Raw Material - 2020

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 4

Tetrahedron Letters 63 (2021) 152718

Contents lists available at ScienceDirect

Tetrahedron Letters
journal homepage: www.elsevier.com/locate/tetlet

Conversion of a readily available carbohydrate raw material into a rare


L-deoxyhexose

Ya-Han Hsu, Che-Chien Chang ⇑


Department of Chemistry, Fu Jen Catholic University, 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City 24205, Taiwan

a r t i c l e i n f o a b s t r a c t

Article history: The highly efficient conversion of D-fructose to a L-deoxyhexose is reported. Starting from D-fructose, ben-
Received 24 October 2020 zoyl protection followed by bromination provided an acyclic radical precursor. Using environmentally
Revised 20 November 2020 benign radical conditions (TTMSS/AIBN), a benzoyl-protected 3-deoxy-L-fructose derivative was pro-
Accepted 26 November 2020
duced through a carbonyl translocation process. This unique radical reaction involves the highly efficient
Available online 24 December 2020
translocation of an a-oxy carbonyl group to produce L-deoxysugars from D-sugars. The benzoyl protecting
groups were enzymatically removed by treatment with a lipase (Candida rugosa) to produce the free 3-
Keywords:
deoxy-L-fructose in only four synthetic steps in an overall yield of 40%. The synthesis uses a cheap, readily
L-deoxysugar
Raw material
available carbohydrate raw material as the source and avoids the use of toxic chemicals, such as tin
Rare reagents. This synthetic methodology is concise, providing one of the most efficient methods for prepar-
Conversion ing L-deoxyhexose derivatives from a readily available carbohydrate raw material.
Concise Ó 2020 Elsevier Ltd. All rights reserved.

Introduction reaction at a specific HO group, as shown in Scheme 1. This


represents a challenging synthetic problem for organic chemists.
L-Deoxysugars are less abundant in nature, compared to their One of the important tasks for organic chemists is to develop
enantiomers, D-deoxysugars [1]. L-Deoxysugars are biological practical synthetic methods for converting inexpensive starting
important molecules, which are found in many important natural materials into rare, expensive, or important organic molecules
products [2]. They are subjects of research in a broad range of [9]. Further, using raw materials as starting materials, developing
fields, ranging from nutritional science to the pharmaceutical concise synthesis, employing environmentally benign reaction
industry [3]. They also serve as carbohydrate building blocks for conditions, and simplifying the purification steps are goals associ-
the synthesis of antitumor and antiviral agents [4], as well as ated with most organic transformations. Among all raw materials
L-DNA and L-deoxynucleosides [5]. Because they are available in from nature, carbohydrates are versatile starting materials for
limited amounts in nature and are in increasing demand in the the synthesis of many chiral organic molecules [10]. They also
pharmaceutical sciences, several methods for their synthesis have could serve as chiral building blocks for the synthesis of commer-
been explored [6–8]. Although the synthesis of L-deoxysugars from cially available pharmaceuticals [11].
To reach this synthetic challenge, a unique radical reaction
L-sugars has been reported [6], the use of D-sugars as starting mate-
involving a carbonyl translocation process [12] was recently devel-
rials would be more practical [7]. The currently available methods
oped in this laboratory, which could allow rare L-deoxysugars to be
for preparing L-hexoses mainly rely on isomerization/conversion of
prepared from inexpensive D-sugars [13]. To cope with the limita-
a D-hexose derivative, chain extension of a pentose, or HO group
tion of this synthetic methodology, an ideal approach would be to
inversion of another D-hexose [8]. Regarding the synthesis of
develop a highly efficient method (only a few synthetic steps) for
L-deoxyhexoses,the conversion of D-hexoses to L-deoxyhexoses is preparing L-deoxysugars from a raw material. D-fructose, a carbo-
complicated and requires multi-synthetic steps. It is necessary to hydrate raw material, is the second cheapest monosaccharide,
convert all of the stereogenic centers of a D-sugar to an found in nature and over 240,000 tons of crystalline D-fructose
L-configuration, followed by a highly selective deoxygenation are produced annually. The conversion of D-fructose to other
important organic molecules has been widely studied [14]. How-
ever, synthesis of L-deoxysugars from D-fructose was not reported,
except by us [13b], as shown in Scheme 2.
⇑ Corresponding author.
E-mail address: 080686@mail.fju.edu.tw (C.-C. Chang).

