Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 1.
Published in final edited form as: Eur J Med Chem. 2012 Jun 23;54:771–783. doi: 10.1016/j.ejmech.2012.06.032

Potent Direct Inhibitors of Factor Xa Based on the Tetrahydroisoquinoline Scaffold

Rami A Al-Horani 1, Akul Y Mehta 1, Umesh R Desai 1,*
PMCID: PMC3408863  NIHMSID: NIHMS389693  PMID: 22770607

Abstract

Direct inhibition of coagulation factor Xa (FXa) carries significant promise for developing effective and safe anticoagulants. Although a large number of FXa inhibitors have been studied, each can be classified as either possessing a highly flexible or a rigid core scaffold. We reasoned that an intermediate level of flexibility will provide high selectivity for FXa considering that its active site is less constrained in comparison to thrombin and more constrained as compared to trypsin. We studied several core scaffolds including 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid for direct FXa inhibition. Using a genetic algorithm-based docking and scoring approach, a promising candidate 23 was identified, synthesized, and found to inhibit FXa with a Ki of 28 μM. Optimization of derivative 23 resulted in the design of a potent dicarboxamide 47, which displayed a Ki of 135 nM. Dicarboxamide 47 displayed at least 1852-fold selectivity for FXa inhibition over other coagulation enzymes and doubled PT and aPTT of human plasma at 17.1 μM and 20.2 μM, respectively, which are comparable to those of clinically relevant agents. Dicarboxamide 47 is expected to serve as an excellent lead for further anticoagulant discovery.

1. Introduction

Anticoagulants represent the basis for treatment and prevention of thromboembolic disorders [1]. A priori, the inhibition of any coagulation enzyme can be expected to reduce or prevent clotting, yet most strategies have targeted two serine proteases, factor IIa (FIIa, or thrombin) and factor Xa (FXa), which belong to the common pathway of the coagulation cascade [2]. Of these, thrombin plays a major role in a number of physiologically relevant responses that rely on its catalytic activity [3,4]. Thus, inhibition of thrombin not only reduces cleavage of fibrinogen to fibrin, a key aspect of clotting, but also other processes such as platelet activation, platelet aggregation and angiogenesis. On the other hand, FXa has a rather limited role in comparison, which is to generate thrombin.

Several lines of evidence suggest that FXa may represent a better target for anticoagulation. FXa’s contribution to amplification of the coagulation signal is higher than that of thrombin [5,6]. Its occurrence earlier in the coagulation pathway suggests that a FXa inhibitor may be more effective in blocking progression of a coagulation signal than a thrombin inhibitor [7,8]. Recent results with indirect parenteral and direct oral FXa inhibitors have shown reduced bleeding complications [9]. Also, thrombin inhibitors have been associated with rebound hypercoagulability [10,11], while peptidomimetic inhibitors have been shown to inhibit both free as well as clot-bound FXa [12,13].

FXa can be inhibited through two major pathways including an indirect, antithrombin-dependent pathway [14,15] or a direct FXa targeting pathway [1618]. Designing indirect FXa inhibitors that activate antithrombin is challenging and has been achieved primarily with heparin-based molecules [14,19], although efforts are afoot to design effective non-saccharide-based antithrombin activators [15,20,21]. In contrast, designing direct inhibitors of FXa has been much more productive and has led to several clinically relevant peptidomimetics, e.g., rivaroxaban [22], apixaban [23], DPC423 [24], and others, that target FXa’s active site.

A large number of scaffolds have been studied for direct inhibition of FXa including the aminopiperidines [25], piperazines [26], diaminocycloalkanes [27], lactams [28,29], oxazolidinones [30], amino acids (e.g., glycine, proline, and β-aminoproionate) [31,32], anthranilamides [33], isoxazoles [34], pyrazoles [24,35], indazoles [36], indoles [37,38], and dihydropyrazolopyridinones [23,39]. The overall philosophy in the design of these scaffolds is to have a three-component system, which includes a core scaffold and two hydrophobic arms that provide a non-linear geometry considered important for FXa recognition. In addition, a key design principle has also been flexibility of the core scaffold. The core scaffolds studied to date can be classified into either a highly flexible class or a fairly rigid class.

It is known that trypsin’s active site is much open than FXa’s, which in turn possesses an active that is more open than that of thrombin. This allows trypsin to work upon practically any arginine containing sequence, while thrombin prefers a fairly rigid proline containing sequence. Thus, we hypothesized that an intermediate level of flexibility in the core scaffold will engineer high selectivity for FXa inhibition. In this work, we studied several core scaffolds with variable flexibility to design potent direct FXa inhibitors. Of the scaffolds studied, the 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (THIQ3CA) scaffold possessing an intermediate level of flexibility was found to be the best. Molecular modeling-based identification of an initial hit 23 was transformed into the discovery of a potent direct FXa inhibitor 47 with a KI of 135 nM. This molecule selectively targeted FXa in comparison to the enzymes of the coagulation and digestive systems, and doubled clotting times at 17 – 20 μM, which are comparable to those of the inhibitors being studied in clinical trials [40]. The work with a library of 39 molecules has revealed interesting structure–activity relationships (SAR) that identify optimal structural units for engineering good FXa inhibition properties. The lead THIQ3CA dicarboxamide 47 is expected to serve as an excellent starting point for further advanced design.

2. Results and Discussion

2.1. Designing the Tetrahydroisoquinoline-3-Carboxylic Acid Scaffold as a Potential Factor Xa Inhibitor Scaffold

The glycine and β-aminopropionate-based inhibitors are examples of a highly flexible core scaffold, while aromatic scaffolds, e.g., aminobenzoic acids, pyrazoles, and indazoles, are examples of fairly rigid scaffolds. An intermediate level of flexibility in the core scaffold has not been studied to date for direct FXa inhibition. A large number of scaffolds possess intermediate level of flexibility including tetrahydroisoquinoline (THIQ), tetrahydroquinoline, tetrahydroquinazoline, dihydrocoumarin, and dihydroindole. In each of these, the five- or six-membered rings are flexible, yet full flexibility is restricted due to ring fusion. Of these, we focused on the THIQ scaffold, which is a widely explored privileged structure [15, 4144]. To enable a non-linear structure for FXa recognition, we introduced a carboxylic acid group at the 3-position of the THIQ scaffold to arrive at THIQ3CA scaffold.

The designed THIQ3CA scaffold was first investigated in silico to assess its potential in binding in factor Xa active site as well as to identify a potential inhibitor for synthesis. A library of THIQ3CA structures was prepared by introducing substituents at positions 2 and 3. A large group of aromatic and non-aromatic carboxylic acid and amine arms at positions 2 and 3 were studied to identify potential ‘hits’ (see Supplementary Information). The virtual library of resulting THIQ3CA dicarboxamides was then computationally docked into active site of human FXa and scored using genetic algorithm-based screening technique developed in our laboratory earlier [15]. As positive controls, two well-established direct FXa inhibitors, apixaban and rivaroxaban, were also studied in an identical manner.

The GOLD-based docking and scoring strategy identified several hits, of which THIQ3CA dicarboxamide 23, containing a 5-chlorothiophen-2-yl moiety (also present in rivaroxaban), was identified as a promising candidate. Figure 1A shows a comparison of the predicted bound form of 23 with apixaban. Both molecules adopted a rather similar binding mode in which the 5-chlorothiophenyl-carbonyl group was found to be oriented toward the S1 subsite and the 4-(piperidin–2–one)–N1–methylaniline moiety oriented into the S4 subsite. Comparison of the binding geometry with rivaroxaban suggested similar corresponding features (not shown). THIQ3CA dicarboxamide 23 was synthesized in three high yielding steps and evaluated for its direct human FXa inhibition potential (see below). An IC50 of 55.9 μM, corresponding to a KI of 28 μM, was measured validating the potential of the THIQ3CA core scaffold in direct human FXa inhibition investigation.

Figure 1.

Figure 1

Virtual screening of a library of potential THIQ3CA-based inhibitors docked and scored onto the active site of FXa (PDB: 2P16) using GOLD resulted in the design of 23 (A: GOLD score of 69.3, Carbons in pink) that appeared to mimick the binding of well known potent FXa inhibitors. The virtual library of THIQ3CA–based dicarboxamides contained structural modifications in arms AN and AC. Apixaban (GOLD score of 93.7, Carbons in blue) and rivaroxaban (GOLD score of 87.2, structure not shown) were also docked and scored as positive controls. Sytematic structural modifications led to the design of the most potent THIQ3CA dicarboxamide 47 (B: GOLD score of 77.2, Carbons in yellow). See text for details.

2.2. Synthesis of THIQ3CA-Based Potential FXa Inhibitors

Based on the structure of 23, a library of THIQ3CA dicarboxamides was designed. The synthesis of the targeted THIQ3CA analogs was achieved using a facile three-step strategy of amidation, deprotection, and amidation that relied on the availability of the two arms, AN and AC, at positions 2 and 3, respectively [45]. Each AN structure designed as a part of the library was commercially available and was introduced using standard amidation reaction (see Scheme 1). In contrast, arm AC at position 3 of THIQ3CA required pre-assembly. This arm was synthesized by aromatic nucleophilic substitution in which 4–bromoaniline derivatives 4a4c were reacted with either valerolactam 5a or morpholin-3-one 5b to quantitatively give the corresponding 4–substituted aniline derivatives 8a8g [46]. Likewise, substituted anilines 8h8j containing the piperazine side chain were also included in the study (see Supplementary Information Scheme S1 for synthetic and characterization details).

Scheme 1.

Scheme 1

a) Appropriate amine (8a–8g), HOBt.H2O, DMAP, EDCI, CH2Cl2, rt/overnight, 73–89%, b) For Cbz-protected intermediates: 10% Pd(OH)2, (1:1) CH3OH: t-butanol, H2, rt/overnight, 65–72%. For Boc-protected intermediates: (1:1) TFA:CH2Cl2, rt/4 h, 75%, c) Appropriate organic acid, EDCI, DMAP, CH2Cl2, rt/overnight, 70–95%.

To introduce either arm AN or AC through an amidation reaction, the THIQ3CA core was first appropriately protected, which involved either esterification of the 3-carboxylic acid or t-butoxycarbonylation (Boc group) or carbonylbenzyloxylation (Cbz group) of the ring nitrogen. For example, THIQ3CAs 911 were amidated using amines 8a8g to yield Boc- or Cbz-protected THIQ3CA mono-carboxamides 1221 in 73–89% yield (Scheme 1). The Boc or Cbz protecting group of 1221 was then removed using either mild acid [47] or catalytic hydrogenation [48], respectively, to afford the corresponding free form in 65–75% yield. The free form of 1221 was then amidated at the 2 position with twenty one carboxylic acids in the presence of EDCI to yield the targeted THIQ3CA dicarboxamides 2249 in 70–95% yield (Scheme 1). This strategy was also exploited for the synthesis of THIQ3CA dicarboxamides 5055, 59, and 62 that contain additional variations on arms AN and AC (see Supplementary Information Scheme S2 and S3).

