Synthesis 2020; 52(14): 2099-2105
DOI: 10.1055/s-0040-1707103
paper
© Georg Thieme Verlag Stuttgart · New York

A Straightforward Synthesis of N-Substituted Ureas from Primary Amides

Nathalie Saraiva Rosa
a   Normandie Univ, ENSICAEN, UNICAEN, CNRS, LCMT, 14000 Caen, France   Email: vincent.reboul@ensicaen.fr
,
a   Normandie Univ, ENSICAEN, UNICAEN, CNRS, LCMT, 14000 Caen, France   Email: vincent.reboul@ensicaen.fr
,
a   Normandie Univ, ENSICAEN, UNICAEN, CNRS, LCMT, 14000 Caen, France   Email: vincent.reboul@ensicaen.fr
,
Jean-François Lohier
a   Normandie Univ, ENSICAEN, UNICAEN, CNRS, LCMT, 14000 Caen, France   Email: vincent.reboul@ensicaen.fr
,
b   Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA, 76000 Rouen, France
,
a   Normandie Univ, ENSICAEN, UNICAEN, CNRS, LCMT, 14000 Caen, France   Email: vincent.reboul@ensicaen.fr
› Author Affiliations
The authors thank the Centre National de la Recherche Scientifique (CNRS), Normandie Université (RIN ChemImaging), Labex SynOrg (Grant No. ANR-11-LABX-0029), the Conseil Régional de Normandie and the Fonds Européen de Développement Régional (FEDER) for financial support.
Further Information

Publication History

Received: 13 February 2020

Accepted after revision: 06 April 2020

Publication Date:
27 April 2020 (online)

 


Abstract

A direct and convenient method for the preparation of N-substituted ureas is achieved by treating primary amides with phenyliodine diacetate (PIDA) in the presence of an ammonia source (NH3 or ammonium carbamate) in MeOH. The use of 2,2,2-trifluoroethanol (TFE) as the solvent increases the electrophilicity of the hypervalent iodine species and allows the synthesis of electron-poor carboxamides. This transformation involves a nucleophilic addition of ammonia on the isocyanate intermediate generated in situ by a Hofmann rearrangement of the starting amide.


#

Due to their interesting physicochemical and biological properties, urea-containing molecules are increasingly used in various research fields such as medicinal chemistry,[1] agrochemistry[­2] and petrochemistry.[3] In particular, the extensive utilization of such moieties in drug discovery as bioisosteres­ of peptide bonds arises from their structural, biological and electronic similarities.[4]

This key structural motif (Figure [1]) can be found in natural products such as the amino acid citrulline (1) and allantoin (2), in marketed drugs such as cetrorelix (a decapeptide for in vitro fertilization)[5] and degarelix (treatment of prostate cancer),[6] and in several molecules with different biological activities such as anticancer (3, a checkpoint kinase inhibitor),[7] anti-obesity (4),[8] antiviral (5)[9] and antibacterial (6).[10] In addition, primary ureas are not only useful in medicinal chemistry but can also be employed as versatile building blocks for further transformations.[11]

Zoom Image
Figure 1 Biologically active compounds containing a primary urea moiety

Since the first synthesis of urea from ammonium cyanate by Wöhler in 1828, historically considered as the birth of organic chemistry,[12] researchers have developed a myriad of synthetic methods in order to prepare them. One of the most common method implies the reaction between ammonia and isocyanates, albeit with safety concerns.[13] Consequently, isocyanates are usually formed in situ thanks to Hofmann, Curtius or Lossen rearrangements,[14] respectively from primary carboxamides, acyl azides and bis-acylated hydroxylamines. Interestingly, hypervalent iodine species appeared to be suitable oxidizing reagents for the Hofmann rearrangement, since they are mild, powerful, and are able to react with amide substrates without added base. Under various conditions, amines,[15] carbamates[16] and symmetric 1,3-disubstituted ureas[17] can be prepared (Scheme [1]).

Herein, we report a straightforward synthesis of primary alkyl- and arylureas (Scheme [1]) by addition of ammonia to in situ generated isocyanates, proceeding via the Hofmann rearrangement of primary amides induced by phenyliodine diacetate (PIDA).

Zoom Image
Scheme 1 Reactions of carboxamides with hypervalent iodine species

Numerous methods have been developed for the synthesis of primary ureas (Scheme [2]) using different precursors. Examples include carboxylic acids,[18] phenyl carbamate,[19] anilines in the presence of urea,[20] aryl chlorides by Pd-catalyzed cross-couplings with benzylurea followed by in situ hydrogenolysis,[21] arylcyanamides by hydration reactions (with or without HCO2H),[22] amines by nucleophilic addition to potassium isocyanate,[23] nitriles via the Tiemann rearrangement,[24] or arenes by CH amination.[25]

Zoom Image
Scheme 2 Literature methods for the preparation of primary ureas

Originally, the unexpected formation of an N-substituted urea was observed during the investigation of the one-pot synthesis of 3H-diazirines from α-amino acids,[26] using PIDA and a methanolic solution of ammonia. Under these conditions, l-glutamine afforded a crystalline compound 7, X-ray analysis of which[27] revealed that the amide moiety was converted into the corresponding primary urea (Scheme [3]).

Zoom Image
Scheme 3 Formation of diazirine 7 (from l-glutamine) and its X-ray crystal structure (packing with H-bonding)

Since the formation of methyl carbamate 8 [19] was not observed, despite the presence of methanol in the reaction medium,[28] we assumed that the isocyanate intermediate, formed in situ via a Hofmann-like rearrangement,[29] was trapped by ammonia which is more nucleophilic (Scheme [4]).

