica gel
(400 MHz, CD3OD, HCl salt) δ 5.00−4.94 (m, 1 H), 4.12−3.97 with 0−3% EtOAc/hexane yielded the desired product 14 as a pale oil
(m, 2 H), 3.90−3.75 (m, 2 H), 3.74−3.58 (m, 2 H), 3.52−3.37 (m, 2 (3.48 g, ∼90% pure). 1H NMR (400 MHz, CDCl3) δ 7.55−7.00 (m,
H), 3.27−2.85 (m, 17 H), 2.84−2.78 (m, 1 H), 2.62−2.48 (m, 1 H), 10 H), 5.11 (s, 2 H), 2.44 (s, 3 H). MS (ESI) m/z calcd for
2.26−2.04 (m, 3 H), 1.68−1.54 (m, 1 H). MS (ESI) m/z calcd for C22H14BrF3IO4 [M − H]− 604.91; found 604.83.
C30H39FN5O7 [M + H]+ 600.29; found 600.09. Phenyl 2-(Benzyloxy)-4-bromo-3-{[(tert-butoxy)carbonyl]-
(5S,9S,10S,12R)-18-{2-[(2,2-Difluoroethyl)amino]ethyl}-9-(di- amino}-6-methyl-5-(trifluoromethoxy)benzoate (15). To com-
methylamino)-15-fluoro-4,5,8,25-tetrahydroxy-2,6-dioxo- pound 14 (5.20 mmol, 90% pure) was added cesium carbonate (2.54
18,23-diazahexacyclo[12.11.0.03,12.05,10.016,24.017,21]pentacosa- g, 7.80 mmol, 1.5 equiv), BocNH2 (0.67 g, 5.70 mmol, 1.1 equiv),
1(25),3,7,14,16(24)-pentaene-7-carboxamide (10g). 1H NMR Xantphos (1.20 g, 2.07 mmol, 0.4 equiv), Pd(OAc)2 (224 mg, 1.00
(400 MHz, CD3OD, HCl salt) δ 6.35 (J = 2.4 Hz, J [19F−1H] = 53.7 mmol, 0.2 equiv), and anhydrous 1,4-dioxane (10 mL). Nitrogen gas

G DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

was bubbled through the mixture for 5 min. The reaction vessel was followed by the addition of enone 7 (147 mg, 0.30 mmol, 1 equiv) in
sealed and heated at 80 °C for 48 h with vigorous stirring. After anhydrous THF (1 mL) dropwise over 1 min. The reaction was stirred
cooling down to rt, water (100 mL) was added. The reaction mixture from −78 to 0 °C for 2 h and quenched with saturated aqueous
was extracted with methylene chloride (100 mL × 1, 50 mL × 2). The sodium bicarbonate (20 mL). The reaction mixture was extracted with
combined extracts were dried over sodium sulfate and concentrated methylene chloride (20 mL × 3). The combined extracts were dried
under reduced pressure. Flash column chromatography on silica gel over sodium sulfate and concentrated under reduced pressure. Flash
with 0−15% EtOAc/hexane yielded the desired product 15 as a white column chromatography on silica gel with 0−20% EtOAc/hexane
solid (0.87 g, 28% overall yield). 1H NMR (400 MHz, CDCl3) δ yielded the two diastereomers of the desired product (diastereomer A,
7.45−7.20 (m, 8 H), 7.03 (d, J = 7.3 Hz, 2 H), 6.07 (br s, 1 H), 5.03 19ba, 100 mg, yellow foam, 33%; diastereomer B, 19bb, 60 mg, yellow
(s, 2 H), 2.46 (s, 3 H), 1.46 (s, 9 H). MS (ESI) m/z calcd for foam, 20%). MS (ESI) m/z calcd for C53H64F3N4O10Si [M + H]+
C27H24BrF3NO6 [M − H]− 594.01; found 594.01. 1001.44, found 1001.47.
Phenyl 2-(Benzyloxy)-3-{[(tert-butoxy)carbonyl]amino}-4- (5S,9S,10S,12R)-9-(Dimethylamino)-4,5,8,25-tetrahydroxy-
formyl-6-methyl-5-(trifluoromethoxy)benzoate (16). To com- 18-methyl-2,6-dioxo-15-(trifluoromethoxy)-18,23-
pound 15 (0.68 g, 1.14 mmol) in anhydrous THF (6 mL) at −78 °C diazahexacyclo[12.11.0.0 3,12 .0 5,10 .0 16,24 .0 17,21 ]pentacosa-1-
(dry ice−acetone bath) was added PhLi (0.95 mL, 1.80 M/n-Bu2O, (25),3,7,14,16(24)-pentaene-7-carboxamide (20b). Compound
1.71 mmol, 1.5 equiv) dropwise over 1 min. After stirring at −78 °C 19ba (33 mg, 0.033 mmol) was deprotected according to previously
for 10 min, n-BuLi (0.86 mL, 1.60 M/hexanes, 1.38 mmol, 1.2 equiv) described desilylation and hydrogenation procedures6 to yield the
was added dropwise over 2 min. The reaction was stirred at −78 °C desired product 20b as an orange solid after reverse phase HPLC
for 5 min. Dry DMF (0.26 mL, 3.36 mmol, 3 equiv) was added purification (13 mg, HCl salt, 55% over 2 steps). 1H NMR (400 MHz,
dropwise. The reaction was stirred from −78 to 0 °C over 1 h and CD3OD) δ 4.75 (d, J = 7.4 Hz, 1 H), 4.08 (s, 1 H), 3.75−3.65 (m, 1
quenched with saturated aqueous sodium bicarbonate (50 mL). The H), 3.50−2.90 (m, 20 H), 2.57−2.47 (m, 1 H), 2.31−2.05 (m, 3 H),
reaction mixture was extracted with methylene chloride (50 mL × 3). 1.68−1.57 (m, 1 H). 13C NMR (400 MHz, CD3OD) δ 194.95, 187.83,
The combined extracts were dried over sodium sulfate and 174.92, 174.26, 150.52, 138.79, 137.70, 123.74, 122.26, 121.17, 115.99,