https://doi.org/10.1016/j.tetlet.2020.152718
0040-4039/Ó 2020 Elsevier Ltd. All rights reserved.
Ya-Han Hsu and Che-Chien Chang Tetrahedron Letters 63 (2021) 152718

by Chatgilialoglu, tris(trimethylsilyl)silane (TTMSS) was


successfully used as an alternative radical reagent to avoid the
use of toxic tin reagents [16]. Under these unique radical condi-
tions (TTMSS/AIBN), the a-oxy carbonyl group was successfully
translocated to produce the protected 3-deoxy-L-fructose deriva-
tive 3 in 76% yield, where the C(3) position was deoxygenated,
Scheme 1. Synthetic strategies for synthesis of L-deoxyhexoses from D-hexoses. and the stereochemistry of C(4) and C(5) were inverted [13b].
Conventional procedures to deprotect the benzoyl (Bz) group of
compound 3, such as acidic methanolysis, Cu(OAc)2 assisted
Although we experienced success in developing a carbonyl
methanolysis [17], hydrazine monohydrate [18], NaOEt, KCN, and
translocation reaction to produce an L-deoxyhexose derivative
thiophenoxide [19], were all not feasible. (please see the Supple-
from D-fructose, the direct deprotection of the benzoyl groups in
mentary Material) We then turned our attention to exploring bio-
the resulting sensitive molecule, which contains a-acidic protons
logical deprotection methods. Since the use of lipases in the
(Ha) and a b-leaving group (b-OBz), met with failure, as shown in
enzymatic hydrolysis of acetates (OAc) and benzoates (OBz) is well
Scheme 2 [13b]. Elimination reactions occurred which were
established in organic synthesis [20], several lipases from different
accompanied by the generation of an undesired product with an
sources (Aspergillus oryzae, Candida Cyclindracea, Candida rugose,
a,b-unsaturated system. Alternatively, indirect procedures using
and Porcine Pancreas), and different benzoyl group acceptors (n-
dithioacetal protection of the carbonyl group were applied to
BuOH, n-hexanol, and n-octanol), and a variety of reaction solvents
eventually afford 3-deoxy-L-fructose. Because the dithioacetal
(phosphate buffer, DMSO/H2O, THF, tBuOMe, diisopropyl ether,
approach was used, two additional synthetic steps (protection
and toluene) were tested. Eventually, a lipase from Candida rugose
and deprotection of the carbonyl group) were needed to
in the presence of n-BuOH and toluene at 37 °C was found to be the
hydrolyze the benzoyl groups and produce the desired 3-deoxy-
best conditions for accomplishing the whole synthesis and produc-
L-fructose. From synthetic points of view, the benzoyl group
ing the free sugar form of 3-deoxy-L-fructose (4) in 59% yield.
functioned well as the protecting group in the carbonyl (please see the Supplementary material) Since the free sugar pro-
translocation process, but further deprotection remained a duct contains a- and b-forms of furanose and pyranose, compli-
challenge [15]. cated 1H/13C spectra were observed, which are identical to
previous work [13b]. We now confirm that D-fructose, a
carbohydrate raw material, could be easily and efficiently
Results and discussion converted into a rare L-deoxyhexose in just four steps in an
overall yield of 39.98%. This example using a carbonyl
Since the benzoyl (Bz) group was still the best protecting group translocation process and enzymatic hydrolysis opens a new
for this unique radical translocation process [12,13abd], we synthetic methodology that can be successfully used to prepare
decided to continue to use the benzoyl group for protecting hydro- rare, expensive, or important L-deoxysugars from carbohydrate
xyl groups, as shown in Scheme 3. To avoid the use of pyridine and raw materials.
DCM, benzoyl protection was first carried out in the presence of
NEt3 in THF, but the tetrabenzoyl fructopyranoside 1 was not suc-
cessfully obtained. Mixtures of tetrabenzoyl furanosides appeared Conclusions
to be generated in these conditions, which is not suitable for fur-
ther bromination reactions. We then examined the use of standard The highly efficient conversion of D-fructose into rare
conditions for achieving benzoyl protection (pyridine and DCM) to 3-deoxy-L-fructose was achieved in four synthetic steps. The first
give tetrabenzoyl fructopyranoside 1 in 91% yield [13b]. two synthetic steps, benzoyl protection and bromination reaction,
Bromination reaction proceeded successfully using CBr4 and PPh3 provided an acyclic carbohydrate precursor. The third step, using
to provide the acyclic precursor 2 in quantitative yield. Pioneered environmentally benign radical conditions (TTMSS/AIBN), allowed

Scheme 2. Configurations of D-fructose and 3-deoxy-L-fructose and carbonyl translocation process.