Zoom Image
Scheme 4 Proposed mechanism

Table 1 Optimization of the Reaction Conditions for the Formation of 9a

Entry

PIDA (equiv)

NH3 (equiv)

Temp

Isolated yield (%)

1

1

 5.0

rt

 47

2

1

 5.0

0 °C to rt

 51

3

2

 5.0

0 °C to rt

 70

4

3

 5.0

0 °C to rt

 50

5

2

10.0

0 °C to rt

 80

6

2

15.0

0 °C to rt

 85

7

2

17.5

0 °C to rt

>99

Based on this encouraging result, optimization of the reaction conditions was carried out using 4-methoxybenz­amide as a model substrate (Table [1]). Using the same amounts of methanolic ammonia and PIDA, the reaction performed at 0 °C afforded the desired urea 9a in a better yield than at room temperature (entries 1 and 2). Increasing the amount of PIDA to 2 equivalents improved the yield of the reaction, however, higher amounts were not beneficial (entries 3 and 4). Finally, 17.5 equivalents of methanolic ammonia were required to obtain the urea in a quantitative yield (entries 5–7).

With optimized conditions in hand (conditions A), different alkyl-, aryl- and heteroaryl-amides were converted into the corresponding primary ureas 9ar (Scheme [5]).

Zoom Image
Scheme 5 Scope of the reaction. a An additional amount of PIDA/NH3 was added and the reaction time was extended to 48 h.

Reactions with aromatic amides bearing electron-donating­ (9ac,n) or deactivating groups (9f,o) afforded the corresponding ureas in good to excellent yields, as well as pyridyl- (9h), phenyl- (9i) and benzylureas (9m). Although aliphatic amides were fully converted into the expected ureas (9k,l), the loss of small amounts of the products during purification was observed due to their volatility and consequently impacted their isolated yields. Furthermore, the reaction time was extended to 48 hours to achieve the synthesis of compounds 9g, 9j and 9p in high yields. On the other hand, the presence of an electron-withdrawing group on the aromatic amide seemed to lower the reaction yield of this transformation since 4-cyanophenylurea 9d was obtained in only 16% yield and 4-nitrophenylurea 9e could not be prepared using this procedure.[30] However, it was possible to increase the electrophilicity of PIDA by using a highly polar and strong hydrogen-bond donating solvent, such as 2,2,2-trifluoroethanol (TFE).[31] Moreover, we reasoned that using a slow ammonia release source, such as ammonium carbamate (AC), would improve the yield as we had already shown in the sulfoximination reaction of sulfides.[32] Hence, it was found that performing the reaction using conditions B (Scheme [5]) allowed the synthesis of compounds 9d and 9e in 95% and 98% yields, respectively. In contrast, a substrate with an aromatic ring bearing a 4-OH group failed to give the desired urea 9q,[33] whereas that with a 2-OH group gave the cyclic carbamate 9r isolated in quantitative yield. However, 2-furamide did not react under both sets of conditions.

In conclusion, an easy, mild and affordable method to convert primary carboxamides into N-substituted ureas has been developed by utilizing a PIDA-induced Hofmann rearrangement followed by addition of ammonia. The reactivity of the hypervalent reagent was increased by using TFE as the solvent and AC as the ammonia source, allowing the synthesis of aromatic ureas bearing electron-withdrawing groups. This reaction allows access to a wide range of alkyl-, aryl- and heteroarylureas which could potentially possess interesting biological activities.

Column chromatography was performed on Merck silica gel (230–400 mesh). Thin-layer chromatography was performed using Merck pre-coated, aluminium-backed silica gel plates. Melting points were determined using a Gallen-Kamp melting point apparatus. IR spectra were recorded on a PerkinElmer ATR-spectrum one spectrometer. 1H NMR spectra were recorded at 500 MHz or 600 MHz and 13C NMR spectra were recorded at 125 MHz or 150 MHz on Bruker Avance III 500 and Avance 600 Neo spectrometers. High-resolution mass spectrometry (HRMS) was performed using a Xevo G2-XS QTof Waters mass spectrometer equipped with an electrospray ion source (ESI) operated in positive ion mode.


#

N-Substituted Ureas; General Procedure A

(Diacetoxyiodo)benzene (1.0 mmol, 2.0 equiv) was added in one portion to a stirred solution of the amide (0.5 mmol, 1.0 equiv) in NH3/MeOH (7 M, 1.25 mL, 17.5 equiv) at 0 °C under argon. After 30 min at 0 °C, the reaction mixture was allowed to reach room temperature and was left to stir for 90 min. After completion (monitored by TLC and 1H NMR), the reaction mixture was concentrated under reduced pressure and the crude product was purified by flash chromatography on silica gel.


#

N-Substituted Ureas; General procedure B

(Diacetoxyiodo)benzene (1.0 mmol, 2.0 equiv) was added in one portion to a stirred solution of the amide (0.5 mmol, 1.0 equiv) and ammonium carbamate (AC) (0.75 mmol, 1.5 equiv) in trifluoroethanol (1.25 mL) at 0 °C under argon. After 30 min at 0 °C, the reaction mixture was allowed to reach room temperature and was left to stir for 9 h. Additional amounts of PIDA and AC (1.0 equiv each) were added at rt and the reaction mixture was stirred for 12 h at rt. After concentration of the reaction mixture under reduced pressure, the crude product was purified by flash chromatography on silica gel.


#

1-(4-Methoxyphenyl)urea (9a)

Prepared according to General Procedure A using 4-methoxybenz­amide (75.6 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9a (83.1 mg, 99%) as a brown solid.