To assess the effect of flexibility for FXa inhibition, we targeted three core scaffolds that afford greater flexibility than the THIQ3CA scaffold as well as can help assess the importance of configurational geometry. These scaffolds were: 1) a ring opened variant of THIQ3CA core, the phenylalanine (PA) derivative 62; 2) mono-cyclic dicarboxamides 6973 belonging to the piperidine–3–carboxylic acid (P3CA) class; and 3) mono-cyclic dicarboxamides 78, 79 and 81 belonging to the piperidine–2–carboxylic acid (P2CA) class (Scheme 2). These molecules were synthesized in a manner similar to the THIQ3CA scaffold using the amidation, deprotection, and amidation strategy.

Scheme 2.

Scheme 2

a)Benzyl chloroformate, Et3N, THF, rt/5 h, 70–73 %, b) 8b or 8c, HOBt.H2O, DMAP, EDCI, CH2Cl2, rt/overnight, 73–89%, c) For Cbz-protected intermediates: 10% Pd(OH)2, (1:1) CH3OH: t-butanol, H2, rt/overnight, 65–72%. For Boc-protected intermediates: (1:1) TFA:CH2Cl2, rt/4 h,75%, d) Appropriate organic acid, EDCI, DMAP, CH2Cl2, rt/overnight, 70–95%.

To understand the importance of the aromatic ring of the THIQ3CA scaffold, and yet mimic its flexibility, we targeted 1) a partially unsaturated, cyclic ring system, i.e., (S)-1,2,3,6–tetrahydropyridine–2–carboxylic acid (THP2CA) and 2) a bridged system, i.e., (1R,3S,4R)-2-azabicyclo[2.2.1]heptane-3-carboxylic acid (ABH3CA) (Scheme 3). The synthesis of either scaffold started with the Boc-protected carboxylic acid 82 or 85, which was converted in three steps to dicarboxamide 84 or 87, respectively, in excellent yield.

Scheme 3.

Scheme 3

a) 8b, HOBt.H2O, DMAP, EDCI, CH2Cl2, rt/overnight, b) (1:1) TFA:CH2Cl2, rt/4 h, c) 4-Chlorophenyl acetic acid, EDCI, DMAP, CH2Cl2, rt/overnight, 70–95%.

Overall, we synthesized twenty eight THIQ3CA, one PA, five P3CA, three P2CA, one THP2CA and one ABH3CA dicarboxamides using a simple three-step protocol. The molecules were purified using standard flash chromatography system and were characterized using 1H and 13C NMR spectroscopy and ESI-MS (see Supplementary Information).

2.3. Inhibition Potential of the Library of FXa Inhibitors

Direct inhibition of FXa was measured by a chromogenic substrate hydrolysis assay in 20 mM TrisHCl buffer, pH 7.4, at 37 °C, as reported earlier [49]. In this assay, hydrolysis of the substrate by FXa results in a linear increase in absorbance at 405 nm, the slope of which corresponds to residual enzyme activity. The change in residual enzyme activity as a function of the concentration of the potential inhibitor is plotted on a logarithmic scale and fitted by the logistic dose-response relationship 2 to derive the potency (IC50), efficacy (ΔY = YMY0) and Hill Slope (HS) of inhibition [18,49]. Figure 2 shows representative inhibition profiles for 20, 47, 49, and 51, which are THIQ3CA derivatives, and 62 and 81, which are PA and P2CA derivatives, respectively. Similar profiles were measured for other THIQ3CA, P2CA, P3CA, THP2CA, and ABH3CA derivatives, except for the variation in potency of inhibition. The HS of inhibition was found to be in the range of 0.7 to 1.2, which suggests absence of the complications arising from non-specific binding or aggregation. The IC50 values of the FXa inhibitors studied in this work were found to exhibit a wide range of activity (high μM to nM), which provide valuable structure – activity relationships.

Figure 2.

Figure 2

Direct inhibition of FXa by designed THIQ3CA and related dicarboxamides. The inhibition of human FXa was determined spectrophotometrically through the chromogenic substrate hydrolysis assay at pH 7.4 and 37°C. Solid lines represent sigmoidal fits to the data to obtain IC50, YM, Y0, and HS, as described in the Experimental Methods section. Experiments were performed in duplicate or triplicate (SE < 20%). See text for details.

2.4. Structural Optimization of Arm AC of the THIQ3CA Scaffold

Our early molecular modeling studies suggested that arm AC at the 3-position of the THIQ3CA core structure bound in the S4 subsite of the FXa active site. An intermediate 13, synthesized early in the library and containing N-(N-methylanilin-4-yl)piperidin-2-one as the AC arm, had shown a reasonable IC50 of 16.4 μM. This formed the starting point for studying the S4 subsite substitutions (Table 1). To optimize the structure of the AC arm, two levels of structural modifications were introduced. These included 1) variation in the carboxamide N-substituent and 2) variation in the para-substituent of the aniline moiety. Replacing the N-methyl group of 13 with ethyl (inhibitor 14) and isopropyl (inhibitor 15) groups resulted in no significant change, however, N-demethylation as in 12 reduced the IC50 by 1.7–fold suggesting the possibility of the formation of a hydrogen bond. This was however not substantiated by molecular modeling (not shown) and we speculated that the alkyl group on the nitrogen may be inducing a conformational preference that is not favorable for binding in the S4 subsite.

Table 1.

Optimization of the AC arm (position 3) of THIQ3CA scaffold for FXa inhibition.

graphic file with name nihms389693u1.jpg
Inhibitor R1 R2 IC50 (μM)
12 -H graphic file with name nihms389693t1.jpg 9.9 ± 0.2
13 -CH3 graphic file with name nihms389693t2.jpg 16.4 ± 0.4
14 -CH2CH3 graphic file with name nihms389693t3.jpg 20 ± 1
15 -CH(CH3)2 graphic file with name nihms389693t4.jpg 19 ± 2
18 -CH3 graphic file with name nihms389693t5.jpg 12.8 ± 0.2
19 -CH2CH3 graphic file with name nihms389693t6.jpg 19 ± 6
20 -H graphic file with name nihms389693t7.jpg 6.0 ± 0.8
50 -H -Cl >500
53 -H graphic file with name nihms389693t8.jpg >500
54 -H graphic file with name nihms389693t9.jpg >500
55 -H graphic file with name nihms389693t10.jpg >500

To optimize the para–substituent of the aniline moiety, we relied on the extensive literature that abounds in such optimizations [2, 16, 50]. The moieties preferred at this position include the piperidone, morpholinone, piperidine, piperazine and other rings. Yet, introducing piperidine and piperazine moieties at the para position of the aniline was catastrophic. The piperidine–containing derivative 53 and the piperazine–containing derivatives 54 and 55 displayed an un-quantifiable IC50 of >500 μM (Table 1). Molecular modeling suggested that the para substituent helps orient the adjacent aromatic ring for better interaction with Tyr99, Phe174, and Trp215 of the S4 subsite (not shown), in a manner similar to that observed with rivaroxaban and apixaban [23,30]. Thus, we studied the morpholin-3-one moiety (derivative 20, Table 1), which exhibited a much improved IC50 of 6.0 μM. The results indicated a significant contribution of the carbonyl group of morpholin-3-one for the THIQ3CA inhibitors.

2.5. Structural Optimization of Arm AN of the THIQ3CA Scaffold

Molecular modeling suggested that the AN arm at position 2 of the THIQ3CA core fits into S1 subsite of the FXa active site. The initial hit 13 (IC50 = 16.4 μM) containing the Cbz group as the AN arm was used as the starting point for studying the S1 subsite substitutions. Considering that the AN arm of 13 is structurally less defined than its AC arm, we decided to explore a wider range of moieties to replace the Cbz group. Thus, optimal chain length of AN, the nature of its aromatic ring, and the position and number of substituents on the aromatic ring were studied.

Replacing the AN arm of 13 with thiophenyl carbonyl (22), 5-chlorothiophenyl carbonyl (23), p-chlorophenyl carbonyl (24), p-methylphenyl carbonyl (25) or p-methoxyphenyl carbonyl (26) arm resulted in 1.2 – 12.7-fold reduction in potency (Table 2). Extending the length of the linker by one carbon atom resulted in significant improvement in IC50. Dicarboxamide 27 containing a 4-chlorophenyl acetyl side chain exhibited an IC50 of 1.3 μM, which was the first high affinity inhibitor (Ki = 0.65 μM) designed in the THIQ3CA series. Yet, most subsequent modifications failed to enhance potency further. For example, one or more electron donating (–CH3, –OCH3) as well as one or more electron withdrawing (–Cl, –CF3, –NO2) substituents on the phenylacetyl side chain weakened the potencyby 1 – 262-fold (Table 2).

Table 2.

Optimization of the AN arm (position 2) of THIQ3CA scaffold containing the piperidone moiety in the AC arm for FXa inhibition.

graphic file with name nihms389693u2.jpg
Inhibitor R3 IC50 (μM)
16 graphic file with name nihms389693t11.jpg 221 ± 19
22 graphic file with name nihms389693t12.jpg 42 ± 2
23 graphic file with name nihms389693t13.jpg 56 ± 2
24 graphic file with name nihms389693t14.jpg 197 ± 39
25 graphic file with name nihms389693t15.jpg 209 ± 31
26 graphic file with name nihms389693t16.jpg 19 ± 2
27 graphic file with name nihms389693t17.jpg 1.3 ± 0.1
28 graphic file with name nihms389693t18.jpg 14 ± 3
29 graphic file with name nihms389693t19.jpg 34 ± 1
30 graphic file with name nihms389693t20.jpg 85 ± 19
31 graphic file with name nihms389693t21.jpg 335 ± 50
32 graphic file with name nihms389693t22.jpg 246 ± 58
33 graphic file with name nihms389693t23.jpg 18 ± 2
34 graphic file with name nihms389693t24.jpg 1.3 ± 0.2
38 graphic file with name nihms389693t25.jpg 103 ± 8

Considering the observations made with the AC structural variants, we studied the morpholin-3-one containing THIQ3CA derivatives also for optimization of the AN arm. Table 3 shows the results obtained in the morpholin-3-one series. Replacing the Cbz group of parent 18 (IC50 = 12.8 μM) with thiophenyl carbonyl (40), p-methoxyphenyl propanoyl (43), or 2,4-dimethoxyphenyl propanoyl (48) resulted in considerable loss of activity. Yet, as in the piperidone series described above, introducing a 4-chlorophenyl acetyl side chain for the Cbz group resulted in an IC50 of 1.1 μM.

Table 3.