concentrated under reduced pressure. Flash column chromatography 111.49, 109.63, 96.11, 75.48, 70.71, 63.64, 55.25, 43.03, 42.70, 41.97,
on silica gel with 0−15% EtOAc/hexane yielded the desired product 41.06, 36.35, 35.81, 35.13, 33.75, 30.38, 27.51. MS (ESI) m/z calcd for
16 as a pale solid (232 mg, 37%). 1H NMR (400 MHz, CDCl3) δ C28H32F3N4O8 [M + H]+ 609.22; found 609.30.
10.21 (s, 1 H), 7.90 (br s, 1 H), 7.45−7.20 (m, 8 H), 7.05 (d, J = 7.3 The following compounds (20a, 20c−20f) were prepared similarly.
Hz, 2 H), 5.00 (s, 2 H), 2.42 (s, 3 H), 1.43 (s, 9 H). MS (ESI) m/z See experimental details in Supporting Information.
calcd for C28H25F3NO7 [M − H]− 544.16; found 544.22. (5S,9S,10S,12R)-9-(Dimethylamino)-4,5,8,25-tetrahydroxy-
Phenyl 2-(Benzyloxy)-3-{[(tert-butoxy)carbonyl](prop-2-en- 2,6-dioxo-15-(trifluoromethoxy)-18,23-diazahexacyclo-
1-yl)amino}-4-formyl-6-methyl-5-(trifluoromethoxy)benzoate [12.11.0.03,12.05,10.01624.017,21]pentacosa-1(25),3,7,14,16(24)-
(17). To compound 16 (232 mg, 0.43 mmol) in dry DMF (2 mL) at rt pentaene-7-carboxamide (20a). 1H NMR (400 MHz, CD3OD,
was added NaH (21 mg, 60% in mineral oil, 0.52 mmol, 1.2 equiv). HCl salt) δ 4.90−4.80 (m, 1 H), 4.09 (s, 1 H), 3.50−2.85 (m, 10 H),
After stirring at rt for 30 min, allyl bromide (56 μL, 0.64 mmol, 1.5 2.82−2.74 (m, 1 H), 2.43−2.15 (m, 7 H), 2.15−2.05 (m, 1 H), 1.62
equiv) was added. The reaction mixture was stirred at rt for 1 h, (dd, J = 10.6 Hz, 1 H). MS (ESI) m/z calcd for C27H30F3N4O8 [M +
diluted with EtOAc (50 mL), washed with water (50 mL × 2) and H]+ 595.20; found 595.31.
brine (50 mL × 1), dried over sodium sulfate, and concentrated under (5S,9S,10S,12R)-9-(Dimethylamino)-4,5,8,25-tetrahydroxy-
reduced pressure. Flash column chromatography on silica gel with 0− 18,19-dimethyl-2,6-dioxo-15-(trifluoromethoxy)-18,23-
8% EtOAc/hexane yielded the desired product 17 as a pale oil (206 diazahexacyclo[12.11.0.0 3,12 .0 5,10 .0 16,24 .0 17,21 ]pentacosa-1-
mg, 82%). 1H NMR (400 MHz, CDCl3) δ 10.16 (s, 1 H), 7.40−6.95 (25),3,7,14,16(24)-pentaene-7-carboxamide (20c). 1H NMR
(m, 10 H), 5.95−5.75 (m, 1 H), 5.10−4.85 (m, 4 H), 4.64, 4.28 (dd, (400 MHz, CD3OD, HCl salt) δ 4.85−4.84 (m, 1 H), 4.11 (s, 1
dd, J = 5.5, 12.8 Hz, J = 4.9, 12.2 Hz, 1 H), 4.00, 3.89 (dd, dd, J = 8.1, H), 3.67−3.63 (m, 1 H), 3.13−2.93 (m, 15 H), 2.76−2.72 (m, 1 H),
10.2 Hz, J = 8.6, 12.8 Hz, 1 H), 2.46, 2.43 (s, s, 3 H), 1.53, 1.50 (s, s, 9 2.35−2.22 (m, 2 H), 1.88−1.83 (m, 1 H), 1.71−1.62 (m, 1 H),1.66 (d,
H). MS (ESI) m/z calcd for C31H29F3NO7 [M − H]− 584.19; found J = 11.2, 3 H). MS (ESI) m/z calcd for C29H34F3N4O8 [M + H]+
584.24. 623.24; found 623.43.
5-tert-Butyl 7-Phenyl 6-(Benzyloxy)-1,8-dimethyl-9-(trifluor- (20S,24S,25S,27R)-24-(Dimethylamino)-15,19,20,23-tetrahy-
omethoxy)-1H,2H,3H,3aH,4H,5H,9bH-pyrrolo[3,2-c]quinoline- droxy-17,21-dioxo-2-(trifluoromethoxy)-5,13-diazaheptacyclo-