Mp 163 °C; Rf = 0.2 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3469, 3301, 1643, 1542, 1215, 1037, 822, 561 cm–1.

1H NMR (600 MHz, DMSO-d 6): δ = 8.30 (s, 1 H, NH), 7.28 (d, J = 9.0 Hz, 2 H, CHAr ), 6.80 (d, J = 9.0 Hz, 2 H, CHAr ), 5.71 (s, 2 H, NH2 ), 3.68 (s, 3 H, OCH3 ).

13C NMR (150 MHz, DMSO-d 6): δ = 156.2 (CO), 153.9 (CAr), 133.7 (CAr), 119.4 (CAr), 113.8 (CAr), 55.1 (OCH3).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C8H11N2O2: 167.0821; found: 167.0821.


#

1-(2-Ethoxyphenyl)urea (9b)

Prepared according to General Procedure A using 2-ethoxybenzamide (82.6 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9b (90.1 mg, 99%) as a brown solid.

Mp 115 °C; Rf  = 0.3 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3484, 3328, 3198, 1661, 1525, 1450, 1249, 1045, 746 cm–1.

1H NMR (600 MHz, DMSO-d 6): δ = 8.07 (dd, J = 7.8, 1.8 Hz, 1 H, CHAr ), 7.75 (s, 1 H, NH), 6.93 (dd, J = 7.8, 1.8 Hz, 1 H, CHAr ), 6.79–6.85 (m, 2 H, CHAr ), 6.24 (br s, 2 H, NH2 ), 4.08 (q, J = 7.0 Hz, 2 H, OCH2 CH3), 1.38 (t, J = 7.0 Hz, 3 H, OCH2CH3 ).

13C NMR (150 MHz, DMSO-d 6): δ = 156.0 (CO), 146.5 (CAr), 129.7 (CAr), 121.0 (CAr), 120.4 (CAr), 118.3 (CAr), 111.6 (CAr), 63.8 (OCH2CH3), 14.7 (OCH2 CH3).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C9H13N2O2: 181.0977; found: 181.0977.


#

1-(p-Tolyl)urea (9c)

Prepared according to General Procedure A using p-toluamide (67.6 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9c (71.3 mg, 95%) as a white solid.

Mp 180 °C; Rf  = 0.4 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3422, 3306, 1650, 1589, 1546, 1354, 811, 550, 501 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 8.37 (s, 1 H, NH), 7.26 (d, J = 8.1 Hz, 2 H, CHAr ), 7.01 (d, J = 8.1 Hz, 2 H, CHAr ), 5.75 (s, 2 H, NH2 ), 2.20 (s, 3 H, CH3 ).

13C NMR (125 MHz, DMSO-d 6): δ = 156.0 (CO), 138.0 (CAr), 129.7 (CAr), 129.0 (CAr), 117.8 (CAr), 20.3 (CH3).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C8H11N2O: 151.0871; found: 151.0873.


#

1-(4-Cyanophenyl)urea (9d)

Prepared according to General Procedure B using 4-cyanobenzamide (73.1 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/EtOH, 97:3) to afford urea 9d (76.5 mg, 95%) as a white solid.

Mp 220 °C; Rf  = 0.2 (CH2Cl2/EtOH, 97:3).

IR (ATR): 3484, 3379, 2219, 1678, 1587, 1538, 1361, 835, 510 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 9.07 (s, 1 H, NH), 7.65 (d, J = 8.5 Hz, 2 H, CHAr ), 7.57 (d, J = 8.5 Hz, 2 H, CHAr ), 6.12 (s, 2 H, NH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 155.5 (CO), 145.1 (CAr), 133.1 (CAr), 119.5 (CN), 117.5 (CAr), 102.4 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C8H8N3O: 162.0667; found: 162.0667.


#

1-(4-Nitrophenyl)urea (9e)

Prepared according to General Procedure B using 4-nitrobenzamide (83.1 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/EtOH, 97:3) to afford urea 9e (88.7 mg, 98%) as a white solid.

Mp 225 °C; Rf  = 0.1 (CH2Cl2/EtOH, 97:3).

IR (ATR): 3486, 3378, 1687, 1547, 1483, 1316, 1095, 854, 692 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 9.31 (br s, 1 H, NH), 8.13 (d, J = 9.1 Hz, 2 H, CHAr ), 7.63 (d, J = 9.1 Hz, 2 H, CHAr ), 6.22 (br s, 2 H, NH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 155.3 (CO), 147.3 (CAr), 140.4 (CAr), 125.1 (CAr), 116.9 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C7H8N3O3: 182.0566; found: 182.0567.


#

1-(4-Bromophenyl)urea (9f)

Prepared according to General Procedure A using 4-bromobenzamide (100.0 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9f (78.0 mg, 73%) as a white solid.

Mp 226 °C; Rf  = 0.3 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3417, 3306, 3213, 1651, 1582, 1544, 1486, 1353, 815, 586 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 8.67 (s, 1 H, NH), 7.37 (s, 4 H, CHAr ), 5.91 (s, 2 H, NH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 155.8 (CO), 140.0 (CAr), 131.3 (CAr), 119.6 (CAr), 112.3 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C7H8N2OBr: 214.9820; found: 214.9832.


#

1-(4-Chlorophenyl)urea (9g)

Prepared according to General Procedure A using 4-chlorobenzamide (77.8 mg, 0.5 mmol). Additional amounts of PIDA (0.25 mmol) and NH3 in MeOH (7 M, 0.3 mL) were added after 16 h and the mixture was left stirring for an additional 24 h. The mixture was concentrated under reduced pressure and the residue was purified by flash chromatography on silica gel (CH2Cl2/EtOH, 95:5) to afford urea 9g (76.8 mg, 90%) as a white solid.