Optimization of the AN arm (position 2) of THIQ3CA scaffold containing the morpholinone moiety in the AC arm for FXa inhibition.

graphic file with name nihms389693u3.jpg
Inhibitor R1 R3 IC50 (μM)
40 -CH3 graphic file with name nihms389693t26.jpg 36 ± 3
41 -CH3 graphic file with name nihms389693t27.jpg 1.1 ± 0.1
42 -H graphic file with name nihms389693t28.jpg 5.4 ± 0.9
43 -CH3 graphic file with name nihms389693t29.jpg 190 ± 16
45 -CH3 graphic file with name nihms389693t30.jpg >500
48 -CH3 graphic file with name nihms389693t31.jpg >500
49 -H graphic file with name nihms389693t32.jpg 2.9 ± 0.4

Analysis of the above results indicated that the best inhibitors carried a 7–atom arm AN (at position 2), e.g., 13, 26, 27, 28, and 41. Within this category, a 7-atom arm containing a small, lipophilic electron–withdrawing group (–Cl) is optimal. This group appears to be the p-chlorophenylacetyl moiety, as exemplified by a considerable increase in affinity observed with 27, 34 and 41, and a loss in affinity for derivative 49 (Tables 2 and 3). Thus, FXa appears to preferentially recognize the chlorophenyl moiety in the THIQ3CA series of inhibitors.

2.6. Rational Design of Analog 47

The majority of inhibitors described above possessed (S)-chirality at position 3. To assess whether an (R)-isomer would be better, we studied THIQ3CA derivative 35, which is a stereoisomer of 27 (IC50 = 1.3 μM). This stereochemical inversion resulted in a 1.5–fold increase in the activity (Table 4). This led to a hypothesis that introducing optimal features derived on arms AN and AC into a single THIQ3CA derivative may lead to the most potent FXa inhibitor designed so far. Thus, analog 47 was designed, which contained a morpholin-3-one moiety, a p-chlorophenylacetyl unit, (R)-chirality at the 3-position, and NH-containing carboxamide at the 3-position. THIQ3CA derivative 47 displayed an IC50 of 270 nM (KI = 135 nM) and represents the highest potency observed in the THIQ3CA series.

Table 4.

Simultaneous optimization of both AN AC arms of THIQ3CA scaffold for optimal FXa inhibition.

graphic file with name nihms389693u4.jpg
Inhibitor R1 X Y R3 Chirality (*) IC50 (μM)
35 -CH3 -CH2- -C(O)- graphic file with name nihms389693t33.jpg (R)- 0.9 ± 0.1
36 -CH3 -CH2- -C(O)- graphic file with name nihms389693t34.jpg (R)- 311 ± 56
37 -H -CH2- -C(O)- graphic file with name nihms389693t35.jpg (S)- 13.3 ± 1.4
46 -CH2CH3 -O- -C(O)- graphic file with name nihms389693t36.jpg (S)- 1.6 ± 0.2
47 -H -O- -C(O)- graphic file with name nihms389693t37.jpg (R)- 0.27 ± 0.03
52 -CH3 -CH2- -SO2- graphic file with name nihms389693t38.jpg (S)- 151 ± 36

The design of 47 reflects a truly additive phenomenon. Inhibitor 47 exhibits ~60–fold decrease in IC50 relative to the starting inhibitor 13 (IC50 = 16.4 μM), which can be rationalized from the 1.5–fold (morpholin-3-one) × 2–fold (unsubstituted carboxamide) × 1.5–fold (stereochemical inversion) × 13–fold (4-chlorophenylacetyl moiety) increases found independently earlier. This is an interesting observation and appears to have considerable consistency across the series of molecules studied here.

2.7. THIQ3CA Core is the Most Optimal Scaffold

Based on the number of flexible carbons constituting the core scaffold, we reasoned that the open-chain PA core will be more flexible than the six-membered cyclic P2CA and P3CA cores, which in turn will exhibit greater flexibility than the unsaturated or bridged THP2CA and ABH3CA cores. The THIQ3CA core was likely to be similar to the unsaturated or bridged cores in terms of conformational flexibility. Table 5 displays the potency of selected PA, P2CA, P3CA, THP2CA, ABH3CA, and THIQ3CA inhibitors. Overall, the potency of PA, P2CA, and P3CA dicarboxamide inhibitors was found to be weak (23 – 371 μM), while that for THP2CA, ABH3CA and THIQ3CA inhibitors were higher (0.27 – 56 μM). Comparison of the unsaturated or bridged six-membered scaffold with the THIQ3CA scaffold shows that the latter is the most optimal structure. For example, THP2CA derivative 84 and ABH3CA derivative 87 exhibit IC50s of 9.1 and 10 μM, which are 33–37-fold greater than the 0.27 μM IC50 of the corresponding THIQ3CA derivative 47 (Table 5).

Table 5.

Comparison of the FXa inhibition potential of different scaffolds.

graphic file with name nihms389693u5.jpg
graphic file with name nihms389693u6.jpg
graphic file with name nihms389693u7.jpg
Inhibitor A IC50 (μM) Inhibitor B IC50 (μM) Inhibitor C IC50 (μM)



27 graphic file with name nihms389693t39.jpg 1.3 ± 0.1 23 graphic file with name nihms389693t40.jpg 56 ± 2 47 graphic file with name nihms389693t41.jpg 0.27 ± 0.03
59 graphic file with name nihms389693t42.jpg 7.8 ± 0.4 78 graphic file with name nihms389693t43.jpg 358 ± 16 81 graphic file with name nihms389693t44.jpg 23 ± 2
62 graphic file with name nihms389693t45.jpg 54 ± 5 79 graphic file with name nihms389693t46.jpg 99 ± 7 84 graphic file with name nihms389693t47.jpg 9 ± 2
72 graphic file with name nihms389693t48.jpg 371 ± 59 69 graphic file with name nihms389693t49.jpg 69 ± 7 87 graphic file with name nihms389693t50.jpg 10 ± 3



Within these broad results, we noticed interesting structure-activity dependence across the different core scaffolds studied here. Nearly all 5-chloro-thiophenyl containing derivatives, e.g., 23 and 40, display weak inhibition potency in comparison to most p-chlorophenyl containing derivatives, e.g., 27 and 41 (Tables 2 and 3). This was against our expectation based on the well established high affinity of rivaroxaban, which contains the 5-chloro-thiophenyl side chain [22, 30]. Another interesting observation was that morpholin-3-one ring consistently induces higher potency than the piperid-2-one ring. For example, morpholin-3-one-containing THIQ3CA derivatives 18, 20, and 40 have better activity than the corresponding piperid-2-one-containing derivatives 13, 12, and 23, respectively (see Tables 1, 2 and 3).

The above results suggest that the THIQ3CA core scaffold was the most optimal for FXa inhibition following optimization of its two arms AN and AC. Yet, the THP2CA and ABH3CA scaffolds are interesting. These are scaffolds are new, readily synthesizable and are relatively unexplored. Screening of a wider chemical space would help better understand their true potential for FXa inhibition.

2.8. Dicarboxamide 47 Selectively Inhibits Factor Xa in Comparison to Other Proteases of the Coagulation and Digestive Systems

Acritical goal of the molecular modeling-based rational design of THIQ3CA inhibitors was selectivity for targeting factor Xa. This enzyme is a trypsin-like serine protease with considerable homology with enzymes of the coagulation and digestive systems including FIIa, FVIIa, FIXa, FXIa, FXIIa, trypsin and chymotrypsin. Chromogenic substrate hydrolysis assays for each of these enzymes were performed in a manner similar to that for FXa. The assays were conducted at 37 °C using literature reported buffer systems that are as close to physiological conditions as possible (see Experimental Methods for details). Initial screening was performed at a high, fixed concentration of the inhibitor (100 – 1000 μM) and fractional residual enzyme activity was measured from the initial rate of hydrolysis in the presence of the inhibitor to that in its absence.

The THIQ3CA inhibitors showed high selectivity for FXa inhibition. No coagulation enzyme was found to be inhibited by THIQ3CA inhibitors at concentrations less than 100 μM. With regard to digestive enzymes, moderate inhibition of chymotrypsin (IC50 = 5–100 μM) was noted with 4-chlorophenyl side chain containing inhibitors. The most potent FXa inhibitor 47 demonstrated a selectivity of at least 1852–fold over FIIa, FVIIa, FIXa, FXIa, and FXIIa (Table 6). Against trypsin and chymotrypsin, the selectivity of 47 was found to be at least 370- and 279-fold, respectively. Few rationally designed inhibitors have been able to achieve such high selectivity for factor Xa against other homologous enzymes [51, 52].

Table 6.

Selectivity of inhibition by THIQ3CA dicarboxamide 47.

Protease IC50(μM) Selectivity Index
Human FIIa >500 >1852
Human FVIIa >1000 >3703
Human FIXa >1000 >3703
Human FXIa >500 >1852
Human FXIIa >500 >1852
Bovine Trypsin >100 >370
Bovine Chymotrypsin 75.3 ± 13.4 279

2.9. Prolongation of Plasma Clotting Times by THIQ3CA, THP2CA and ABH3CA Inhibitors

Clotting assays, prothrombin and activated partial thromboplastin time (PT and aPTT, respectively), are routinely used to assess anticoagulation potential of new enzyme inhibitors in an in vitro setting [18,53,54]. Whereas PT measures the effect of an inhibitor on the extrinsic pathway of coagulation, aPTT measures the effect on the intrinsic pathway. The prolongation of the human plasma clotting time as a function of the concentration of the inhibitors followed a pattern typical of other well-studied anticoagulants (data not shown), except for the range of active concentrations. Table 7 lists the concentrations of selected potent FXa inhibitors required to double the PT and aPTT. Overall, for the inhibitors studied, doubling the PT required 17–2347 μM concentration, while doubling of aPTT required 20–810 μM suggesting that most inhibitors essentially affect both pathways equally (Figure 3). This is to be expected because of their selectivity for targeting FXa, which is an enzyme of the common pathway. The inhibitors studied included THIQ3CA derivatives 20, 27, 34, 42, 47, and 49, and THP2CA/ABH3CA derivatives 84 and 87. Inhibitor 47 doubled the PT clotting time at 17.1 μM, which is comparable to razaxaban (3.8 μM) and DPC423 (4.9 μM), two direct FXa inhibitors being pursued in clinical trials [51].

Table 7.

Effect of designed FXa dicarboxamide inhibitors on human plasma clotting time.

Inhibitor PT (μM) aPTT (μM)
20 1387 NDa
27 71 80
34 219 260
42 2160 NDa
47 17.1 20.2
49 645 614
84 952 810
87 2347 NDa
a

Not determined.

Figure 3.

Figure 3

Prolongation of clotting time as a function of concentration of designed THIQ3CA and related dicarboxamides in either prothrombin time assay (PT) (A) or activated partial thromboplastin time assay (aPTT) (B). Solid lines are trend lines from which the concentration necessary to double clotting time was deduced. Clotting assays were performed in duplicate (SE ≤ 10%) as described in Experimental Methods.