2
Ya-Han Hsu and Che-Chien Chang Tetrahedron Letters 63 (2021) 152718

Scheme 3. Conversion of D-fructose into 3-deoxy-L-fructose in four steps.

a 3-deoxy-L-fructose derivative to be produced through our unique Salas, J. A.; Bechthold, A. Genes and Enzymes Involved in Deoxysugar
Biosynthesis in Bacteria. Nat. Prod. Rep. 1999, 16, 283-299.
carbonyl translocation process, thus representing a highly efficient
[3] (a) C. Rupprath, T. Schumacher, L. Elling, Nucleotide deoxysugars: essential
approach to the production of L-deoxysugars from D-sugars. The tools for the glycosylation engineering of novel bioactive compounds, Curr.
fourth step, enzymatic deprotection using a lipase, resulted in Med. Chem. 12 (2005) 1637–1675;
the formation of the free sugar, 3-deoxy-L-fructose in an overall (b) X. He, H.-W. Liu, Formation of unusual sugars: mechanistic studies and
biosynthetic applications, Annu. Rev. Biochem. 71 (2002) 701–754;
yield of 40%. The synthesis used a readily available carbohydrate (c) T.M. Hallis, H.-W. Liu, Learning nature’s strategies for making deoxy
raw material as the starting material and avoided the use of toxic sugars: pathways, mechanisms, and combinatorial applications, Acc. Chem.
tin chemicals. To the best of our knowledge, this synthetic method- Res. 32 (1999) 579–588.
[4] (a) Vater, A.; Klussmann, S. Turning Mirroe-Image Oligonucleotides into Drugs:
ology is sufficiently concise to provide one of the most efficient The Evolution of SpiegelmerÒ Therapeutics. Drug. Discov. Today 20 (2015)
methods for preparing L-deoxyhexoses from a simple raw material. 147-155.
This method could also serve as the standard procedures for fur- (b) Perrone, D.; Capobianco, M. L. in Chemical Synthesis of Nucleoside
Analogues, (Ed: Merino, P.), John Wiley & Sons, Weinheim, 2013, p. 473-534.
ther applications in the pharmaceutical industry. Meanwhile, with (c) Mathé, C.; Gosselin, G. L-Nucleoside Enantiomers as Antivirals Drugs: A
this methodology in hand, the synthesis of other rare L-deoxysug- Mini-Review. Antivir. Res. 71 (2006) 276-281.
ars using other raw materials is currently being investigated. (d) Dukhan, D.; Leroy, F.; Peyronnet, J.; Bosc, E.; Chaves, D.; Durka, M.; Storer,
R.; La Colla, P.; Seela, F.; Gosselin, G. Synthesis of 5-Aza-7-deazaguanine
Nucleoside Derivatives as Potential Anti-flavivirus Agents. Nucleosides
Declaration of Competing Interest Nucleotides Nucleic Acids 24 (2005) 671-674.
(e) Lombó, F.; Gibsob, M.; Greenwell, L.; Brańa, A. F.; Rohr, J.; Jose, A.; Méndes,
C. Engineering Biosynthetic Pathways for Deoxysugars: Branched-chain Sugar
The authors declare that they have no known competing finan- Pathways and Derivatives from The Antitumor Tetracenomycin. Chem. Biol. 11