Mp 209 °C; Rf  = 0.3 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3419, 3311, 1651, 1545, 1490, 1090, 820, 585, 489 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 8.66 (s, 1 H, NH), 7.42 (d, J = 8.5 Hz, 2 H, CHAr ), 7.24 (d, J = 8.5 Hz, 2 H, CHAr ), 5.90 (s, 2 H, NH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 155.8 (CO), 139.6 (CAr), 128.4 (CAr), 124.5 (CAr), 119.2 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C7H8N2OCl: 171.0325; found: 171.0328.


#

1-(Pyridin-3-yl)urea (9h)

Prepared according to General Procedure A using nicotinamide (66.2 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 90:10 + a few drops of NH3) to afford urea 9h (68.6 mg, 99%) as a yellow solid.

Mp 199 °C; Rf  = 0.3 (CH2Cl2/MeOH, 90:10 + a few drops of NH3).

IR (ATR): 3374, 3198, 1672, 1550, 1485, 1356, 1302, 571 cm–1.

1H NMR (600 MHz, DMSO-d 6): δ = 8.73 (s, 1 H, NH), 8.51 (d, J = 2.4 Hz, 1 H, CHAr ), 8.10 (dd, J = 4.6, 1.4 Hz, 1 H, CHAr ), 7.89 (ddd, J = 8.3, 2.6, 1.5 Hz, 1 H, CHAr ), 2.23 (ddd, J = 8.3, 4.6, 0.5 Hz, 1 H, CHAr ), 6.01 (br s, 2 H, NH2 ).

13C NMR (150 MHz, DMSO-d 6): δ = 156.0 (CO), 142.1 (CAr), 139.6 (CAr), 137.2 (CAr), 124.5 (CAr), 123.5 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C6H8N3O: 138.0667; found: 138.0670.


#

1-Phenylurea (9i)

Prepared according to General Procedure A using benzamide (60.6 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/EtOH, 97:3) to afford urea 9i (67.8 mg, >99%) as a beige solid.

Mp 149 °C; Rf  = 0.2 (CH2Cl2/EtOH, 97:3).

IR (ATR): 3420, 3311, 3214, 1651, 1590, 1548, 1353, 750, 694, 584 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 8.49 (s, 1 H, NH), 7.38 (d, J = 7.7 Hz, 2 H, CHAr ), 7.20 (t, J = 7.7 Hz, 2 H, CHAr ), 6.88 (t, J = 7.7 Hz, 1 H, CHAr ), 5.82 (s, 2 H, NH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 156.0 (CO), 140.6 (CAr), 128.6 (CAr), 121.0 (CAr), 117.7 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C7H9N2O: 137.0715; found: 137.0713.


#

1-Cyclohexylurea (9j)

Prepared according to General Procedure A using cyclohexanecarboxamide (63.6 mg, 0.5 mmol). Additional amounts of PIDA (0.15 mmol) and NH3 in MeOH (7 M, 0.18 mL) were added after 16 h and the mixture was left stirring for an additional 24 h. The mixture was concentrated under reduced pressure and the residue was purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9j (55.0 mg, 77%) as a white solid.

Mp 195 °C; Rf  = 0.2 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3328, 3196, 2928, 2852, 1649, 1544, 1351, 1157, 609 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 8.81 (d, J = 8.0 Hz, 1 H, NH), 5.28 (br s, 2 H, NH2 ), 3.26–3.32 (m, 1 H, CH), 1.71–1.73 (m, 2 H, CH 2 ), 1.61–1.64 (m, 2 H, CH 2 ), 1.50–1.52 (m, 1 H, CH), 1.20–1.28 (m, 2 H, CH2 ), 1.10–1.20 (m, 1 H, CHH), 1.01–1.10 (m, 2 H, CH).

13C NMR (125 MHz, DMSO-d 6): δ = 157.9 (CO), 47.6 (CAr), 33.3 (CAr), 25.3 (CAr), 24.5 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C7H15N2O: 143.1184; found: 143.1185.


#

1-Butylurea (9k)

Prepared according to General Procedure A using valeramide (50.6 mg; 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/EtOH, 9:1) to afford urea 9k (47.5 mg, 82%) as colorless needles.

Mp 98 °C; Rf  = 0.3 (CH2Cl2/EtOH, 90:10).

IR (ATR): 3411, 3348, 3208, 2968, 2932, 1554, 1339, 1157, 533 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 5.87 (s, 1 H, NH), 5.34 (s, 2 H, NH2 ), 2.93 (q, J = 6.5 Hz, 2 H, NHCH2 CH2), 1.23–1.34 (m, 4 H, H3CCH 2 CH2 CH2), 0.86 (t, J = 7.2 Hz, 3 H, CH3 ).

13C NMR (125 MHz, DMSO-d 6): δ = 158.7 (CO), 38.8 (CH2), 32.1 (CH2), 19.5 (CH2), 13.7 (CH3).

HRMS (ESI-QTOF): m/z [M + Na]+ calcd for C7H12N2ONa: 139.0847; found: 139.0844.


#

1-Ethylurea (9l)

Prepared according to General Procedure A using propionamide (73.1 mg, 1.0 mmol) and purified by flash chromatography on silica gel (CH2Cl2/EtOH, 9:1) to afford urea 9l (74.3 mg, 84%) as colorless needles.

Mp 83 °C; Rf  = 0.2 (CH2Cl2/EtOH, 90:10).