3. Significance

Our work investigates the dependence of direct FXa inhibition on the flexibility of core scaffold and attempts to optimize the structure of two arms AN and AC of a bifunctional cyclic ring system. We discovered interesting structural preference for both arms and a fine additive relationship that culminated in the design of THIQ3CA analog 47, which displays direct inhibition IC50 of 270 nM (KI = 135 nM). Synthetically, the FXa inhibitors were readily assembled using three high yielding steps, which required room temperature conditions and avoided harsh reactions. Our work suggests that the morpholin-3-one ring engineers better inhibitor potential than any other ring system; that the nitrogen of the carboxamide group at position-3 should be optimally unsubstituted; that a (R)–stereochemistry at position-3 is more preferred than its (S)-enantiomer; and that a 7-atom AN arm containing a para-chloro substituted aromatic ring is critical for potent, direct FXa inhibition. It is also worth mentioning that the AN and AC arms should be adjacent to each other (1,2-substitution) rather than one carbon away (1,3-disubstitution). These conclusions may help design more potent molecules.

4. Experimental Methods

4.1. Chemicals, Reagents, and Analytical Chemistry

Anhydrous CH2Cl2, THF, CH3CN, DMF, toluene, and acetone were purchased from Sigma-Aldrich (Milwaukee, WI) or Fisher (Pittsburgh, PA) and used as such. Other solvents used were of reagent gradient and used as purchased. Analytical TLC was performed using UNIPLATE silica gel GHLF 250 um pre-coated plates (ANALTECH, Newark, DE). Column chromatography was performed using silica gel (200–400 mesh, 60 Å) from Sigma-Aldrich. Chemical reactions sensitive to air or moisture were carried out under nitrogen atmosphere in oven-dried glassware. Reagent solutions, unless otherwise noted, were handled under a nitrogen atmosphere using syringe techniques. Flash chromatography was performed using Teledyne ISCO (Lincoln, NE) Combiflash RF system and disposable normal silica cartridges of 30–50 μ particle size, 230–400 mesh size and 60 Å pore size. The flow rate of the mobile phase was in the range of 18 to 35 ml/min and mobile phase gradients of ethyl acetate/hexanes and CH2Cl2/CH3OH were used to elute compounds.

4.2. Chemical Characterization of Compounds

1H and 13C NMR were recorded on Bruker-400 MHz spectrometer in either CDCl3, CD3OD, or acetone-d6. Signals, in part per million (ppm), are either relative to the internal standard (tetramethyl silane, TMS) or to the residual peak of the solvent. The NMR data are reported as chemical shift (ppm), multiplicity of signal (s= singlet, d= doublet, t= triplet, q= quartet, dd= doublet of doublet, m= multiplet), coupling constants (Hz), and integration. Compounds with dicarboxamide functionalities exhibit amide rotamerism resulting in complexity of NMR signals. ESI-MS of compounds were recorded using Waters Acquity TQD MS spectrometer in positive ion mode. Samples were dissolved in methanol and infused at a rate of 20 μL/min. Mass scans were obtained in the range of 200–700 amu with a scan time of 1 s and a scan rate of 500 amu/s. The capillary voltage was varied between 3 and 4 kV, while the cone voltage ranged from 38 to103 V. Ionization conditions were optimized for each compound to maximize the ionization of the parent ion. Generally, the extractor voltage was set to 3 V, the Rf lens voltage was 0.1 V, the source block temperature was set to 150 °C, and the desolvation temperature was about 250 °C. The purity of each final compound was greater than 95% as determined by uPLC-MS.

4.3. Proteins

Human plasma proteinases including FIIa, FXa, FXIa, FIXa, FVIIa, and recombinant tissue factor was obtained from Haematologic Technologies (Essex Junction, VT). FXIIa was purchased from Enzyme Research Laboratories (South Bend, IN). Bovine α-chymotrypsin and bovine trypsin were obtained from Sigma-Aldrich (St. Louis, MO). The substrates Spectrozyme TH, Spectrozyme FXa, Spectrozyme FXIIa, Spectrozyme FIXa, Spectrozyme VIIa, and Spectrozyme CTY were obtained from American Diagnostica (Greenwich, CT). Factor XIa substrate, S-2366, trypsin substrate, S-2222, were obtained from Diapharma (West Chester, OH). Factor Xa and FVIIa were prepared in 20 mM TrisHCl buffer, pH 7.4, containing 100 mM NaCl, 2.5 mM CaCl2, 0.1% PEG8000, and 0.02% Tween80. FIXa was prepared in 20 mM TrisHCl buffer, pH 7.4, containing 100 mM NaCl, 2.5 mM CaCl2, 0.1% PEG8000, 0.02% Tween80, and 33% v/v Ethyleneglycol. Other enzymes were prepared in 50 mM TrisHCl buffer, pH 7.4, containing 150 mM NaCl, 0.1% PEG8000, and 0.02% Tween80.

4.4. Modeling Factor Xa and the Virtual Library of (S)-THIQ3CA Dicarboxamides

Sybyl 8.1 (Tripos Associates, St. Louis, MO) was used for modeling of FXa–inhibitor complexes. The structure of human FXa (ID: 2P16) [23] was acquired from the Protein Data Bank (www.rcsb.org). To prepare the protein structure for modeling experiments, hydrogen atoms were added, while inorganic ions and water molecules were removed. Individual atoms were assigned Gasteiger–Hückel charges. Energy minimization was then performed using a Tripos force field so as to reach a terminating gradient of 0.5 kcal/mol Å2 or a maximum of 100,000 iterations.

(S)–THIQ3CA was modified in silico at positions 2 and 3 with appropriate substituents to “synthesize” a virtual chemical library of nearly 150 dicarboxamide derivatives. The different combinations at 2- and 3-positions included substituted derivatives of aromatic carboxylic acids and aromatic amines, respectively. These acids and amines were further substituted at varying positions with halogen, t–butyl, cyclopropyl, methoxy, amino–methyl, substituted phenyl, substituted imidazole, pyridine, pyridine–2–one, N–oxide pyridine, N-methyl piperazine, or cyclic amides such as piperidone and morpholinone (See Supplementary Information). The resulting molecules were assigned Gasteiger – Hückel charges and were then energetically minimized using a Tripos force field until a terminating gradient of 0.5 kcal/mol Å2 or a maximum of 100,000 iterations.

4.5. Docking and Scoring

Docking of the (S)-THIQ3CA dicarboxamides onto the active site of FXa was performed with GOLD 5.1 (Cambridge Crystallographic Data Center, UK). The docking experiment was performed as reported earlier [15]. The binding site in human FXa was defined to include all atoms within 6 Å of the co–crystallized ligand. The docking protocol was validated by docking ligand for which the adopted docking pose was found to have RMSD < 1 Å relative to that reported in the crystal structure [23]. Docking was driven by the GOLD scoring function, while for ranking the docked solutions; a linear, modified form of the same scoring function Eq. 1 was used, as reported earlier [15]. In this equation, HBEXT and VDWEXT are the “external” (nonbonded interactions taking place between the ligand and the target protein) hydrogen bonding and van der Waals terms, respectively.

GOLDScore=HBEXT+1.375×VDWEXT Eq. 1

4.6. FXa Inhibition Studies

Direct inhibition of FXa was measured by a chromogenic substrate hydrolysis assay, as reported earlier [49] using a microplate reader (FlexStation III, Molecular Devices) at wavelength of 405 nm and incubating temperature of 37 °C. Generally, each well of the 96-well microplate had 185 μL pH 7.4 buffer to which 5 μL potential FXa inhibitor (or solvent reference) was added, to which 5 μL FXa (stock conc. 43.5 nM) was further added. After 10 min incubation at 37 °C, 5 μL FXa substrate (stock conc. 5 mM) was rapidly added and the residual FXa activity was measured from the initial rate of increase in absorbance at 405 nm. The concentration of the organic solvent in which the inhibitors were dissolved was maintained constant and was less than 2.5% (v/v). Stocks of FXa inhibitors were prepared at 20 mM concentration and then serially diluted to give twelve different aliquots spanning a range of 0.015–500 μM in the plate wells. Each inhibitor was studied in duplicate or triplicate at each concentration. Relative residual FXa activity at each concentration of the inhibitor was calculated from the ratio of FXa activity in the presence and absence of the inhibitor. Logistic Eq. 2 was used to fit the dose-dependence of residual proteinases activity to obtain the potency (IC50) and efficacy (ΔY) of inhibition. In this equation, Y is the ratio of residual factor Xa activity in the presence of inhibitor to that in its absence (fractional residual activity), YM and Y0 are the maximum and minimum possible values of the fractional residual proteinase activity, IC50 is the concentration of the inhibitor that results in 50% inhibition of enzyme activity, and HS is the Hill slope (which was between 0.7 and 1.2 in all measurements). Nonlinear curve fitting resulted in YM, Y0, IC50 and HS values. The reported IC50 is an average of the two or three measurements with standard error (SE) of <20%. The efficacy of inhibition ΔY was found to be >80% for each studied FXa inhibitor.

Y=Y0+YM-Y01+10(Log[I]0-LogIC50)(HS) Eq. 2

4.7. Inhibition of Proteases of the Coagulation and Digestive Systems

The potential of the FXa inhibitors against coagulation enzymes including FIIa, FVIIa, FIXa, FXIa, and FXIIa, and digestive enzymes including trypsin and chymotrypsin was performed using chromogenic substrate hydrolysis assays reported in the literature [55]. These assays were performed using substrates appropriate for the enzyme being studied under conditions closest to the physiological condition (37 °C and pH 7.4), except for FIIa, which was performed at 25 °C and pH 7.4. For selectivity analysis, a single concentration point assays was utilized in which 0.5 – 1 mM FXa inhibitor was tested in duplicate. The fractional residual enzyme activity was measured and if found to be less than 50%, the inhibition profile was measured over a range of inhibitor concentrations to determine the IC50 of the enzyme–inhibitor complex. The KM of the substrate for its enzyme was used to identify the concentration of the substrate to be used for inhibition studies. The concentrations of enzymes and substrates in microplate cells were: 6 nM and 50 μM for FIIa; 0.765 nM and 345 μM for FXIa; 5 nM and 125 μM for FXIIa; 89 nM and 850 μM for FIXa; 8 nM and 1000 μM for FVIIa (along with 40 nM recombinant tissue factor); 72.5 ng/ml and 80 μM for bovine trypsin; and 500 ng/ml and 240 μM for bovine chymotrypsin.