5,7-dicarboxylate (18b). To compound 17 (206 mg, 0.35 mmol) in [14.12.0.03,14.04,11.05,9.018,27.020,25]octacosa-1,3(14),15,18,22-
DMF (2 mL) was added N-methyl glycine (47 mg, 0.53 mmol, 1.5 pentaene-22-carboxamide (20d). 1H NMR (400 MHz, CD3OD,
equiv). The reaction mixture was heated at 100 °C for 24 h. After HCl salt) δ 4.80 (d, J = 6.0, 1 H), 4.41−4.38 (m, 1 H), 4.13 (s, 1 H),
cooling to rt, the reaction mixture was diluted with EtOAc (50 mL), 3.80−3.77 (m, 1 H), 3.66−3.60 (m, 1 H), 3.52−3.48 (m, 1 H), 3.16−
washed with aqueous saturated sodium bicarbonate (50 mL × 2) and 3.03 (m, 12 H), 2.42−2.21 (m, 6 H), 2.09−2.01 (m, 1 H), 1.96−1.94
brine (50 mL × 1), dried over sodium sulfate, and concentrated under (m, 1 H), 1.69−1.60 (m, 1 H). MS (ESI) m/z calcd for C30H34F3N4O8
reduced pressure. Flash column chromatography on silica gel with 0− [M + H]+ 635.24; found 635.17.
15% EtOAc/hexane yielded the desired product 18b as a white foam (21S,25S,26S,28R)-25-(Dimethylamino)-16,20,21,24-tetrahy-
(190 mg, 89%). 1H NMR (400 MHz, CDCl3), broad and complex due droxy-18,22-dioxo-2-(trifluoromethoxy)-5,14-diazaheptacyclo-
to the presence of various rotamers and/or conformers. MS (ESI) m/z [15.12.0.03,15.04,12.05,10.019,28.021,26]nonacosa-1,3(15),16,19,23-
calcd for C33H36F3N2O6 [M + H]+ 613.25; found 613.36. pentaene-23-carboxamide (20e). 1H NMR (400 MHz, CD3OD,
tert-Butyl (1R,19S,26S,27S)-14,22-Bis(benzyloxy)-19-[(tert- HCl salt) δ 5.00 (d, J = 9.2, 1 H), 4.13 (s, 1 H), 3.73−3.70 (m, 1 H),
butyldimethylsilyl)oxy]-26-(dimethylamino)-18-hydroxy-7- 3.45−3.38 (m, 1 H), 3.29−3.26 (m, 1 H), 3.16−3.03 (m, 11 H), 2.49−
methyl-16,20-dioxo-4-(trifluoromethoxy)-24-oxa-7,12,23- 2.22 (m, 5 H), 1.97−1.79 (m, 5 H), 1.69−1.61 (m, 2 H). MS (ESI)
triazaheptacyclo[15.11.0.03,15.05,13.06,10.019,27.021,25]octacosa-3- m/z calcd for C31H36F3N4O8 [M + H]+ 649.25; found 649.12.
(15),4,13,17,21(25),22-hexaene-12-carboxylate (19b). To diiso- (21S,25S,26S,28R)-25-(Dimethylamino)-16,20,21,24-tetrahy-
propylamine (51 μL, 0.36 mmol, 1.2 equiv) in anhydrous THF (1 mL) droxy-18,22-dioxo-2-(trifluoromethoxy)-8-oxa-5,14-
at −78 °C (dry ice−acetone bath) was added n-BuLi (0.23 mL, 1.60 diazaheptacyclo[15.12.0.03,15.04,12.05,10.019,28.021,26]nonacosa-
M/hexanes, 0.36 mmol, 1.2 equiv) dropwise. The reaction was stirred 1,3(15),16,19,23-pentaene-23-carboxamide (20f). 1H NMR
at 0 °C for 10 min and cooled to −78 °C. TMEDA (56 μL, 0.36 (400 MHz, CD3OD, HCl salt) δ 5.11 (d, J = 7.6, 1 H), 4.14 (s, 1
mmol, 1.2 equiv) was added, followed by the addition of compound H), 4.07−4.04 (m, 1 H), 3.94−3.88 (m, 3 H), 3.70−2.90 (m, 8 H),
18b (186 mg, 0.30 mmol) in anhydrous THF (2 mL) dropwise over 2 3.20−2.99 (m, 8 H), 2.66−2.59 (m, 1 H), 2.35−2.22 (m, 2 H), 1.68−
min. The resulting deep-red solution was stirred at −78 °C for 30 min. 1.62 (m, 1 H). MS (ESI) m/z calcd for C30H34F3N4O9 [M + H]+
LHMDS (0.36 mL, 1.0 M/THF, 0.36 mmol, 1.2 equiv) was added, 651.23; found 651.14.

H DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry


Article

In Vitro Coupled P. aeruginosa Transcription/Translation AUTHOR INFORMATION


Assay. Antitranslational activity (IC50 values) was assessed in a P.
aeruginosa in vitro coupled transcription/translation assay (TnT) with Corresponding Author
a firefly luciferase readout (catalogue no. L1020, Promega, Madison, *Phone: 617-715-3553. Fax: 617-926-3557. E-mail: xyxiao@
WI), as described by Fyfe et al.15 Reactions were run at a volume of 20 tphase.com.
μL in Costar black 96-well assay plates (Costar catalogue no. 3915) for Notes
60 min at 37 °C. The reaction was stopped by placing on ice for 5 min,
The authors declare no competing financial interest.
followed by addition of 25 μL of luciferase assay substrate (catalogue
no. E1500, Promega, Madison, WI). Plates were read on a LUMIStar
Optima (BMG Labtech, Ortenberg, Germany) with gain set to 3600,
0.2 s read, 0 s between wells. Percent luminescence was plotted against
■ ACKNOWLEDGMENTS
We thank Drs. Andrew Myers, Eric Gordon, and Joaquim Trias
inhibitor concentration with 50% inhibition versus untreated controls for valuable discussions during the course of this work. We also
marked as the IC50 value. thank Dr. Shu-hui Chen and his colleagues at WuXi Apptec for
Susceptibility Testing. Compound stocks were prepared and
external chemistry support.


serially diluted in sterile deionized water. Minimal inhibitory
concentration (MIC) determinations were performed in liquid
medium in 96-well microtiter plates according to the methods ABBREVIATIONS USED
described by the Clinical and Laboratory Standards Institute (CLSI).16 aq, aqueous; ATCC, American Type Culture Collection; CFU,
Cation-adjusted Mueller Hinton broth was obtained from BBL colony-forming units; CLSI, Clinical and Laboratory Standards
(catalogue no. 212322, Becton Dickinson, Sparks, MD), prepared Institute; CRE, carbapenem-resistant Enterobacteriaceae; DMF,
fresh and kept at 4 °C prior to testing. Lysed horse blood (catalogue N,N-dimethylformamide; HPLC, high-performance liquid
no. 205, Quad Five, Ryegate, Montana) was used to supplement
chromatography; IC50, half-minimum inhibitory concentration;
medium, as appropriate. All test methods met acceptable standards
based on recommended quality control ranges for all comparator
IV, intravenous; LDA, lithium diisopropylamide; LHMDS,
antibiotics and the appropriate ATCC quality control strains. lithium hexamethydisilazide; MDR, multidrug-resistant; MIC,
Immunocompetent Lung Infection Model. The mouse lung minimum inhibitory concentration; MRSA, methicillin-resistant
infection model was performed at Vivisource, Waltham, MA. Female Staphylococcus aureus; MS, mass spectrometry; NMR, nuclear
BALB/c mice weighing 18−20 g were infected with ∼2 × 107 CFU/ magnetic resonance; PBS, phosphate buffered saline; SAR,
mouse of P. aeruginosa PA1145, a cystic fibrosis isolate from Children’s structure−activity relationship; THF, tetrahydrofuran; TLC,
Hospital, Boston, via intranasal administration of 0.05 mL of cell thin-layer chromatography; TMEDA, N,N,N′,N′-tetramethyle-
suspension under light anesthesia. One group did not receive drug thylenediamine; TnT, coupled transcription/translation assay