IR (ATR): 3350, 3207, 2931, 1548, 1340, 1157, 536 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 5.86 (s, 1 H, NH), 5.36 (s, 2 H, NH2 ), 2.96 (quin, J = 7.0 Hz, 2 H, CH2 ), 0.96 (t, J = 7.2 Hz, 3 H, CH3 ).

13C NMR (125 MHz, DMSO-d 6): δ = 158.6 (CO), 34.0 (CH2), 15.7 (CH3).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C3H9N2O: 89.0715; found: 89.0713.


#

1-Benzylurea (9m)

Prepared according to General Procedure A using 2-phenylacetamide (67.6 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9m (75.1 mg, 86%) as a white solid.

Mp 150 °C; Rf  = 0.3 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3428, 3328, 1647, 1598, 1557, 695, 583, 546 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 7.20–7.32 (m, 5 H, CHAr ), 6.40 (t, J = 6.0 Hz, 1 H, NH), 5.52 (br s, 2 H, NH2 ), 4.17 (d, J = 6.0 Hz, 2 H, CH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 158.7 (CO), 140.9 (CAr), 128.2 (CAr), 127.0 (CAr), 126.5 (CAr), 42.8 (CH2).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C8H11N2O: 151.0871; found: 151.0870.


#

1-(3-Ethoxyphenyl)urea (9n)

Prepared according to General Procedure A using 3-ethoxybenzamide (82.6 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9n (90.0 mg, 99%) as a brown solid.

Mp 112 °C; Rf  = 0.3 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3434, 3307, 3208, 1652, 1531, 1191, 1044, 766, 593 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 8.48 (s, 1 H, NH), 7.06–7.12 (m, 2 H, CHAr ), 6.83 (d, J = 8.0 Hz, 1 H, CHAr ), 6.44 (d, J = 8.0 Hz, 1 H, CHAr ), 5.82 (s, 2 H, NH2 ), 3.94 (q, J = 6.8 Hz, 2 H, CH2 ), 1.30 (t, J = 6.8 Hz, 3 H, CH3 ).

13C NMR (125 MHz, DMSO-d 6): δ = 158.9 (CO), 155.9 (CAr), 141.8 (CAr), 129.3 (CAr), 110.0 (CAr), 106.9 (CAr), 104.0 (CAr), 62.7 (CH2), 14.7 (CH3).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C9H13N2O2: 181.0977; found: 181.0977.


#

1-(2-Bromophenyl)urea (9o)

Prepared according to General Procedure A using 2-bromobenzamide (100.0 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9o (107.5 mg, 99%) as an off-white solid.

Mp 206 °C; Rf  = 0.3 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3419, 3287, 3196, 1651, 1516, 1474, 1354, 755, 539 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 8.00 (d, J = 8.2 Hz, 1 H, CHAr ), 7.85 (s, 1 H, NH), 7.54 (d, J = 7.8 Hz, 1 H, CHAr ), 7.26 (t, J = 7.8 Hz, 1 H, CHAr ), 7.89 (t, J = 7.8 Hz, 1 H, CHAr ), 6.40 (s, 2 H, NH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 155.6 (CO), 137.9 (CAr), 132.3 (CAr), 127.9 (CAr), 123.3 (CAr), 121.9 (CAr), 112.4 (CAr).

HRMS (ESI-QTOF): m/z [M + Na]+ calcd for C7H8N2OBrNa: 236.9639; found: 236.9640.


#

1-(3-Chlorophenyl)urea (9p)

Prepared according to General Procedure A using 3-chlorobenzamide (77.8 mg, 0.5 mmol). Additional amounts of PIDA (0.5 mmol) and NH3 in MeOH (7 M, 0.60 mL) were added after 4 h and the mixture was left to stir for an additional 24 h. The mixture was concentrated under reduced pressure and the residue was purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9p (76.2 mg, 89%) as a pale brown solid.

Mp 143 °C; Rf  = 0.2 (CH2Cl2/MeOH, 95:5).

1H NMR (500 MHz, DMSO-d 6): δ = 8.73 (s, 1 H, NH), 7.69 (t, J = 2.0 Hz, 1 H, CHAr ), 7.22 (t, J = 8.2 Hz, 1 H, CHAr ), 7.16 (ddd, J = 8.2, 1.9, 1.0 Hz, 1 H, CHAr ), 6.92 (ddd, J = 7.8, 2.0, 1.0 Hz, 1 H, CHAr ), 5.96 (s, 2 H, NH2 ).

13C NMR (125 MHz, DMSO-d 6): δ = 155.8 (CO), 142.2 (CAr), 133.1 (CAr), 130.2 (CAr), 120.6 (CAr), 117.0 (CAr), 116.0 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C7H8N2OCl: 171.0325; found: 171.0325.


#

2(3H)-Benzoxazolone (9r)

Prepared according to General Procedure A using salicylamide (68.6 mg, 0.5 mmol) and purified by flash chromatography on silica gel (CH2Cl2/MeOH, 95:5) to afford urea 9r (67 mg, 99%) as a brown solid.

Mp 140 °C; Rf  = 0.4 (CH2Cl2/MeOH, 95:5).

IR (ATR): 3201, 1732, 1478, 1253, 937, 686 cm–1.

1H NMR (500 MHz, DMSO-d 6): δ = 11.6 (s, 1 H, NH), 7.27 (d, J = 8.0 Hz, 1 H, CHAr ), 7.12–7.16 (m, 1 H, CHAr ), 7.06–7.09 (m, 2 H, CHAr ).

13C NMR (125 MHz, DMSO-d 6): δ = 154.4 (CO), 143.3 (CAr), 130.4 (CAr), 123.8 (CAr), 121.8 (CAr), 109.8 (CAr), 109.5 (CAr).