4.8. Prothrombin Time (PT) and Activated Partial Thromboplastin Time (aPTT)

Clotting time was measured in a standard one-stage recalcification assay with a BBL Fibrosystem fibrometer (Becton-Dickinson, Sparles, MD), as described previously [18]. For PT assays, thromboplastin was reconstituted according to the manufacturer’s directions and warmed to 37 °C. A 10 μL sample of the FXa inhibitor, to give the desired concentration, was brought up to 100 μL with citrated human plasma, incubated for 30 s at 37° C followed by addition of 200 μL of prewarmed thromboplastin. For the aPTT assay, 10 μL of inhibitor was mixed with 90 μL of citrated human plasma and 100 μL of prewarmed aPTT reagent (0.2% ellagic acid). After incubation for 4 min at 37 °C, clotting was initiated by adding 100 μL of prewarmed 25 mM CaCl2 and time to clot noted. The data were fit to a quadratic trendline, which was used to determine the concentration of the inhibitor necessary to double the clotting time. Clotting time in the absence of an anticoagulant was determined in similar fashion using 10 μL of deionized water and/or appropriate organic vehicle and was found to be 21.0 s for PT and 46.4 s for aPTT.

4.9. General Procedure for Nucleophilic Amidation of Halogenated Aniline Derivatives 8a–8g

An oven-dried, two-neck round bottom flask fitted with a condenser was charged with CuI (0.05 mmol) and K2CO3 (2.0 mmol) under nitrogen atmosphere. N, N′-dimethylethylenediamine (0.10 mmol), 4-haloaniline derivative (4a4c) (1.0 mmol), piperidin-2-one (5a)or morpholin-3-one ( 5b) (1.5 mmol), and anhydrous toluene (1 mL) were added. The reaction mixture was stirred and refluxed overnight. The resulting mixture was allowed to reach RT and filtered through Celite, which was further washed with methanol (10 mL). The organic filtrate was concentrated in vacuo and the residue was purified by flash chromatography using gradient mobile phase of EtOAc/hexanes to afford the corresponding para-amidated aniline derivatives (8a–8g) as solid products in 65–90% yield. The full spectral characterization of these products is provided as Supplementary Information.

4.9.1. 1-(4-aminophenyl)piperidin-2-one (8a)

1H-NMR (CDCl3, 400 MHz): 7.00 (d, J = 8.76 Hz, 2 H), 6.56 (d, J = 8.76 Hz, 2 H), 3.56 (t, J = 5.44 Hz, 2 H), 2.52 (t, J = 5.24 Hz, 2 H), 1.92-1.89 (m, 4 H). 13C-NMR (CDCl3, 100 MHz): 170.11, 146.25, 132.89, 127.19, 113.35, 52.17, 32.88, 23.04, 21.20. MS (ESI) calculated for C11H14N2O, [M+H]+, m/z 191.25, found for [M+H]+, m/z 191.98.

4.10. General Procedure for Catalytic Hydrogenation of Nitro Group for Synthesis of 8i and 8j

4-Nitrophenylpiperazine (7a or 7b) and 10% Pd/C was mixed in methanol (10 mL) containing concentrated HCl (2 mL). Hydrogen gas was then pumped into the mixture at RT. After stirring the solution for 5 h, the catalyst was filtered on Celite and the organic filtrate concentrated in vacuo to afford the corresponding aniline derivative (8h and 8j) in ~100% yield. The full spectral characterization of these products is provided as Supplementary Information.

4.10.1. 1-(4-(4-aminophenyl)piperazin-1-yl)ethanone (8i)

1H-NMR (CDCl3, 400 MHz): 6.81 (d, J = 8.76 Hz, 2 H), 6.66 (d, J = 8.76 Hz, 2 H), 3.75 (t, J = 5.08 Hz, 2 H), 3.60 (t, J = 4.92 Hz, 2 H), 3.01 (t, J = 5.20 Hz, 2 H), 2.98 (t, J = 5.12 Hz, 2 H), 2.13 (s, 3 H). 13C-NMR (CDCl3, 100 MHz): 168.98, 144.04, 140.90, 119.32, 116.17, 51.55, 51.14, 46.51, 41.61, 21.33. MS (ESI) calculated for C12H17N3O, [M+H]+, m/z 220.29, found for [M+]+, m/z 220.15.

4.10.2. (4-(4-aminophenyl)piperazin-1-yl)(thiophen-2-yl)methanone (8j)

1H-NMR (acetone-d6, 400 MHz): 7.57 (d, J = 4.96 Hz, 1 H), 7.34 (d, J = 3.56 Hz, 1 H), 7.03 (d, J = 4.32 Hz, 1 H), 6.86 (d, J = 8.64 Hz, 2 H), 6.54 (d, J = 8.60 Hz, 2 H), 3.77 (t, J = 4.96 Hz, 4 H), 3.08 (t, J = 5.16 Hz, 4 H). 13C-NMR (acetone-d6, 100 MHz): 163.60, 148.27, 138.75, 129.72, 127.70, 122.94, 121.33, 120.06, 118.43, 116.14, 52.39, 51.20. MS (ESI) calculated for C15H17N3OS, [M+H]+, m/z 288.39, found for [M+H]+, m/z 288.25.

4.11. General Procedure for Deprotection of t-Butyloxycarbonyl (Boc) Group of 16, 17, 21, 61, 76, 77, 80, 83, or 86

To a solution of 1,2,3,4-tetrahydroisoquinoline carboxamides (16, 17, 21), phenylalanine carboxamide (61), piperidine-2-carboxamides (76, 77, 80), 1,2,3,6-tetrahydropyridine-2-carboxamides (83), or 2-azabicyclo[2.2.1] heptane-3-carboxamides (86) (1.0 mmol)in CH 2Cl2 (5 mL), trifluoroacetic acid (TFA, 5 mL) was added drop-wise at 0 °C, and the mixture was warmed to RT. After stirring for 4 h, the reaction mixture was diluted with CH2Cl2 (25 mL) and neutralized by drop-wise addition of saturated aqueous NaHCO3 (20 mL). The organic layer was separated and the aqueous phase was extracted with EtOAc (2 × 25 mL). The organic extracts were combined, washed with saturated NaCl solution (25 mL), and dried over anhydrous Na2SO4. Removal of the solvent under reduced pressure afforded the desired unprotected carboxamides in quantitative yields and sufficient purity (as indicated by TLC) to be directly used in the next reactions without any further treatment.

4.12. General Procedure for Deprotection of Carbobenzyloxy (Cbz) Group of 12–15, 18–20, 50, 67or 68

1,2,3,4-tetrahydroisoquinoline carboxamides (1215, 18, 19, 50) or piperidine-3-carboxamides (67 or 68) and 10% Pd(OH)2 on activated charcoal were mixed in CH3OH: tert-butanol (1:1) mixture (10 mL). Hydrogen gas was then pumped into the mixture at RT. After stirring the solution overnight, the catalyst was filtered on Celite and the organic filtrate concentrated in vacuo to afford the corresponding desired unprotected carboxamides in quantitative yields and sufficient purity (as indicated by TLC) to be directly used in the next reactions without any further treatment.

4.13. General Procedure for Amine Protection by Carbobenzyloxy (Cbz) Group in the Synthesis of 65 or 66

Piperidine-3-carboxylic acid (63 or 64) (1 mmol) was dissolved in dry THF (10 mL) and stirred at RT. Tri-ethylamine (Et3N) (2 mmol) was then added followed by benzyl chloroformate (1.5 mmol). The reaction mixture became initially cloudy and turned to clear solution after 5 h. The reaction mixture was then partitioned between acidified water (15 mL) and EtOAc (20 mL). The aqueous layer was further washed with EtOAc (2 × 20 mL). The organic extracts were combined, dried over anhydrous Na2SO3, filtered, and concentrated in vacuo. The N-Cbz protected product (65 or 66) was isolated as white solids by flash chromatography using gradient of EtOAc/hexanes as eluant in 70–73 % yield.

4.13.1. (S)-1-(benzyloxycarbonyl)piperidine-3-carboxylic acid (65)

1H-NMR (acetone-d6, 400 MHz): 7.43-7.30 (m, 5 H), 5.15 (s, 2 H), 4.35-4.04 (m, 1 H), 3.93 (d, J = 12.08 Hz, 1 H), 3.18-3.02 (m, 1 H), 3.01-2.92 (m, 1 H), 2.53-2.46 (m, 1 H), 2.09-2.062 (m, 1 H), 1.75-1.66 (m, 2 H), 1.53-1.45 (m, 1 H). 13C-NMR (acetone-d6, 100 MHz): 174.88, 155.77, 138.25, 129.30, 128.70, 128.60, 67.49, 46.62, 44.90, 41.64, 27.87, 24.98. MS (ESI) calculated for C14H17NO4, [M+H]+, m/z 264.30, found for [M+H]+, m/z 264.37.

4.13.2. (R)-1-(benzyloxycarbonyl)piperidine-3-carboxylic acid (66)

1H-NMR (acetone-d6, 400 MHz): 7.43-7.30 (m, 5 H), 5.15 (s, 2 H), 4.35-4.04 (m, 1 H), 3.93 (d, J = 12.08 Hz, 1 H), 3.18-3.02 (m, 1 H), 3.01-2.92 (m, 1 H), 2.53-2.46 (m, 1 H), 2.09-2.062 (m, 1 H), 1.75-1.66 (m, 2 H), 1.53-1.45 (m, 1 H). 13C-NMR (acetone-d6, 100 MHz): 174.88, 155.77, 138.25, 129.30, 128.70, 128.60, 67.49, 46.62, 44.90, 41.64, 27.87, 24.98. MS (ESI) calculated for C14H17NO4, [M+H]+, m/z 264.30, found for [M+Na]+, m/z 286.28.

4.14. General Procedure for Amidation of 3-Carboxylic Acid to Yield 12–21, 50, 53–55, 59,61,67, 68,76, 77,80, 83, or 86

To a stirred solution of the free carboxylic acid of (9–11, 58, 60, 65,66, 74, 75, 82, or 85) (1.0 mmol) in anhydrous CH2Cl2 (5 mL) was added hydrated N-hydroxybenzotrizole (HOBT.H2O, 1.1 mmol), DMAP (1.1 mmol), and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDCI, 1.1 mmol) at RT under nitrogen atmosphere. Appropriate amine (8a – 8j or4-chloroaniline) (1.1 mmol) in anhydrous CH 2Cl2 (5 mL) was then added dropwise. After stirring overnight, the reaction mixture was partitioned between 2.0 N HCl solution (20 mL) and CH2Cl2 (30 mL). The organic layer was washed further with 2 N HCl (2 × 10 mL) and saturated NaCl solution (20 mL), dried using anhydrous Na2SO4, and concentrated to give a crude, which purified by flash chromatography using gradient of CH2Cl2/CH3OH as eluant to give the desired carboxamide product in 73 – 89 % yield. The full spectral characterization of these products is provided as Supplementary Information.