cial interests or personal relationships that could have appeared (2004) 1709-1781.
to influence the work reported in this paper. (f) Gumina, G.; Song, G.-Y.; Chu, C. K. L-Nucleosides as Chemotherapeutic
Agents. FEMS Microbiol. Lett. 202 (2001) 9-15.
(g) Graciet, J.-C, G.; Schinazi, R. F. From D- to L-Nucleoside Analogs as Antiviral
Acknowledgement Agents. Adv. Antiviral Drug Des. 3 (1999) 1-68.
[5] D. D’Alonzo, A. Guaragna, G. Palumbo, Exploring the role of chirality in nucleic
acid recognition, Chem. Biodiversity 8 (2011) 373–413.
We wish to thank the Ministry of Science and Technology, Tai- [6] (a) M.L. Sznaidman, M.R. Almond, P. Amir, New synthesis of L-FMAU from L-
wan (grant number: MOST 108-2113-M-030-008) for financial arabinose, Nucleosides Nucleotides 21 (2002) 155–163;
support. YHH held Department of Chemistry Master Student Fel- (b) M.E. June, Y. Xu, Efficient synthesis of 2-deoxy L-ribose from L-arabinose:
mechanistic information on the 1,2-acyloxy shift in alkyl radicals, Org. Lett. 1
lowships. Tai-Ni Lu is acknowledged for prelimary experiments. (1999) 1517–1519;
(c) W. Zhang, K.S. Ramasamy, D.R. Averett, Improved synthesis of 2-deoxy-L-
ribose, Nucleosides Nucleotides 18 (1999) 2357–2365;
Appendix A. Supplementary data (d) E. Moyroud, P. Strazewski, L-ribonucleosides from L-xylose, Tetrahedron
55 (1999) 1277–1284;
Supplementary data to this article can be found online at (e) J. Du, Y. Choi, K. Lee, B.K. Chun, J.H. Hong, C.K. Chu, A practical synthesis of
L-FMAU from L-arabinose, Nucleosides Nucleotides 18 (1999) 187–195.
https://doi.org/10.1016/j.tetlet.2020.152718.
[7] (a) Q. Ji, M. Pang, J. Han, S. Feng, X. Zhang, Y. Ma, J. Meng, A simple and efficient
synthesis of 2-deoxy-L-ribose from 2-deoxy-D-ribose, Synlett. 15 (2006)
References 2498–2500;
(b) Z.-D. Shi, B.-H. Yang, Y.-L. Wu, A stereospecific synthesis of L-deoxyribose,
L-ribose, and L-ribosides, Tetrahedron 58 (2002) 3287–3296;
[1] (a) M. Nuedo, G. Copper, S.A. Sandford, Deoxyribose and deoxysugar
(c) M.E. Jung, Y. Xu, Efficient syntheses of L-ribose and 2-deoxy-L-ribose from
derivatives from photoprocessed astrophysical ice analogues and comparsion
D-ribose and L-arabinose, Tetrahedron Lett. 38 (1997) 4199–4202;
to meteorites, Nat. Comm. 9 (2018) 5276;
(d) M.E. Jung, C.J. Nichols, O. Kretscik, Y. Xu, Synthesis and testing of new
(b) R.M. de Lederkremer, C. Marino, Deoxy sugars: occurrence and synthesis,
modified nucleotides, Nucleosides Nucelotides 18 (1999) 541–546.
Adv. Carbohydr. Chem. Biochem. 61 (2007) 143–216.
[8] (a) T.G. Frihed, M. Bols, C.M. Pedersen, Synthesis of L-Hexoses, Chem. Rev. 115
[2] (a) Johnson, D. A.; Liu, H.-W. in Comprehensive Natural product Chemistry,
(2015) 3615–3676;
(Eds.: Pinto, P. M.), Elsevier, Amsterdam, 1999, p. 311-365. (b) Trefzer, A.;