HRMS (ESI-QTOF): m/z [M + H]+ calcd for C7H6NO2: 136.0399; found: 136.0401.


#
#

Acknowledgment

We gratefully acknowledged K. Jarsalé for HRMS analyses, and H. El Slibani and R. Legay for NMR studies carried out at Caen.

Supporting Information

  • References

    • 1a Ghosh AK, Brindisi M. J. Med. Chem. 2020; 63: 2751
    • 1b Jagtap AD, Kondekar NB, Sadani AA, Chern JW. Curr. Med. Chem. 2017; 24: 622
    • 1c Cioffi CL, Dobri N, Freeman EE, Conlon MP, Chen P, Stafford DG, Schwarz DM. C, Golden KC, Zhu L, Kitchen DB, Barnes KD, Racz B, Qin Q, Michelotti E, Cywin CL, Martin WH, Pearson PG, Johnson G, Petrukhin K. J. Med. Chem. 2014; 57: 7731
    • 1d Belfrage AK, Gising J, Svensson F, Åkerblom E, Sköld C, Sandström A. Eur. J. Med. Chem. 2015; 978
    • 1e Nowotarski SL, Pachaiyappan B, Holshouser SL, Kutz CJ, Li Y, Huang Y, Sharma SK, Casero RA. Jr, Woster PM. Bioorg. Med. Chem. 2015; 23: 1601
    • 1f Darvesh S, Pottie IR, Darvesh KV, McDonald RS, Walsh R, Conrad S, Penwell A, Mataija D, Martin E. Bioorg. Med. Chem. 2010; 18: 2232
    • 1g Manickam M, Pillaiyar T, Boggu P, Venkateswararao E, Jalani HB, Kim N.-D, Lee SK, Jeon JS, Kim SK, Jung S.-H. Eur. J. Med. Chem. 2016; 117: 113
    • 1h Nepali K, Sharma S, Sharma M, Bedi PM. S, Dhar KL. Eur. J. Med. Chem. 2014; 77: 422
    • 1i Gong H, Yang M, Xiao YZ, Doweyko AM, Cunningham M, Wang J, Habte S, Holloway D, Burke C, Shuster D, Gao L, Carman J, Somerville JE, Nadler SG, Salter-Cid L, Barrish JC, Weinstein DS. Bioorg. Med. Chem. Lett. 2014; 24: 3268
    • 1j Li H.-Q, Lv PC, Yan T, Zhu H.-L. Anti-Cancer Agents Med. Chem. 2009; 9: 471
    • 2a Zakrzewski J, Krawczyk M. Heteroat. Chem. 2006; 17: 393
    • 2b Wada Y, Kamada Y, Hanaki K. US Patent 5833733, 1998
    • 2c Landes M, Sievernich B, Kibler E, Nuyken W, Walter H, Westphalen K.-O, Mayer H, Haden E, Mulder C, Schönhammer A, Hamprecht G. US Patent 6054410, 2000
    • 2d Achgill RK, Call LW. US Patent 4987233, 1991
    • 2e Arnold WR. US Patent 4165229, 1979
  • 3 Klingstedt F, Arve K, Eränen K, Murzin DY. Acc. Chem. Res. 2006; 39: 273
  • 4 Volz N, Clayden J. Angew. Chem. Int. Ed. 2011; 50: 12148
  • 5 Pullagurla MR, Rangisetty JB. US Patent 20190382447, 2019
  • 6 Guryanov I, Orlandin A, Viola A, Biondi B, Badocco D, Formaggio F, Ricci A, Cabri W. Org. Process Res. Dev. 2019; 23: 2746
  • 7 Oza V, Ashwell S, Almeida L, Brassil P, Breed J, Deng C, Gero T, Grondine M, Horn C, Ioannidis S, Liu D, Lyne P, Newcombe N, Pass M, Read J, Ready S, Rowsell S, Su M, Toader D, Vasbinder M, Yu D, Yu Y, Xue Y, Zabludoff S, Janetka J. J. Med. Chem. 2012; 55: 5130
  • 8 Cui K, Chen J, Wang M. PCT Int. Appl WO2016201662, 2016
  • 9 Luedtke NW, Liu Q, Tor Y. Bioorg. Med. Chem. 2003; 11: 5235
  • 10 Katayama N, Fukusumi S, Funabashi Y, Iwahi T, Ono H. J. Antibiot. 1993; 46: 606
    • 11a Lee S.-H, Yoshida K, Matsushita H, Clapham B, Koch G, Zimmermann J, Janda KD. J. Org. Chem. 2004; 69: 8829
    • 11b Lee S.-H, Clapham B, Koch G, Zimmermann J, Janda KD. Org. Lett. 2003; 5: 511
    • 11c Belfrage AK, Gising J, Svensson F, Åkerblom E, Sköld C, Sandström A. Eur. J. Org. Chem. 2015; 978
    • 11d Wu J, Xie Y, Chen X, Deng G.-J. Adv. Synth. Catal. 2016; 358: 3206
    • 11e Rakesh KP, Ramesha AB, Shantharam CS, Mantelingu K, Mallesha N. RSC Adv. 2016; 6: 108315
    • 11f Rekunge DS, Khatri CK, Chaturbhuj GU. Tetrahedron Lett. 2017; 58: 4304
  • 12 Wöhler F. Ann. Phys. Chem. 1828; 88: 253
  • 13 Browne DL, O’Brien M, Koos P, Cranwell PB, Polyzos A, Ley SV. Synlett 2012; 23: 1402
  • 14 Aubé J, Fehl C, Liu R, McLeod MC, Motiwala HF. In Comprehensive Organic Synthesis II, Vol. 6. Elsevier; Amsterdam: 2014: 598
  • 15 Loudon GM, Radhakrishna AS, Almond MR, Blodgett JK, Boutin RH. J. Org. Chem. 1984; 49: 4272
  • 16 Moriarty RM, Chany CJ. II, Vaid RK, Prakash O, Tuladhar SM. J. Org. Chem. 1993; 58: 2478
  • 18 Kumar A, Kumar N, Sharma R, Bhargava G, Mahajan D. J. Org. Chem. 2019; 84: 11323
  • 19 Thavonekham B. Synthesis 1997; 1189
  • 20 Nagarkar AG, Telvekar VN. Lett. Org. Chem. 2017; 15: 926
  • 21 Breitler S, Oldenhuis NJ, Fors BP, Buchwald SL. Org. Lett. 2011; 13: 3262
  • 22 Habibi D, Heydari S, Faraji A, Keypour H, Mahmoudabadi M. Polyhedron 2018; 151: 520
    • 23a Sardarian AR, Inaloo ID. RSC Adv. 2015; 5: 76626
    • 23b Tiwari L, Kumar V, Kumar B, Mahajan D. RSC Adv. 2018; 8: 21585
  • 24 Wang CH, Hsieh TH, Lin CC, Yeh WH, Lin CA, Chien TC. Synlett 2015; 26: 1823
  • 25 Das S, Natarajan P, König B. Chem. Eur. J. 2017; 23: 18161
  • 26 Glachet T, Marzag H, Saraiva Rosa N, Colell JF. P, Zhang G, Warren WS, Franck X, Theis T, Reboul V. J. Am. Chem. Soc. 2019; 141: 13689
  • 27 Single crystals suitable for X-ray crystallographic analysis were obtained by slow evaporation from an Et2O solution. CCDC 1896523 contains the supplementary crystallographic data for this paper. The data can be obtained free of charge from The Cambridge Crystallographic Data Centre via www.ccdc.cam.ac.uk/getstructures.
  • 28 No reaction occurred between (4-methoxyphenyl)methyl carbamate and ammonia in methanol at rt.
  • 29 Boutin RH, Loudon GM. J. Org. Chem. 1984; 49: 4277
  • 30 The same trend was observed with other methods: see references 18, 20 and 23b.
  • 31 Chaabouni S, Lohier JF, Barthelemy AL, Glachet T, Anselmi E, Dagousset G, Diter P, Pégot B, Magnier E, Reboul V. Chem. Eur. J. 2018; 24: 17006
  • 33 Pouységu L, Deffieux D, Quideau S. Tetrahedron 2010; 66: 2235