treatment, and lungs were harvested at 2 h postinfection. At 2 and 12 h
postinfection, mice were treated with compound 10c, meropenem, or REFERENCES
amikacin intravenously. Six mice per group were treated with each
drug concentration. Twenty-four h post initiation of treatment, mice (1) Gupta, N.; Limbago, B. M.; Patel, J. B.; Kallen, A. J. Carbapenem-
were euthanized by CO2 inhalation. The lungs of the mice were resistant Enterobacteriaceae: Epidemiology and Prevention. Clin. Infect.
aseptically removed, weighed, homogenized, serially diluted, and Dis. 2011, 53, 60−67.
plated on MacConkey medium. The plates were incubated overnight (2) Mulligan, M. E.; Murray-Leisure, K. A.; Ribner, B. S.; Standiford,
H. C.; John, J. F.; Korvick, J. A.; Kauffman, C. A.; Yu, Y. L. Methicillin-
at 37 °C in 5% CO2. CFU per gram of lung was calculated by
Resistant Staphylococcus aureus: A Consensus Review of the Micro-
enumerating the plated colonies then adjusting for serial dilutions and
biology, Pathogenesis, and Epidemiology with Implications for
the weight of the lung. Individual animal CFU/g lung data was plotted
Prevention and Management. Am. J. Med. 1993, 94, 313−328.
using GraphPad Prism. (3) Conover, L H. Discovery of Drugs from Microbiological Sources.
Immunocompetent Thigh Infection Model. The mouse thigh In Drug Discovery, Science and Development in a Changing Society;
infection model was performed at University of North Texas Health American Chemical Society: Washington, DC, 1971; Vol. 108, pp 33−
Science Center, Fort Worth, TX. Groups of five female specific- 80.
pathogen-free CD-1 mice weighing 22 ± 2 g were used. On day 0, (4) (a) Chopra, I.; Roberts, M. Tetracycline Antibiotics: Model of
animals were inoculated intramuscularly (0.1 mL/thigh) with 3−5 × Action, Applications, Molecular Biology, and Epidemiology of
106 CFU/mouse of P. aeruginosa PA694, a clinical isolate from Bacterial Resistance. Microbiol. Mol. Biol. Rev. 2001, 65, 232−260.
Eurofins-Medinet, into the right thigh. Two groups did not receive (b) Levy, S. B. Evolution and Spread of Tetracycline Resistance
drug treatment, and thighs were harvested at 2 and 24 h postinfection. Determinants. J. Antimicrob. Chemother. 1989, 24, 1−3. (c) Lomov-
Remaining mice were administered intravenously with either vehicle, skaya, O.; Watkins, W. J. Efflux Pimps: Their Role in Antibacterial
compound 10c, or levofloxacin at 2 and 12 h postinfection. The Drug Discovery. Curr. Med. Chem. 2001, 8, 1699−1711.
muscle of the right thigh of each animal was harvested at 24 h (5) Sum, P.-E.; Lee, V. J.; Testa, R. T.; Hlavka, J. J.; Ellestad, G. A.;
postinfection. Harvested thigh tissues were homogenized in 2 mL of Bloom, J. D.; Gluzman, Y.; Tally, F. P. Glycylcyclines. 1. A New
PBS (pH 7.4) with a Polytron tissue homogenizer. The homogenates Generation of Potent Antibacterial Agents through Modification of 9-
were serially diluted and plated on Brain Heart Infusion agar + 0.5% Aminotetracyclines. J. Med. Chem. 1994, 37, 184−188.
charcoal (w/v) for CFU determination per gram of thigh. Individual (6) Xiao, X.-Y.; Hunt, D. K.; Zhou, J.; Clark, R. B.; Dunwoody, N.;
animal CFU/gram thigh data was plotted using GraphPad Prism. Fyfe, C.; Grossman, T. H.; O’Brien, W. J.; Plamondon, L.; Ronn, M.;


Sun, C.; Zhang, W.-Y.; Sutcliffe, J. A. Fluorocyclines. 1. 7-Fluoro-9-
pyrrolidinoacetamido-6-demethyl-6-deoxytetracycline: A Potent,
ASSOCIATED CONTENT Broad Spectrum Antibacterial Agent. J. Med. Chem. 2012, 55, 597−
*
S Supporting Information 605.
Detailed information on the synthesis and analytical data of (7) Solomkin, J. S.; Ramesh, M. K.; Cesnauskas, G.; Novikovs, N.;
Stefanova, P.; Sutcliffe, J. A.; Walpole, S. M.; Horn, P. T. Phase 2,
compounds 10b, 10c, 10f−10h, 11a−11c, 20a, and 20c−20f. Randomized, Double-blind Study of the Efficacy and Safety of Two
The Supporting Information is available free of charge on the Dose Regimens of Eravacycline versus Ertapenem for Adult
ACS Publications website at DOI: 10.1021/acs.jmed- Community-Acquired Complicated Intra-abdominal Infections. Anti-
chem.5b00262. microb. Agents Chemother. 2014, 58, 1847−1854.

I DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry Article

(8) (a) Sun, C.; Hunt, D. K.; Clark, R. B.; Lofland, D.; O’Brien, W. J.;
Plamondon, L.; Xiao, X.-Y. Synthesis and Antibacterial Activity of
Pentacyclines: A Novel Class of Tetracycline Analogs. J. Med. Chem.
2011, 54, 3704−3731. (b) Sun, C.; Wang, Q.; Brubaker, J. D.; Wright,
P. M.; Lerner, C. D.; Noson, K.; Charest, M.; Siegel, D. R.; Wang, Y.-
M.; Myers, A. G. A Robust Platform for the Synthesis of New
Tetracycline Antibiotics. J. Am. Chem. Soc. 2008, 130, 17913−17927.
(9) (a) Clark, R. B.; He, M.; Fyfe, C.; Lofland, D.; O’Brien, W. J.;
Plamondon, L.; Xiao, X.-Y. 8-Azatetracyclines: Synthesis and
Evaluation of a Novel Class of Tetracycline Antibacterial Agents. J.
Med. Chem. 2011, 54, 1511−1528. (b) Clark, R. B.; Hunt, D. K.; Me,
H.; Achorn, C.; Chen, C.-L.; Deng, D.; Fyfe, C.; Grossman, T. H.;
Hogan, P. C.; O’Brien, W. J.; Plamondon, L.; Ronn, M.; Sutcliffe, J. A.;
Zhu, Z.; Xiao, X.-Y. Fluorocyclines. 2. Optimization of the C-9 Side-
Chain for Antibacterial Activity and Oral Efficacy. J. Med. Chem. 2012,
55, 606−622. (c) Clark, R. B.; He, M.; Deng, D.; Sun, C.; Chen, C.-L.;
Hunt, D. K.; O’Brien, W. J.; Fyfe, C.; Grossman, T. H.; Sutcliffe, J. A.;
Achorn, C.; Hogan, P. C.; Katz, C. E.; Niu, J.; Zhang, W.-Y.; Zhu, Z.;
Ronn, M.; Xiao, X.-Y. Synthesis and Biological Evaluation of 8-
Aminomethyltetracycline Derivatives as Novel Antibacterial Agents. J.
Med. Chem. 2013, 56, 8112−8138.
(10) Charest, M. G.; Lerner, C. D.; Brubaker, J. D.; Siegel, D. R.;
Myers, A. G. A Convergent Enantioselective Route to Structurally
Diverse 6-Deoxytetracycline Antibiotics. Science 2005, 308, 395−398.
(11) Brubaker, J. D.; Myers, A. G. A Practical, Enantioselective
Synthetic Route to a Key Precursor to the Tetracycline Antibiotics.
Org. Lett. 2007, 9, 3523−3525.
(12) He, Y.; Mahmud, H.; Moningka, R.; Lovely, C. J.; Dias, H. V. R.
Cyclization Reactions of N-Acryloyl-2-aminobenzaldehyde Deriva-
tives: Formal Total Synthesis of Martinellic Acid. Tetrahedron 2006,
62, 8755−8769.
(13) Chen, C.-L.; Clark, R. B.; Deng, Y.; Plamondon, L.; Sun, C.;
Xiao, X.-Y. Tetracycline Analogs and Their Therapeutic Use against
Infections. PCT Int. Appl. WO 201202712, 2102.
(14) Muci, A. R.; Buchwald, S. L. Practical Palladium Catalysts for
C−N and C−O Bond Formation. Top. Curr. Chem. 2002, 219, 131−
209.
(15) Fyfe, C.; Sutcliffe, J. A.; Grossman, T. H. Development and
Characterization of a Pseudomonas aeruginosa in Vitro Coupled
Transcription−Translation Assay System for Evaluation of Translation
Inhibitors. J. Microbiol. Methods 2012, 90, 256−261.
(16) Methods for Dilution Antimicrobial Susceptibility for Bacteria That
Grow Aerobically: Approved Standard, 9th ed.; CLSI document M07-
A9E; Clinical and Laboratory Standards Institute (CLSI): 940 West
Valley Road, Suite 1400, Wayne, Pennsylvania, 2012; Vol. 32, no. 2.

J DOI: 10.1021/acs.jmedchem.5b00262
J. Med. Chem. XXXX, XXX, XXX−XXX

You might also like