3
Ya-Han Hsu and Che-Chien Chang Tetrahedron Letters 63 (2021) 152718

(b) M.M. Zulueta, Y.-Q. Zhong, S.-C. Hung, Synthesis of L-hexoses and their [16] (a) C. Chatgilialoglu, C. Ferreri, Y. Landais, V.I. Timokhin, Thirty years of (TMS)
related biomolecules, Chem. Commun. 49 (2013) 3275–3287; 3SiH: A milestone in radical-based synthetic chemistry, Chem. Rev. 118 (2018)
(c) D. D’Alonzo, A. Guaragna, G. Palumbo, Recent advances in monosaccharide 6516–6572;
synthesis: a journey into L-hexose world, Curr. Org. Chem. 13 (2009) 71–98; (b) C. Chatgilialoglu, (Me3Si)3SiH: twenty years after its discovery as a
(d) A. Kirschning, M. Jesberger, K.-U. Schöning, Concepts for the total synthesis radical-based reducing agent, Chem. Eur. J. 14 (2008) 2310–2320;
of deoxy sugars, Synthesis 4 (2001) 507–540. (c) C. Chatgilialoglu, J. Lalevee, Recent applications of the (TMS)3SiH radical-
[9] K.C. Nicolaou, T. Montagnon, Molecules That Changed the World, WILEY-VCH, based reagent, Molecules 17 (2012) 527–555;
Weinheim, 2008. (d) C. Chatgilialoglu, Tris(trimethylsilyl)silane. A new reducing agent, J. Org.
[10] (a) H.V. Bekkum, H. Röper, A.G. Vorhaben, Carbohydrates and Organic Raw Chem. 53 (1988) 3641–3642.
Materials III, WILEY-VCH, 1996; [17] T. Sammakia, J.S. Jacobs, Picolinic acid as a partner in the mitsunobu reaction:
(b) B. Ernst, G.W. Hart, P. Sinaÿ, Carbohydrates in Chemistry and Biology, subsequent hydrolysis of picolinate esters under essentially neutral conditions
WILEY-VCH, 2000; with copper acetate in methanol, Tetrahedron Lett. 40 (1999) 2685–2688.
(c) P.G. Wang, C.R. Bertozzi, Glycochemistry, Marcel Dekker, 2001. [18] Y. Ishido, N. Nakazaki, N. Sakairi, Partial protection of carbohydrate
[11] (a) M. Bols, Carbohydrate Building Blocks, John Wiley & Sons, 1996; derivatives. Part 3. Regioselective 20 -O-deacylation of fully acylated purine
(b) D.E. Levy, P. Fügedi, The Organic Chemistry of Sugars, Taylor & Francis, and pyrimidine ribonucleosides with hydrazine hydrate, J. Chem. Soc. Perkin
2006; Trans. I (1979) 2088–2098.
(c) S. Hanessian, Preparative Carbohydrate Chemistry, Marcel Dekker, 1997. [19] J. Herzig, A. Nudelman, H.E. Gottlieb, B. Fischer, Studies in sugar chemistry. 2. A
[12] L.-A. Chien, C.-C. Chang, Radical cyclization followed by the fragmentation of simple method for O-deacylation of polyacylated sugars, J. Org. Chem. 51
carbonyl compounds: effect of an a-benzoyl group, J. Org. Chem. 80 (2015) (1986) 727–730.
11294–11301. [20] (a) J. Parve, I. Reile, T. Aid, M. Kudrjašova, A.-M. Müürisepp, I. Vallikivi, L. Villo,
[13] (a) W.-S. Song, S.-X. Liu, C.-C. Chang, Synthesis of L-deoxyribonucleosides R. Aav, T. Pehk, L. Vares, O. Parve, Lipase-catalyzed stereosolution of long-chain
from D-ribose, J. Org. Chem. 83 (2018) 14923–14932; 1,2-alkanediols: A screening of preferable reaction conditions, J. Mol. Cat. B:
(b) T.-N. Lu, C.-C. Chang, Synthesis of 3-deoxy-L-ketohexoses through group Enzymatic 116 (2015) 60–69;
transfer, J. Org. Chem. 81 (2016) 469–475; (b) E. Santaniello, P. Ciuffreda, S. Casati, L. Alessandrini, A. Repetto, Selective
(c) H.-P. Wu, N.-Y. Hsu, T.-N. Lu, C.-C. Chang, Chemical synthesis of 1-deoxy-L- lipase-catalyzed preparation of diol monobenzoates by transesterification and
fructose and L-sorbose through carbonyl translocation, Eur. J. Org. Chem. 378– alcoholysis reactions in organic solvents, J. Mol. Cat. B: Enzymatic 40 (2006)
382 (2015); 81–85;
(d) N.-Y. Hsu, C.-C. Chang, A unique synthetic method to convert a D-sugar (c) E. Santaniello, S. Casati, P. Ciuffreda, L. Gamberoni, Lipase-catalyzed
into 2-deoxy-L-ribitol through carbonyl translocation, Eur. J. Org. Chem. 658– alcoholysis of diol dibenzoates: selective enzymatic access to the 2-benzoyl
661 (2013). ester of 1,2-propanediol and preparation of the enantiomerically pure (R)-1-O-
[14] J. Esteban, P. Yustos, M. Ladero, Catalytic processes from biomass-drived benzoyl-2-methylpropane-1,3-diol, Tetrahedron: Asymmetry 16 (2005) 1705–
hexoses and pentoses: a recent literature overview, Catalysts 8 (2018) 637. 1708;
[15] An ideal approach is to develop a synthetic method to prepare rare L- (d) P. Ciuffreda, L. Alessandrini, G. Terraneo, E. Santaniello, Lipase-catalyzed
deoxysugar from inexpensive D-sugars within four steps, which is presumably selective benzoylation of 1,3-Diols with vinyl benzoate in organic solvents,
suitable for industry production. Tetrahedron: Asymmetry 14 (2003) 3197–3201.

You might also like