  • References

    • 1a Ghosh AK, Brindisi M. J. Med. Chem. 2020; 63: 2751
    • 1b Jagtap AD, Kondekar NB, Sadani AA, Chern JW. Curr. Med. Chem. 2017; 24: 622
    • 1c Cioffi CL, Dobri N, Freeman EE, Conlon MP, Chen P, Stafford DG, Schwarz DM. C, Golden KC, Zhu L, Kitchen DB, Barnes KD, Racz B, Qin Q, Michelotti E, Cywin CL, Martin WH, Pearson PG, Johnson G, Petrukhin K. J. Med. Chem. 2014; 57: 7731
    • 1d Belfrage AK, Gising J, Svensson F, Åkerblom E, Sköld C, Sandström A. Eur. J. Med. Chem. 2015; 978
    • 1e Nowotarski SL, Pachaiyappan B, Holshouser SL, Kutz CJ, Li Y, Huang Y, Sharma SK, Casero RA. Jr, Woster PM. Bioorg. Med. Chem. 2015; 23: 1601
    • 1f Darvesh S, Pottie IR, Darvesh KV, McDonald RS, Walsh R, Conrad S, Penwell A, Mataija D, Martin E. Bioorg. Med. Chem. 2010; 18: 2232
    • 1g Manickam M, Pillaiyar T, Boggu P, Venkateswararao E, Jalani HB, Kim N.-D, Lee SK, Jeon JS, Kim SK, Jung S.-H. Eur. J. Med. Chem. 2016; 117: 113
    • 1h Nepali K, Sharma S, Sharma M, Bedi PM. S, Dhar KL. Eur. J. Med. Chem. 2014; 77: 422
    • 1i Gong H, Yang M, Xiao YZ, Doweyko AM, Cunningham M, Wang J, Habte S, Holloway D, Burke C, Shuster D, Gao L, Carman J, Somerville JE, Nadler SG, Salter-Cid L, Barrish JC, Weinstein DS. Bioorg. Med. Chem. Lett. 2014; 24: 3268
    • 1j Li H.-Q, Lv PC, Yan T, Zhu H.-L. Anti-Cancer Agents Med. Chem. 2009; 9: 471
    • 2a Zakrzewski J, Krawczyk M. Heteroat. Chem. 2006; 17: 393
    • 2b Wada Y, Kamada Y, Hanaki K. US Patent 5833733, 1998
    • 2c Landes M, Sievernich B, Kibler E, Nuyken W, Walter H, Westphalen K.-O, Mayer H, Haden E, Mulder C, Schönhammer A, Hamprecht G. US Patent 6054410, 2000
    • 2d Achgill RK, Call LW. US Patent 4987233, 1991
    • 2e Arnold WR. US Patent 4165229, 1979
  • 3 Klingstedt F, Arve K, Eränen K, Murzin DY. Acc. Chem. Res. 2006; 39: 273
  • 4 Volz N, Clayden J. Angew. Chem. Int. Ed. 2011; 50: 12148
  • 5 Pullagurla MR, Rangisetty JB. US Patent 20190382447, 2019
  • 6 Guryanov I, Orlandin A, Viola A, Biondi B, Badocco D, Formaggio F, Ricci A, Cabri W. Org. Process Res. Dev. 2019; 23: 2746
  • 7 Oza V, Ashwell S, Almeida L, Brassil P, Breed J, Deng C, Gero T, Grondine M, Horn C, Ioannidis S, Liu D, Lyne P, Newcombe N, Pass M, Read J, Ready S, Rowsell S, Su M, Toader D, Vasbinder M, Yu D, Yu Y, Xue Y, Zabludoff S, Janetka J. J. Med. Chem. 2012; 55: 5130
  • 8 Cui K, Chen J, Wang M. PCT Int. Appl WO2016201662, 2016
  • 9 Luedtke NW, Liu Q, Tor Y. Bioorg. Med. Chem. 2003; 11: 5235
  • 10 Katayama N, Fukusumi S, Funabashi Y, Iwahi T, Ono H. J. Antibiot. 1993; 46: 606
    • 11a Lee S.-H, Yoshida K, Matsushita H, Clapham B, Koch G, Zimmermann J, Janda KD. J. Org. Chem. 2004; 69: 8829
    • 11b Lee S.-H, Clapham B, Koch G, Zimmermann J, Janda KD. Org. Lett. 2003; 5: 511
    • 11c Belfrage AK, Gising J, Svensson F, Åkerblom E, Sköld C, Sandström A. Eur. J. Org. Chem. 2015; 978