4.14.1. (S)-benzyl 3-(4-(2-oxopiperidin-1-yl)phenylcarbamoyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate (12)

1H-NMR (CDCl3, 400 MHz): 7.50-7.27 (m, 6 H), 7.25-7.08 (m, 5 H), 7.06 (d, J = 8.44, 2 H), 5.23-5.12 (m, 2 H), 4.96-4.54 (m, 3 H), 3.55 (t, J = 5.00 Hz, 2 H), 3.40-3.30 (m, 1 H), 3.20-3.09 (m, 1 H), 2.52 (t, J = 5.00 Hz, 2 H), 2.00-1.87 (m, 4 H). 13C-NMR (CDCl3, 100 MHz): 170.23, 169.22, 156.94, 139.31, 136.09, 135.47, 134.33, 133.32,128.64, 128.32, 128.07, 127.76, 127.23, 126.90, 126.58,125.99, 120.87, 68.04, 64.35, 51.75, 45.08, 32.82, 30.31, 23.51, 21.41. MS (ESI) calculated for C29H29N3O4, [M+H]+, m/z 484.57, found for [M+Na]+, m/z 506.3.

4.14.2. (S)-benzyl 3-(4-chlorophenylcarbamoyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50)

1H-NMR (CDCl3, 400 MHz): 7.50-7.35 (m, 3 H), 7.34-7.26 (m, 2 H), 7.25-7.18 (m, 4 H), 7.17-7.00 (m, 4 H), 5.39-5.10 (m, 2 H), 4.96-4.49 (m, 3 H), 3.35-3.21 (m, 1 H), 3.20-3.3.03 (m, 1 H). 13C-NMR (CDCl3, 100 MHz): 169.12, 157.30, 136.24, 135.95, 133.44, 132.70, 129.08, 128.81, 128.70, 128.57, 128.43, 128.11, 127.77, 126.78, 125.94, 121.13, 68.19, 57.04, 45.11, 28.30. MS (ESI) calculated for C24H21ClN2O3, [M+H]+, m/z 421.90, found for [M+Na]+, m/z 443.26.

4.14.3. (S)-tert-butyl 6-(4-(3-oxomorpholino)phenylcarbamoyl)-5,6-dihydropyridine-1(2H)-carboxylate (83)

1H-NMR (CDCl3, 400 MHz): 7.50 (d, J = 8.80 Hz, 2 H), 7.22 (d, J = 8.64 Hz, 2 H), 5.85-5.80 (m, 1 H), 4.65-4.60 (m, 1 H), 5.00-4.93 (m, 1 H), 4.28 (s, 2 H), 4.20-4.07 (m, 1 H), 3.94 (t, J = 4.95 Hz, 2 H), 3.65 (t, J = 4.40 Hz, 2 H), 3.60-3.63 (m, 1 H), 2.60-3.72 (m, 1 H), 2.34-2.28 (m, 1 H). 13C-NMR (CDCl3, 100 MHz): 169.54, 166.82, 156.90, 137.99, 126.12, 123.36, 122.20, 120.36, 81.41, 68.54, 64.11, 61.4, 49.79, 41.50, 28.39, 24.10. MS (ESI) calculated for C22H29N3O5, [M+H]+, m/z 402.46, found for [M+Na]+, m/z 424.36.

4.15. General Procedure for Amidation at the Ring Nitrogen of the Core Structure to Yield 7a, 7b, 22–49, 51, 58, 62, 69–73, 78, 79, 81, 84, or87

To a stirred solution of organic acid (1.0 mmol) in anhydrous CH2Cl2 (5 mL) was added 1-Ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDCI, 1.1 mmol) and DMAP (1.1 mmol) at RT under nitrogen atmosphere. The unprotected form (free amine) of (6,12–21, 50, 57, 61,67, 68, 76, 77,80, 83, or 86) (1.1 mmol) in anhydrous CH2Cl2 (5 mL) was then added drop-wise. After stirring overnight, the reaction mixture was partitioned between 2.0 N HCl solution (20 mL) and CH2Cl2 (30 mL). The organic layer was washed further with 2 N HCl (2 × 10 mL) and saturated NaCl solution (20 mL), dried using anhydrous Na2SO4, and concentrated to give a crude, which purified by flash chromatography using gradient of CH2Cl2/CH3OH as eluting phase to give the desired dicarboxamide product in 70 – 95 % yield. The full spectral characterization of these products is provided as Supplementary Information.

4.15.1. (S)-N-methyl-N-(4-(2-oxopiperidin-1-yl)phenyl)-2-(thiophene-2-carbonyl)-1,2,3,4-tetrahydroisoquinoline-3-carboxamide (22)

1H-NMR (CDCl3, 400 MHz): 7.51-7.42 (m, 2 H), 7.41 (d, J = 0.88 Hz, 1 H), 7.4-7.39 (m, 1 H), 7.37 (d, J = 20.08 Hz, 2 H), 7.12-7.09 (m, 2 H), 7.04-7.00 (m, 3 H), 4.97-4.76 (dd, J = 14.96 Hz, J = 69.36 Hz, 2 H), 4.9-4.82 (m, 1 H), 3.55 (t, J= 5.44 Hz, 2 H), 3.21 (s, 3 H), 2.98-2.85 (m, 2 H), 2.50 (t, J= 6.44 Hz, 2 H), 1.90-1.84 (m, 4 H). 13C-NMR (CDCl3, 100 MHz): 171.32, 170.07, 162.67, 142.93, 141.12, 137.40, 134.54, 133.10, 129.36, 129.01, 128.17, 127.60, 127.36, 126.88, 126.74, 125.48, 53.06, 51.37, 48.92, 38.00, 32.90, 31.24, 23.50, 21.36. MS (ESI) calculated for C27H27N3O3S, [M+H]+, m/z 474.59, found for [M+Na]+, m/z 496.21.

4.15.2. (R)-2-(2-(4-chlorophenyl)acetyl)-N-(4-(3-oxomorpholino)phenyl)-1,2,3,4-tetrahydro-isoquinoline-3-carboxamide (47)

1H-NMR (CDCl3, 400 MHz): 7.57-7.30 (m, 2 H), 7.26-7.20 (m, 2 H), 7.19-7.17 (m, 2 H), 7.16-7.00 (m, 4 H), 6.99-6.68 (m, 2 H). 5.14-4.12 (m, 1 H), 4.75-4.50 (m, 2 H), 4.25 (s, 2 H), 3.95 (t, J = 4.72 Hz, 2 H), 3.81 (d, J = 10.40 Hz, 2 H), 3.66 (t, J = 5.20 Hz, 2 H), 3..38-3.29 (m, 1 H), 3.14-2.92 (m, 1 H). 13C-NMR (CDCl3, 100 MHz): 172.23, 169.13, 167.34, 136.97, 136.81, 133.53, 133.09, 132.45, 130.72, 130.28, 130.11, 129.54, 129.00, 128.01, 127.46, 126.75, 125.95, 125.39, 124.21, 122.79, 120.63, 68.29, 63.94, 60.56, 52.53, 49.78, 46.88, 40.45, 28.12. MS (ESI) calculated for C28H26ClN3O4, [M+H]+, m/z 504.98, found for [M+Na]+, m/z 526.21.

4.15.3. (S)-2-(2-(4-chlorophenyl)acetamido)-N-methyl-N-(4-(2-oxopiperidin-1-yl)phenyl)-3-phenyl propanamide (62)

1H-NMR (CDCl3, 400 MHz): 7.30-7.22 (m, 4 H), 7.21-7.19 (m, 3 H), 7.11 (d, J= 8.36 Hz, 2 H), 6.93 (d, J = 7.72 Hz, 2 H), 6.84 (d, J = 7.12 Hz, 2 H), 4.87-4.82 (q, J = 7.16 Hz, J = 15.08 Hz, 1 H), 3.69 (t, J = 4.96 Hz, 2 H), 3.47 (s, 2 H), 3.22 (s, 3 H), 2.90-2.85 (dd, J = 7.16 Hz, J = 13.32, 1 H), 2.71-2.66 (dd, J = 7.00 Hz, J = 13.36 Hz, 1 H), 2.59 (t, J = 6.40, 2 H), 2.05-1.95 (m, 4 H). 13C-NMR (CDCl3, 100 MHz): 171.30, 170.07, 169.63, 143.01, 140.21, 136.00, 133.15, 130.62, 129.24, 128.95, 128.44, 127.95, 127.25, 126.90, 51.42, 51.19, 42.76, 38.91, 37.60, 32.93, 23.53, 21.40. MS (ESI) calculated for C29H30ClN3O3, [M+H]+, m/z 505.03, found for [M+Na]+, m/z 526.33.

4.15.4. (1R, 3S, 4R)-2-(2-(4-chlorophenyl)acetyl)-N-(4-(3-oxomorpholino)phenyl)-2-azabicyclo [2.2.1]heptane-3-carboxamide (87)

1H-NMR (CDCl3, 400 MHz): 7.52 (d, J = 8.64 Hz, 2 H), 7.32 (d, J = 8.32 Hz, 2 H), 7.23 (d, J = 8.36 Hz, 2 H), 7.22 (d, J = 8.20 Hz, 2H), 4.32 (s, 2 H), 4.26 (s, 2 H), 4.01 (t, J = 4.96 Hz, 2 H), 3.76-3.65 (m, 4 H), 3.11-3.95 (m, 1 H), 2.04-1.99 (m, 1 H), 1.89-1.74 (m, 3 H), 1.63-1.45 (m, 3 H). 13C-NMR (CD3OD, 100 MHz): 171.24, 170.63, 169.48, 138.89, 138.48, 135.06, 133.86, 132.02, 129.63, 127.56, 122.03, 69.07, 67.43, 65.09, 60.21, 51.20, 43.46, 41.58, 36.43, 32.14, 28.55. MS (ESI) calculated for C25H26ClN3O4, [M+H]+, m/z 468.95, found for [M+Na]+, m/z 490.27.

Supplementary Material

01

Acknowledgments

This work was supported by grants RC1 HL099420, R01 HL090586 and P01 HL107152 from the National Institutes of Health.