    • 11d Wu J, Xie Y, Chen X, Deng G.-J. Adv. Synth. Catal. 2016; 358: 3206
    • 11e Rakesh KP, Ramesha AB, Shantharam CS, Mantelingu K, Mallesha N. RSC Adv. 2016; 6: 108315
    • 11f Rekunge DS, Khatri CK, Chaturbhuj GU. Tetrahedron Lett. 2017; 58: 4304
  • 12 Wöhler F. Ann. Phys. Chem. 1828; 88: 253
  • 13 Browne DL, O’Brien M, Koos P, Cranwell PB, Polyzos A, Ley SV. Synlett 2012; 23: 1402
  • 14 Aubé J, Fehl C, Liu R, McLeod MC, Motiwala HF. In Comprehensive Organic Synthesis II, Vol. 6. Elsevier; Amsterdam: 2014: 598
  • 15 Loudon GM, Radhakrishna AS, Almond MR, Blodgett JK, Boutin RH. J. Org. Chem. 1984; 49: 4272
  • 16 Moriarty RM, Chany CJ. II, Vaid RK, Prakash O, Tuladhar SM. J. Org. Chem. 1993; 58: 2478
  • 18 Kumar A, Kumar N, Sharma R, Bhargava G, Mahajan D. J. Org. Chem. 2019; 84: 11323
  • 19 Thavonekham B. Synthesis 1997; 1189
  • 20 Nagarkar AG, Telvekar VN. Lett. Org. Chem. 2017; 15: 926
  • 21 Breitler S, Oldenhuis NJ, Fors BP, Buchwald SL. Org. Lett. 2011; 13: 3262
  • 22 Habibi D, Heydari S, Faraji A, Keypour H, Mahmoudabadi M. Polyhedron 2018; 151: 520
    • 23a Sardarian AR, Inaloo ID. RSC Adv. 2015; 5: 76626
    • 23b Tiwari L, Kumar V, Kumar B, Mahajan D. RSC Adv. 2018; 8: 21585
  • 24 Wang CH, Hsieh TH, Lin CC, Yeh WH, Lin CA, Chien TC. Synlett 2015; 26: 1823
  • 25 Das S, Natarajan P, König B. Chem. Eur. J. 2017; 23: 18161
  • 26 Glachet T, Marzag H, Saraiva Rosa N, Colell JF. P, Zhang G, Warren WS, Franck X, Theis T, Reboul V. J. Am. Chem. Soc. 2019; 141: 13689
  • 27 Single crystals suitable for X-ray crystallographic analysis were obtained by slow evaporation from an Et2O solution. CCDC 1896523 contains the supplementary crystallographic data for this paper. The data can be obtained free of charge from The Cambridge Crystallographic Data Centre via www.ccdc.cam.ac.uk/getstructures.
  • 28 No reaction occurred between (4-methoxyphenyl)methyl carbamate and ammonia in methanol at rt.
  • 29 Boutin RH, Loudon GM. J. Org. Chem. 1984; 49: 4277
  • 30 The same trend was observed with other methods: see references 18, 20 and 23b.
  • 31 Chaabouni S, Lohier JF, Barthelemy AL, Glachet T, Anselmi E, Dagousset G, Diter P, Pégot B, Magnier E, Reboul V. Chem. Eur. J. 2018; 24: 17006
  • 33 Pouységu L, Deffieux D, Quideau S. Tetrahedron 2010; 66: 2235

Zoom Image
Figure 1 Biologically active compounds containing a primary urea moiety
Zoom Image
Scheme 1 Reactions of carboxamides with hypervalent iodine species
Zoom Image
Scheme 2 Literature methods for the preparation of primary ureas
Zoom Image
Scheme 3 Formation of diazirine 7 (from l-glutamine) and its X-ray crystal structure (packing with H-bonding)
Zoom Image
Scheme 4 Proposed mechanism
Zoom Image
Scheme 5 Scope of the reaction. a An additional amount of PIDA/NH3 was added and the reaction time was extended to 48 h.