Abbreviations

ABH3CA

2-azabicyclo [2.2.1]heptane-3-carboxylic acid

APTT

activated partial thromboplastin time

FIIa

factor IIa (thrombin)

FXa

factor Xa

HS

Hill slope

P2CA

piperidine–2–carboxylic acid

P3CA

piperidine–3–carboxylic acid

PA

phenylalanine

PT

prothrombin time

THIQ

1,2,3,4-tetrahydroisoquinoline

THIQ3CA

1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid

THP2CA

1,2,3,6–tetrahydropyridine–2–carboxylic acid

Footnotes

Supplementary Information

General description of the virtual library of (3S)-THIQ3CA and synthesis of chemical arm 8a8g, 8i, and 8j are included. Synthetic schemes for derivatives 5055, 59, and 62 are also provided. 1H and 13C NMR and ESI MS characterization of all synthesized compounds are also available. Included also figures to describe: structural exploration of 1,3–disubstituted, six–membered, cyclic dicarboxamides of piperidine–3–carboxylic acid, general structure of THIQ3CA-based FXa inhibitors with the corresponding modifications at different positions, and lastly flow chart of systematic structural modifications which led to the most potent compound 47. This material is available free of charde via the internet.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Henry BL, Desai UR. Anticoagulants: Drug discovery and development. In: Rotella D, Abraham DJ, editors. Burger’s Medicinal Chemistry. John Wiley and Sons; New York: 2010. pp. 365–408. [Google Scholar]
  • 2.Lee YK, Player MR. Developments in factor Xa inhibitors for the treatment of thromboembolic disorders. Med Res Rev. 2011;31:202–283. doi: 10.1002/med.20183. [DOI] [PubMed] [Google Scholar]
  • 3.Huntington JA, Baglin TP. Targeting thrombin–rational drug design from natural mechanisms. Trends Pharmacol Sci. 2003;24:589–595. doi: 10.1016/j.tips.2003.09.002. [DOI] [PubMed] [Google Scholar]
  • 4.Popović M, Smiljanić K, Dobutović B, Syrovets T, Simmet T, Isenović ER. Thrombin and vascular inflammation. Mol Cell Biochem. 2012;359:301–313. doi: 10.1007/s11010-011-1024-x. [DOI] [PubMed] [Google Scholar]
  • 5.Mann KG, Brummel K, Butenas S. What is all that thrombin for. J Thromb Haemost. 2003;1:1504–1514. doi: 10.1046/j.1538-7836.2003.00298.x. [DOI] [PubMed] [Google Scholar]
  • 6.Yin ET, Wessler S. Investigation of the apparent thrombogenicity of thrombin. Thromb Diath Haemorrh. 1968;20:465–468. [PubMed] [Google Scholar]
  • 7.Yin ET, Wessler S. Heparin-accelerated inhibition of activated factor X by its natural inhibitor. Biochem Biophys Acta. 1970;201:387–390. doi: 10.1016/0304-4165(70)90316-8. [DOI] [PubMed] [Google Scholar]
  • 8.Wessler S, Yin ET. Theory and practice of minidose heparin in surgical patients. Circulation. 1973;47:671–676. doi: 10.1161/01.cir.47.4.671. [DOI] [PubMed] [Google Scholar]
  • 9.Ansell J. Factor Xa or thrombin: Is factor Xa a better target? J Thromb Haemost. 2007;5(Suppl 1):60–64. doi: 10.1111/j.1538-7836.2007.02473.x. [DOI] [PubMed] [Google Scholar]
  • 10.Merlini PA, Ardissino D, Bauer KA, Oltrona L, Pezzano A, Bottasso B, Rosenberg RD, Mannucci PM. Persistent thrombin generation during heparin therapy in patients with acute coronary syndromes. Arterioscler Thromb Vasc Biol. 1977;17:1325–1330. doi: 10.1161/01.atv.17.7.1325. [DOI] [PubMed] [Google Scholar]
  • 11.Hermans C, Claeys D. Review of the rebound phenomenon in new anticoagulant treatments. Curr Med Res Opin. 2006;22:471–481. doi: 10.1185/030079906X89801. [DOI] [PubMed] [Google Scholar]
  • 12.Rezaie RA. Prothrombin protects factor Xa in the prothrombinase complex from inhibition by the heparin-antithrombin complex. Blood. 2001;97:2308–2313. doi: 10.1182/blood.v97.8.2308. [DOI] [PubMed] [Google Scholar]
  • 13.Weitz JI, Hudoba M, Massel D, Maraganore J, Hirsh J. Clot-bound thrombin is protected from inhibition by heparin-antithrombin III independent inhibitors. J Clin Invest. 1990;86:385–391. doi: 10.1172/JCI114723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Desai UR. New antithrombin-based anticoagulants. Med Res Rev. 2004;24:151–181. doi: 10.1002/med.10058. [DOI] [PubMed] [Google Scholar]
  • 15.Al-Horani RA, Liang A, Desai UR. Designing nonsaccharide allosteric activators of antithrombin for accelerated inhibition of factor Xa. J Med Chem. 2011;54:6125–6138. doi: 10.1021/jm2008387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Straub A, Roehrig S, Hillisch A. Oral, direct thrombin and factor Xa inhibitors: The replacement for warfarin, leeches, and pig intestines? Angew Chem Int Ed. 2011;50:4574–4590. doi: 10.1002/anie.201004575. [DOI] [PubMed] [Google Scholar]
  • 17.Henry BL, Monien BH, Bock PE, Desai UR. A novel allosteric pathway of thrombin inhibition. Exosite II mediated potent inhibition of thrombin by chemo-enzymatic, sulfated dehydropolymers of 4-hydroxycinnamic acids. J Biol Chem. 2007;282:31891–31899. doi: 10.1074/jbc.M704257200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sidhu PS, Liang A, Mehta AY, Abdel Aziz MH, Zhou Q, Desai UR. Rational design of potent, small, synthetic allosteric inhibitors of thrombin. J Med Chem. 2011;54:5522–5531. doi: 10.1021/jm2005767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Xu Y, Masuko S, Takieddin M, Xu H, Liu R, Jing J, Mousa SA, Linhardt RJ, Liu J. Chemoenzymatic synthesis of homogeneous ultralow molecular weight heparins. Science. 2011;334:498–501. doi: 10.1126/science.1207478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gunnarsson GT, Desai UR. Designing small, non-sugar activators of antithrombin using hydropathic interaction analyses. J Med Chem. 2002;45:1233–1243. doi: 10.1021/jm020012q. [DOI] [PubMed] [Google Scholar]
  • 21.Raghuraman A, Liang A, Krishnasamy C, Lauck T, Gunnarsson CT, Desai UR. On designing non-saccharide, allosteric activators of antithrombin. Eur J Med Chem. 2009;44:2626–2631. doi: 10.1016/j.ejmech.2008.09.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. The discovery and development of rivaroxaban, an oral, direct factor Xa inhibitor. Nature Rev Drug Discov. 2011;10:61–75. doi: 10.1038/nrd3185. [DOI] [PubMed] [Google Scholar]
  • 23.Pinto DJP, Orwat MJ, Koch S, Rossi KA, Alexander RS, Smallwood A, Wong PC, Rendina AR, Luettgen JM, Knabb RM, He K, Xin B, Wexler RR, Lam PYS. Discovery of 1-(4-methoxyphenyl)-7-oxo-6-(4-(2-oxopiperidin-1-yl)phenyl)-4,5,6,7-tetrahydro- 1H-pyrazolo[3,4-c]pyridine-3-carboxamide (Apixaban, BMS-562247), a highly potent, selective, efficacious, and orally bioavailable inhibitor of blood coagulation factor Xa. J Med Chem. 2007;50:5339–5356. doi: 10.1021/jm070245n. [DOI] [PubMed] [Google Scholar]
  • 24.Pinto DJ, Orwat DMJ, Wang S, Fevig JM, Quan ML, Amparo E, Cacciola J, Rossi KA, Alexander RS, Smallwood AM, Luettgen JM, Liang L, Aungst BJ, Wright MR, Knabb RM, Wong PC, Wexler RR, Lam PY. Discovery of 1-[3-(aminomethyl)phenyl]-N-[3-fluoro-2′-(methylsulfonyl)- [1,1′-biphenyl]-4-yl]-3-(trifluoromethyl)-1H-pyrazole-5-carboxamide (DPC423), a highly potent, selective, and orally bioavailable inhibitor of blood coagulation factor Xa. J Med Chem. 2001;44:566–578. doi: 10.1021/jm000409z. [DOI] [PubMed] [Google Scholar]
  • 25.Imaeda Y, Kuroita T, Sakamoto H, Kawamoto T, Tobisu M, Konishi N, Hiroe K, Kawamura M, Tanaka T, Kubo K. Discovery of imidazo[1,5-c]imidazol-3-ones: Weakly basic, orally active factor Xa inhibitors. J Med Chem. 2008;51:3422–3436. doi: 10.1021/jm701548u. [DOI] [PubMed] [Google Scholar]
  • 26.Maignan S, Guilloteau JP, Choi-Sledeski YM, Becker MR, Ewing WR, Pauls HW, Spada AP, Mikol V. Molecular structures of human factor Xa complexed with ketopiperazine inhibitors: Preference for a neutral group in the S1 pocket. J Med Chem. 2003;46:685–690. doi: 10.1021/jm0203837. [DOI] [PubMed] [Google Scholar]
  • 27.Nagata T, Yoshino T, Haginoya N, Yoshikawa K, Nagamochi M, Kobayashi S, Komoriya S, Yokomizo A, Muto R, Yamaguchi M, Osanai K, Suzuki M, Kanno H. Discovery of N- [(1R,2S,5S)-2-{[(5-chloroindol-2-yl)carbonyl]amino}-5-(dimethylcarbamoyl)cyclohexyl]-5-methyl- 4,5,6,7-tetrahydrothiazolo[5,4-c]pyridine-2-carboxamide hydrochloride: A novel, potent and orally active direct inhibitor of factor Xa. Bioorg Med Chem. 2009;17:1193–1206. doi: 10.1016/j.bmc.2008.12.037. [DOI] [PubMed] [Google Scholar]
  • 28.Young RJ, Borthwick AD, Brown D, Burns-Kurtis CL, Campbell M, Chan C, Charbaut M, Chung CW, Convery MA, Kelly HA, King NP, Kleanthous S, Mason AM, Pateman AJ, Patikis AN, Pinto IL, Pollard DR, Senger S, Shah GP, Toomey JR, Watson NS, Weston HE. Structure and property based design of factor Xa inhibitors: Pyrrolidin-2-ones with biaryl P4 motifs. Bioorg Med Chem Lett. 2008;18:23–27. doi: 10.1016/j.bmcl.2007.11.023. [DOI] [PubMed] [Google Scholar]
  • 29.Smallheer JM, Wang S, Laws ML, Nakajima S, Hu Z, Han W, Jacobson I, Luettgen JM, Rossi KA, Rendina AR, Knabb RM, Wexler RR, Lam PYS, Quan ML. Sulfonamidolactam inhibitors of coagulation factor Xa. Bioorg Med Chem Lett. 2008;18:2428–2433. doi: 10.1016/j.bmcl.2008.02.054. [DOI] [PubMed] [Google Scholar]
  • 30.Roehrig S, Straub A, Pohlmann J, Lampe T, Pernerstorfer S, Schlemmer KH, Reinemer P, Perzborn E. Discovery of the novel antithrombotic agent 5-chloro-N-({(5S)-2-oxo-3- [4-(3-oxomorpholin-4-yl)phenyl]-1,3-oxazolidin-5-yl}methyl)thiophene- 2-carboxamide (BAY 59-7939): An oral, direct factor Xa inhibitor. J Med Chem. 2005;48:5900–5908. doi: 10.1021/jm050101d. [DOI] [PubMed] [Google Scholar]
  • 31.Kohrt JT, Filipski KJ, Cody WL, Bigge CF, La F, Welch K, Dahring T, Bryant JW, Leonard D, Bolton G, Narasimhan L, Zhang E, Peterson JT, Haarer S, Sahasrabudhe V, Janiczek N, Desiraju S, Hena M, Fiakpui C, Saraswat N, Sharma R, Sun S, Maiti SN, Leadley R, Edmunds JJ. The discovery of glycine and related amino acid-based factor Xa inhibitors. Bioorg Med Chem. 2006;14:4379–4392. doi: 10.1016/j.bmc.2006.02.040. [DOI] [PubMed] [Google Scholar]
  • 32.Van Huis CA, Casimiro-Garcia A, Bigge CF, Cody WL, Dudley DA, Filipski KJ, Heemstra RJ, Kohrt JT, Jr, Leadley RJ, Narasimhan LS, McClanahan T, Mochalkin I, Pamment M, Thomas Peterson J, Sahasrabudhe V, Schaum RP, Edmunds JJ. Exploration of 4,4-disubstituted pyrrolidine-1,2-dicarboxamides as potent, orally active factor Xa inhibitors with extended duration of action. Bioorg Med Chem. 2009;17:2501–2511. doi: 10.1016/j.bmc.2009.01.063. [DOI] [PubMed] [Google Scholar]
  • 33.Ye B, Arnaiz DO, Chou YL, Griedel BD, Karanjawala R, Lee W, Morrissey MM, Sacchi KL, Sakata ST, Shaw KJ, Wu SC, Zhao Z, Adler M, Cheeseman S, Dole WP, Ewing J, Fitch R, Lentz D, Liang A, Light D, Morser J, Post J, Rumennik G, Subramanyam B, Sullivan ME, Vergona R, Walters J, Wang YX, White KA, Whitlow M, Kochanny MJ. Thiophene-anthranilamides as highly potent and orally available factor Xa inhibitors. J Med Chem. 2007;50:2967–2980. doi: 10.1021/jm070125f. [DOI] [PubMed] [Google Scholar]
  • 34.Pruitt JR, Pinto DJ, Estrella MJ, Bostrom LL, Knabb RM, Wong PC, Wright MR, Wexler RR. Isoxazolines and isoxazoles as factor Xa inhibitors. Bioorg Med Chem Lett. 2000;10:685–689. doi: 10.1016/s0960-894x(00)00097-4. [DOI] [PubMed] [Google Scholar]
  • 35.Quan ML, Lam PYS, Han Q, Pinto DJP, He MY, Renhua E, Christopher D, Clark CG, Teleha CA, Sun JH, Alexander RS, Bai S, Luettgen JM, Knabb RM, Wong PC, Wexler RR. Discovery of 1-(3′-aminobenzisoxazol-5′-yl)-3-trifluoromethyl-N-[2-fluoro-4- [(2′-dimethylamino-methyl)imidazol-1-yl]phenyl]-1H-pyrazole-5-carboxyamide hydrochloride (Razaxaban), a highly potent, selective, and orally bioavailable factor Xa inhibitor. J Med Chem. 2005;48:1729–1744. doi: 10.1021/jm0497949. [DOI] [PubMed] [Google Scholar]
  • 36.Lee YK, Parks DJ, Lu T, Thieu TV, Markotan T, Pan W, McComsey DF, Milkiewicz KL, Crysler CS, Ninan N, Abad MC, Giardino EC, Maryanoff BE, Damiano BP, Player MR. 7-Fluoroindazoles as potent and selective inhibitors of factor Xa. J Med Chem. 2008;51:282–297. doi: 10.1021/jm701217r. [DOI] [PubMed] [Google Scholar]
  • 37.Matter H, Defossa E, Heinelt U, Blohm PM, Schneider D, Müller A, Herok S, Schreuder H, Liesum A, Brachvogel V, Lönze P, Walser A, Al-Obeidi F, Wildgoose P. Design and quantitative structure-activity relationship of 3-amidinobenzyl-1H-indole-2-carboxamides as potent, nonchiral, and selective inhibitors of blood coagulation factor Xa. J Med Chem. 2002;45:2749–2769. doi: 10.1021/jm0111346. [DOI] [PubMed] [Google Scholar]
  • 38.Nazaré, Will DW, Matter H, Schreuder H, Ritter K, Urmann M, Essrich M, Bauer A, Wagner M, Czech J, Lorenz M, Laux V, Wehner V. Probing the subpockets of factor Xa reveals two binding modes for inhibitors based on a 2-carboxyindole scaffold: a studycombining structure-activity relationship and X-ray crystallography. J Med Chem. 2005;48:4511–4525. doi: 10.1021/jm0490540. [DOI] [PubMed] [Google Scholar]
  • 39.Pinto DJP, Jr, Galemmo RA, Quan ML, Orwat MJ, Clark C, Li R, Wells B, Woerner F, Alexander RS, Rossi KA, Smallwood A, Wong PC, Luettgen JM, Rendina AR, Knabb RM, He K, Wexler RR, Lam PYS. Discovery of potent, efficacious, and orally bioavailable inhibitors of blood coagulation factor Xa with neutral P1 moieties. Bioorg Med Chem Lett. 2006;16:5584–5589. doi: 10.1016/j.bmcl.2006.08.027. [DOI] [PubMed] [Google Scholar]
  • 40.Eriksson BI, Quinlan DJ, Weitz JI. Comparative pharmacodynamics and pharmacokinetics of oral direct thrombin and factor Xa inhibitors in development. Clin Pharmacokinet. 2009;48:1–22. doi: 10.2165/0003088-200948010-00001. [DOI] [PubMed] [Google Scholar]
  • 41.Zhang Y, Feng J, Liu C, Zhang L, Jiao J, Fang H, Su L, Zhang X, Zhang J, Li M, Wang B, Xu W. Design, synthesis, and preliminary activity assay of 1,2,3,4-tetrahydro-isquinoline-3-carboxylic acid derivatives as novel histine deacetylases (HDACs) inhibitors. Bioorg Med Chem. 2010;18:1761–1772. doi: 10.1016/j.bmc.2010.01.060. [DOI] [PubMed] [Google Scholar]
  • 42.Cheng S, Zhang X, Wang W, Zhao M, Zheng M, Chang HW, Wu J, Peng S. A class of novel N-(3S-1,2,3,4-tetrahydroisoquinoline-3-carbonyl)-L-amino acid derivatives: Their synthesis, antithrombotic activity evaluation, and 3D QSAR analysis. Eur J Med Chem. 2009;44:4904–4919. doi: 10.1016/j.ejmech.2009.08.002. [DOI] [PubMed] [Google Scholar]
  • 43.Azukizawa S, Kasai M, Takahashi K, Miike T, Kunishiro K, Kanda M, Mukai C, Shirahase H. Synthesis and biological evaluation of (S)-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acids: A novel series of PPARγ agonists. Chem Pharm Bull. 2008;56:335–345. doi: 10.1248/cpb.56.335. [DOI] [PubMed] [Google Scholar]
  • 44.Cai TB, Zou Z, Thomas JB, Brieaddy L, Navarro HA, Carroll FI. Synthesis and in vitro opioid receptor functional antagonism of analogues of the selective kappa opioid receptor antagonist (3R)-7-hydroxy-N-((1S)-1-{[(3R,4R)-4-(3-hydroxyphenyl)-3,4-dimethyl-1-piperidinyl] methyl}-2-methylpropyl)-1,2,3,4-tetrahydro-3-isoquinolinecarboxamide (JDTic) J Med Chem. 2008;51:1849–1860. doi: 10.1021/jm701344b. [DOI] [PubMed] [Google Scholar]
  • 45.Montalbetti CAGN, Falque V. Amide bond formation and peptide coupling. Tetrahedron. 2005;61:10827–10852. [Google Scholar]
  • 46.Klapars A, Huang X, Buchwald SL. A general and efficient copper catalyst for the amidation of aryl halides. J Am Chem Soc. 2002;124:7421–7428. doi: 10.1021/ja0260465. [DOI] [PubMed] [Google Scholar]
  • 47.Shendage DM, Froehlich R, Haufe G. Highly efficient stereoconservative amidation and deamidation of α-Amino Acids. Org Lett. 2004;6:3675–3678. doi: 10.1021/ol048771l. [DOI] [PubMed] [Google Scholar]
  • 48.Mathias LJ, Fuller WD, Nissen D, Goodman M. Polydepsipeptides. 6. Synthesis of sequential polymers containing varying ratios of L-alanine and L-lactic Acid. Macromolecules. 1978;11:534–539. [Google Scholar]
  • 49.Verghese J, Liang A, Sidhu PP, Hindle M, Zhou Q, Desai UR. First steps in the direction of synthetic, allosteric, direct inhibitors of thrombin and factor Xa. Bioorg Med Chem Lett. 2009;19:4126–4129. doi: 10.1016/j.bmcl.2009.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pinto DJP, Smallheer JM, Cheney DL, Knabb RM, Wexler RR. Factor Xa inhibitors: Next-generation antithrombotic agents. J Med Chem. 2010;53:6243–6274. doi: 10.1021/jm100146h. [DOI] [PubMed] [Google Scholar]
  • 51.Young RJ. The successful quest for oral factor Xa inhibitors; Learnings for all of medicinal chemistry? Bioorg Med Chem Lett. 2011;21:6228–6235. doi: 10.1016/j.bmcl.2011.08.119. [DOI] [PubMed] [Google Scholar]
  • 52.Rupprecht H-J, Blank R. Clinical pharmacology of direct and indirect factor Xa inhibitors. Drugs. 2010;70:2153–2170. doi: 10.2165/11538030-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 53.Henry BL, Thakkar JN, Martin EJ, Brophy DF, Desai UR. Characterization of the plasma and blood anticoagulant potential of structurally and mechanistically novel oligomers of 4-hydroxycinnamic acids. Blood Coagul Fibrinolysis. 2009;20:27–34. doi: 10.1097/MBC.0b013e328304e077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Monien BH, Henry BL, Raghuraman A, Hindle M, Desai UR. Novel chemo-enzymatic oligomers of cinnamic acids as direct and indirect inhibitors of coagulation proteinases. Bioorg Med Chem. 2006;14:7988–7998. doi: 10.1016/j.bmc.2006.07.066. [DOI] [PubMed] [Google Scholar]
  • 55.Schumacher WA, Seiler SE, Steinbacher TE, Stewart AB, Bostwick JS, Hartl KS, Liu EC, Ogletree ML. Antithrombotic and hemostatic effects of a small molecule factor XIa inhibitor in rats. Eur J Pharmacol. 2007;570:167–174. doi: 10.1016/j.ejphar.2007.05.043. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES