Next Article in Journal
The Apicomplexan Parasite Toxoplasma gondii
Previous Article in Journal
Mechanics and Mathematics in Ancient Greece
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Entry

Enantiomers and Their Resolution

by
Rodrigo Santos
1,2,
Karen V. Pontes
1,* and
Idelfonso B. R. Nogueira
2,*
1
Programa de Pós-Graduação em Engenharia Industrial (Industrial Engineering Program), Escola Politécnica (Polytechnique Institut), Universidade Federal da Bahia, Salvador 40210-630, Brazil
2
Laboratory of Separation and Reaction Engineering, Associate Laboratory LSRE-LCM, Department of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
*
Authors to whom correspondence should be addressed.
Encyclopedia 2022, 2(1), 151-188; https://doi.org/10.3390/encyclopedia2010011
Submission received: 19 December 2021 / Revised: 7 January 2022 / Accepted: 12 January 2022 / Published: 14 January 2022
(This article belongs to the Section Chemistry)

Definition

:
Enantiomers share the same chemical formula but have different chemical structures, i.e., type of isomers. Enantiomers are present in several drugs, perfumes, food, and are a fundamental part of biomolecules. This subject is highly important for pharmaceutical companies. Enantiomeric drugs present different actuation in the human body; depending on the compound, one might combat the symptom, whereas its pair might cause damage. The separation of pairs of enantiomers requires a chiral environment that provokes a structural imbalance that conventional methods cannot provide. Enantioresolution is one of the most promissory studies that benefit several areas, such as pharmaceutical, biotechnology, food industry, and fine chemistry. Its resolution is of great importance, therefore, its main mechanisms of resolution will be explained herein.

1. Introduction

Isomers are different compounds that share the same chemical formula but have different chemical structures. They are classified into structural and stereoisomers. Structural isomers might be subclassified into function, chain, position, metamerism and tautomerism, as exemplified in Table 1, the latter only existing in equilibrium. Stereoisomers are subclassified into diastereomers (diastereoisomers) and enantiomers (optical isomers). Isomers have different physicochemical properties such as their melting point, boiling point, solubility, and density. Enantiomers, unlike other isomers, share all these properties, but optical activity. Enantiomers are analogous to a pair of hands, an enantiomer is a specular image of the other.
Enantiomerism is present in several drugs, perfumes, food, and even in our own body. The response of human organisms to certain enantiomeric compounds might change dramatically depending on the enantiomer: one enantiomer in a drug might treat the disease whereas the other might cause harmful side effect. Thalidomide’s side effect is, perhaps, the most remarkable and unfortunate known case in the literature. It is a drug that was used by pregnant women in the 1960’s in the United Kingdom in order to combat morning sickness. However, the drug caused several birth defects [1]. This drug was sold in an equimolar mixture of enantiomer in which only one of them combats the symptom (eutomer), and the other caused side effects (deutomer) [2].
Large pharmaceutical companies aim to synthesize drugs that are quickly able to combat symptoms or illnesses with the least number of side effects by removing the deutomer. For this reason, enantioresolution is a crucial issue in the pharmaceutical industry. Distillation, decantation, and filtration are common methods of separation for compounds based on boiling point, density, and size, respectively. The resolution of compounds with different physicochemical properties might be simpler in comparison to enantioresolution. The separation of pairs of enantiomers, though, is not straightforward, and conventional methods are nearly ineffective. Crystallization, membrane, or chromatography are examples of processes used to separate enantiomers in a chiral environment, molecules that tend to bind to an enantiomer instead of the other [3].
Optical activity is the phenomenon of shifting in the direction of the light plane when it passes through a compound. Light is an electromagnetic radiation that, when interacting with electrons in a molecule, slightly diverts its direction. Some molecules, though, do not present optical activity, i.e., for any light shift, either to the left or right, there is a molecule that shifts it in the opposite direction, nulling the optical activity. On the other hand, when light passes through pure there is a shift of the light; when light shifts to left, the enantiomer is called levorotatory (receives the prefix l or ( ) ) and to the right it is called dextrorotatory (receives the prefix d or ( + ) ).
A polarimeter is the instrument responsible for reading the light rotation in degrees of a certain enantiomer concentration. This rotation must be converted into a specific rotation to take in account the polarimeter length and sample concentration, as given by:
[ α ] λ T = α l p o l · c
where [ α ] is the specific rotation, superscript T designates the temperature (usually 20 °C), superscript λ the light wavelength (usually at 589.6 nm of Na D-lines), l p o l is the polarimeter length in dm, c is the enantiomer concentration in g·cm−3, and α is the observed rotation in degrees in the polarimeter. It means that an l enantiomer has a negative specific rotation, a d enantiomer a positive one, and a racemate has a [ α ] λ T of zero.
A pair of enantiomers does not overlap by either transposition or rotation. The non-overlapping of molecules is known in chemistry as chirality and these compounds are therefore called chirals (from the Greek word χέριa [quéria], meaning hands). In order to be considered chiral, the molecule must have at least one tetrahedral carbon bound to four different groups, as shown in Figure 1. This carbon is known as chiral or asymmetric carbon [4]. There is also the possibility of chirality involving other atoms such as silicon, nitrogen, sulfur and phosphorus, coordination complexes, allenes and atropisomers, as shown in Figure 2; however, these are less common [5].
Chirality in a molecule’s structural formula is represented by solid and hashed wedges ( Encyclopedia 02 00011 i065 and Encyclopedia 02 00011 i066) representing bonds that are projected out towards and retreating from the viewer, respectively. At least two equal bonds make up the atom achiral; therefore it does not form optical isomers. The number of chiral carbons determines the number of enantiomers of a compound, as given by:
N E N = 2 n
where N E N and n are the number of enantiomers and chiral carbons, respectively, as exemplified in Figure 3. However, this correlation is not always true; molecules with at least two chiral carbons that present a plane of symmetry are not chiral molecules. One of its carbons shifts the light plan to left, whereas the other nulls its effect, an internal compensation. An example is illustrated in Figure 4, although the tartaric acid has two chiral carbons, it has only two enantiomers and one meso isomer, a diastereomer and a non-optically active stereoisomer (see Table 1).

2. Nomenclature

A pair of enantiomers is formed by two different compounds that must have two different names. The Chemical Abstracts Service Registry Numbers (CAS RN or CAS Number) is a registration system of different compounds. Chemical Abstract Service has registered over 190 million compounds since the XIX century and thousands are registered every day [6]. Its database includes inorganic and organic compounds; metals and alloys; polymers; isotopes; protein and nucleic acids; coordination compounds and organometallics; minerals; and even mixtures. In fact, not only the pure enantiomers, but racemates, a mixture of compounds, have their own CAS Number. For instance, limonene racemate’s, ( + )-limonene’s and ( )-limonene’s CAS Numbers are, respectively, 138-86-3, 5989-27-5 and 5989-54-8. The CAS Number does not have a significance itself, as it is just based on the order of registration. On the other hand, prefixes ( d   or   + ) and ( l   o r ), mentioned in the previous section, designates whether the compound shifts the light to the right or left, and does not distinguish enantiomers when there are four or more enantiomers, as shown in Figure 3b; therefore, this is not a form of naming either.
Pairs of enantiomers are named following a sequence of rules created by the chemists R.S. Cahn, C. Ingold, and V. Prelog. This is known as Cahn-Ingold-Prelog rules or R/S configuration, where R and S stand for rectus and sinister (right and left in Latin). The rules for enantiomer nomenclature are detailed in Table 2.
First, it is necessary to rank the atoms directly bounded to the chiral carbon from largest to smallest atomic number, even in the case of isotopes. When the atoms have the same atomic number, one must follow down to the next substituent until there is a divergence point. Then, the least atomic number substituent is posed at the rear from a 3D perspective (a molecular modeling kit might help), as shown in Table 2. Finally, one draws circle arrows from the first to the third position. If the arrow goes counterclockwise, the enantiomer is S, otherwise is R. This procedure must be repeated when the molecule has other chiral carbons.
It is worth noticing that the prefixes (R) and (S) have no correlation with the shift of light plan, as an enantiomer might receive a prefix (R) and shifts light to left and vice versa. For instance: (R)-limonene has a specific rotation of +12° whereas (R)-butanol presents −13.5°. Another important issue that should be noted that there is another form of nomenclature exclusive for amino acids and monosaccharides, L and D , which have nothing to do with prefixes (R)/(S) or ( d )/( l ). L -amino acids and D -monosaccharides are by far the most common types that are present in all forms of life, with D -amino acids and R -monosaccharides nearly inexistent. They receive the prefix L when the amine or hydroxy group on the farthest carbon chiral from the carbonyl group is on the left side in a fisher projection, and prefix D when is on the right, as shown in Figure 5.

3. Enantiomers and the Human Body

Several natural compounds have two or more enantiomeric forms due to the complexity of their molecules that hold one or more chiral carbons. The human body, in turn, is essentially asymmetric; several organic molecules present chiral carbon, such as deoxyribonucleic and ribonucleic acid and their monosaccharides deoxyribose and ribose, respectively, and monosaccharides of the natural polymer DNA and RNA, shown in Figure 6, are present in all human cells [7]. “Chirality represents an intrinsic property of the so-called ‘structural blocks of life’, such as amino acids and monosaccharides and, consequently, peptides, proteins and polysaccharides” [8].
In their composition, many drugs have enantiomeric compounds that, in an asymmetric environment such as the human body, act completely different. There are several cases where one of the enantiomer is the active principle, while the other might cause side effects, prove to be toxic, ineffective or antagonist in the treatment [2,9,10,11,12]. Many drugs must be sold in its pure enantiomer form in order to be effective and/or harmless, for this reason, enantioresolution is one of the most important issues of the pharmaceutical industry. There are many known cases in the literature of different behaviors of enantiomers in pharmaceutical, agronomic and food applications, as shown in Table 3 [4,13,14].
The reason for the distinct actuation of virtually identical compounds is found in the biological receptors in human cells. These are macromolecules present in cell membranes that mediate the effects of chemical messages, hormones, and drug actions in the body [13]. They are responsible for selecting which substance from extracellular fluid may or may not enter the cytoplasm [15]. Figure 7 illustrates this interaction between biological receptors and chiral molecules. In Figure 7a, the enantiomer interacts perfectly with the receptor, whereas in Figure 7b the connection is compromised, which might cause a side effect if any effect at all. The pharmacological properties, pharmacokinetic of adsorption, distribution, biotransformation and excretion, and drug toxicology of the enantiomeric drug must be well known and identified [8]. It is important to define the safe degree of purity for an enantiomeric drug before commercialization. Pharmaceutical companies are also interested in selling safe and efficient medicaments, especially in a highly competitive market of medicaments.

4. Enantiomeric Drug Market

Nearly 60% of prescribed drugs have an enantiomeric pair [16,17]. However, for a long time, it was a common practice to commercialize chiral drugs in their racemic mixture. Regulatory authorities have put pressure on pharmaceutical companies to sell enantiomeric medicaments in their pure form [2,4]. Ethambutol was the first drug to be commercialized as a single enantiomer in 1961 [18]. Since then, the number of enantiomers sold in their pure form has increased year after year. In 1994, only 20% of the top selling drugs were enantiomers in their pure form [4]. In 2005 this number was already 37% [19] and currently it is over 50% [20]. This increase is justified to regulators that increasingly require clinical control and commercialization of pure enantiomers [8]. An example is the American Federal Agency FDA (Food and Drug Administration), which requires toxicological data of each enantiomer individually [4,21,22].
In economic terms, there was a worldwide growth of USD 30 billion to USD 100 billion from 1992 to 2000 with about 24 companies specializing in enantiomer separation [8,23]. In 2002, this value was already USD 159 billion and in 2005 USD 225 billion [8,19,24]. Table 4 shows the top pharmaceutical companies worldwide in 2017 with regard to chiral active ingredients, according to Pharmacompass (2019), which taken together account for almost USD 58.4 billion.
In such a market, pharmaceutical industries increasingly require refined compounds, which act quickly, in order to stand out in a competitive market. Due to the difference in the performance of different enantiomers, the complexity of some compounds, and the economic-market importance, it is crucial to efficiently and economically obtain each compound separately, either by synthesis or separation.

5. Enantioresolution

Currently, there are two pathways to obtain chiral compounds in their pure form, namely, synthesis (chiral route) or separation (racemic route), as shown in Figure 8. In the first case, each enantiomer is produced separately by using a chiral catalyst that induces selectivity for a given enantiomer, as shown in Figure 8a [4,8]. If the other enantiomer is also desired, it is necessary to develop a second synthesis with a different chiral catalyst. At first glance, the chiral route seems to be more advantageous than the racemic route (Figure 8b); they synthesize 100% of the intended enantiomer and there is no need for further separation and energetic expenses. On the other hand, racemic routes achieve up to 50% of the desired optically active isomer and racemic routes show inherently poor “atom economy”, i.e., part of the raw material is wasted, and the methods are not “elegant” [25,26,27,28]. However, chiral routes usually have low overall yields and only a few of them are applied for industrial purpose, especially at the early stages of new drugs development that requires pure enantiomers for pharmacological tests [2]. Keith, Larrow and Jacobsen (2001) list the following conditions that must be met in order to make the chiral route feasible: cheap racemate; poor CSP (Chiral Stationary Phase) enantioselectivity; highly selective catalyst for one enantiomer; inexpensive or efficiently recyclable catalyst and economical and safe reaction. Usually, these conditions are difficult to achieve, in a way that the production of the racemic mixture for further separation (Figure 8b) is normally favored. Furthermore, economic interest is the driving force that boosts the development of new enantioresolution technologies [2]. The cost of separation depends on the desired degree of purity and this affects the separation route [29].
Several methods for enantioresolution have been carried out in chiral environments using CSP. Chiral stationary phases are more effective at binding to one of the enantiomers, according to Dalgliesh’s three-point rule [30], and analogous to biological receptor in Figure 7. This rule states that a chiral environment should have three interaction points with the retained compound either by H-bonding, electrostatic interaction, dipole stacking, inclusion complexion or steric bulk hindrance. When the enantiomer does not interact perfectly with the CSP, the connection weakens and the enantiomer is less retained in the chiral environment, promoting enantioseparation [31,32,33].
Enantioresolution methods have existed since the early works of Louis Pasteur, who was able to separate tartaric acid manually by crystallization [4]. Currently, there are several methods for enantioresolution, such as chromatography, crystallization, and membrane. Figure 9 summarizes the main methods for enantiomer separation that might be Crystallization, Membrane, and Chromatography. The latter can be divided into Gas Chromatography (GC), Supercritical Fluid Chromatography (SFC), and Liquid Chromatography (LC). The latter can be further classified into High Pressure Liquid Chromatography (HPLC), True Moving Bed (TMB) and Simulated Moving Bed (SMB) chromatography. All these methods have advantages and drawbacks and choosing the best one among numerous technologies is not straightforward; therefore, there is no single resolution method suitable for all racemate [8,29,34,35]. The main mechanisms for enantioresolution are going to be detailed in the following sections.

5.1. Crystallization

Crystallization is a stochastic phenomenon of molecular dynamics based on many variables such as temperature, pressure, glass-forming ability of liquids, among other not well understood [36]. Table 5 presents some examples of enantiomers resolved by crystallization methods in the literature. Crystals might be formed by either conglomerates or racemic crystals [37]. Conglomerates are mechanical mixtures of crystals macroscopically distinguished from the pure enantiomers. The first enantiomeric resolution was carried out by Louis Pasteur in 1848, who manually separated conglomerate crystals of sodium ammonium tartrate [37,38]. In racemic crystals, the two enantiomers crystallize together forming a one-phase crystal containing the same amounts of each enantiomer, as shown in Figure 10.
Conglomerate crystal separations are generally straightforward; during the process of crystallization, enantiomers separate spontaneously into two stable crystal-phases, this mechanism is called preferential (or direct) crystallization and it is a non-expensive and efficient method, even on large scales [39,40]. Unfortunately, crystals from conglomerates are rather rare, comprising only 5–10% of all racemate crystals [38,40,41,42].
Table 5. Experimental studies of enantioresolutions applying crystallization.
Table 5. Experimental studies of enantioresolutions applying crystallization.
EnantiomersRef.
Ketoprofen[36]
5-ethyl-5-methylhydantoin[39]
Threonine[42]
Aspartic acid and glutamic acid[43]
Propranolol[44]
N-methylamphetamine[45]
Threonine[46]
Mandelic acid[47]
Chiral microspheres based on poly(N-vinyl a-L-phenylalanine)[48]
Benzo-(c)phenanthrene, 3,4-dehydroproline anhydride, and 2,6-dimethylglycoluril[49]
2-(2-oxopyrrolidin-1-yl)butanamide[50]
Allenyl-bis-phosphine oxides[51]
Leucine[52]
Ibuprofen lysine[53]
Racemic crystals, in turn, require suitable resolving agents, which are optically active compounds that aid in their non-spontaneous separation. Resolving agents convert the enantiomeric pair into two different diastereomers (geometric isomers, see Table 1) with different Physico-chemical properties, as they are different compounds, such as solubility, thus, facilitating their separation [44,54]. This mechanism is the so-called Classical Resolution [38]. The main idea is to break the symmetry between enantiomers by adding a chiral agent in an achiral solvent, possibly forming non-covalent and covalent diastereomers. The former involves salt formation by adding acidic or basic substrates and is the most common resolution method and the latter is applied for molecules unable to form salt [40,54,55].

5.2. Membrane

Membranes for enantiomer separation have advantages over other methods because they tend to be low energy consuming, can work in continuous operation, even in large scales, can be scaled up and down, have higher throughput than other methods and are eco-friendly [31,40,56,57,58]. The membranes provide good a transport rate, high selectivity, stability in wide range of pH among different solvents and might be produced from different polymeric materials [59]. Enantioselective membranes work as barriers with chiral recognition sites that selectively transport one of the enantiomers based on affinity between the enantiomer and chiral selectors. Table 6 presents some examples of enantiomers resolved by membrane methods in the literature.
Enantioselective membranes might be either liquid or solid [2,56]. Liquid membranes are formed by organic liquid with Chiral Selectors (CS) dissolved or suspended in it, such as cyclodextrins, crown ethers, chiral copper complexes, DNA, polypeptides, and enzymes. Solid membranes for enantiomer separation use chiral polymers as enantioselective compounds that interact to a specific enantiomer, as shown in Figure 11 [62]. They are either matrices of molecularly imprinted chiral polymers or membranes with the chiral polymeric selector immobilized (by impregnation, esterification, or grafting) on porous membranes [33,40,56,68,71].

5.3. Chromatography

Chromatographic processes are largely applied in several fields such as chemistry, pharmacy, and bioprocesses either for purification or recovery of different compounds [83]. Its separation is based on a physico-chemical phenomenon whereby a compound in a mobile phase known as the eluent, liquid or gaseous phase, is adsorbed onto the surface of a solid phase, usually a porous adsorbent. Chromatography displays some advantages over crystallization and membrane separations once it may be applied for a mixture of more than two chiral compounds and it can run with samples of small amount for analytical purposes [84]. Its separation is based on the chemical affinity of each compound to a stationary phase. In a chromatography equipment there are a solid stationary phase, generally porous, and a liquid or gaseous (solvent) mobile phase, which contains the mixture to be separated. Chromatographic methods have been the most effective for obtaining enantiomers with very high purity [4,8].
There are different chromatographic techniques for chiral resolution that are able to separate virtually all racemic compounds, that includes GC, SFC, and LC, as shown in Figure 9. Regardless of the chromatographic method applied, a Chiral Stationary Phase is applied, according to Dalgliesh’s three-point rule [30]. Figure 12 depicts adsorption of Enantiomer 1 (E1) onto chiral environment, following Dalgliesh’s three-point rules, whereas Enantiomer 2 (E2) has a compromised interaction to the surface, not fixing on it. CSPs are adsorbents usually based on cellulose such as the commercial products Chiralcel OD, CHIRALCEL OZ, CHIRALCEL OJ and CHIRALPAK AD, whereas common eluents are hexane, ethanol, and methanol. The enantiomers commonly resolved are pharmaceutical drugs such as Praziquantel and Guaifenesin.
All chromatographic techniques addresed to enatioresolution have their advantages and disadvantages. The next sections detail the differences among these chromatographic techniques, their functionalities and particularities.

5.3.1. GC—Gas Chromatography

Gas chromatography can be used by indirect or direct mechanisms [85]. In the former, enantiomers are converted into diastereomers by a resolving agent, analogous to a crystallization in racemic crystals, then they are separated in an achiral filling CG. The latter uses CSP that separates one of the enantiomers preferentially in a straightforward way by means of chemical affinity based on the three-point rule dismissing a pretreatment [86]. For the direct mechanism, GC requires the use of volatile and thermally stable compounds and the choice for a CSP is of paramount importance in order to resolve racemic mixtures [87].
The first GC with a Chiral Stationary Phase was applied to resolve 2-nalkanols [85]. Since this achievement, GC has been extensively used in academia and industry due to its advantages of being high efficient, simple, and sensitive in comparison to other methods [85,86,88,89]. On the other hand, GC has the drawbacks of being very difficult to scale up and their CSPs usually racemize, decompose and bleed at high temperatures, thereby diminishing its separation factor [87,90]. Table 7 presents some examples of enantiomers resolved by gas chromatography methods.

5.3.2. SFC—Supercritical Fluid Chromatography

Supercritical fluids display transient properties between liquid and gas phases; therefore SFC works as an intermediate between GC and LC [117]. At the supercritical condition, the substances are above its critical temperature and pressure [118,119]. Supercritical fluid was firstly used as an eluent for chromatographic separation in 1962 by Klesper, Iiber and Clark using chlorofluoromethanes at 140 bar and 150–170 °C [120,121]. Supercritical fluids have advantages over liquid and gas states and they are a better solvent due to their higher density in comparison to gas states as they are faster (shorter run times), require lower pressure across column and require lower volume due to its lower viscosity and higher diffusivity over liquid states [118,119,120,122,123,124,125,126,127]. The most commonly applied supercritical fluid is carbon dioxide; however, the mentioned features are not exclusive to fluids over the critical point, and equal properties are seen in subcritical regions, since there are no abrupt changes of properties in the transition to a supercritical phase. This means that the fluid must not be at a supercritical condition to perform an SFC [118]. Table 8 presents some examples of enantiomers resolved by SFC methods reported in the literature.
Additionally, CO2 is the most used eluent in SFC due to its relatively low critical point (304.12 K and 73 atm), and it can be purified and reused after analysis; moreover it is inert, non-toxic, non-flammable cheap and relatively safe gas, therefore being considered a green solvent [118,119]. Due to its low critical temperature point, operating CO2 in SFC reduces the likelihood of CSP racemization, improving enantioselectivity [124,153].

5.3.3. LC—Liquid Chromatography

Liquid chromatography has been growing in importance in the last decade due to the variety of enantioseparation modalities [5,154,155]. In LC, the eluent and compounds cross the fixed bed column (adsorber) by means of gravity and this can prove to be lengthy in some procedures. The time-consumption of LC can be overcome by means of a pump, low-dead-volume injectors, and detectors. An enhancement of LC is the so-called High-Performance Liquid Chromatography (HPLC—also known as High-Pressure Liquid Chromatography). HPLC is suitable for a wide range of applications, such as pharmaceuticals and food analysis. HPLC has the advantage of running a fast analysis and high resolution; however, in HPLC (and LC) the adsorber must be constantly regenerated, once it will be contaminated by the more retained enantiomer. Table 9 presents some examples of enantiomers resolved by HPLC methods.
True Moving Bed (TMB) is an attempt to develop a continuous liquid chromatography based on the counter-current movement of eluent and solid. Its separation is maximized by a constant flow of solid and liquid phases counter-currently. In this chromatography, the CSP adsorber retaines one of the compounds due to its enantioselectivity nature. This leads to different velocities of displacement of enantiomers along the column. The more retained enantiomer takes longer to reach the end of the column than the less retained compound and, for better separation, the column must be long enough [83]. In TMB chromatography, the adsorber is constantly being regenerated by the eluent. In TMB chromatography, there is constant liquid and solid recirculation, as shown in Figure 13. The liquid stream leaves the top of the column at Section IV and is recycled to Section I, while the solid stream (adsorbent) moves in the opposite direction, being recycled from Section I to Section IV. The eluent and racemate feed the system (respectively, streams E and F in Figure 13). As shown in Figure 13, the more retained compound leaves the system in the Extract Stream (X), whereas the less retained compound is removed in the Raffinate Stream (R). The system has four sections with different functions [83], as explained in Table 10. Due to the counter-current flows, TMB should reach higher purity, even if the adsorbent (solid phase) presents low selectivity, in contrast to conventional chromatography where high selectivity is crucial [83]. However, from an engineering perspective, solid movement is difficult to attain and can cause mechanical erosion in the adsorbent phase, equipment abrasion, and difficulties in maintaining plug flow for the solid [4,83]. The TMB system is a theoretical concept and to solve such problems, an SMB chromatographic unit was developed.
A Simulated Moving Bed is a powerful technology for the preparative and analytical scale in laboratory or in industry [83]. SMB chromatography was first presented by UOP (Universal Oil Products) through a United States Patent [4,179,180,181], known as the Sorbex Process, which was designed for oil refining purposes [4]. The technology soon found other industrial applications, such as in biotechnology, pharmaceutical and fine chemistry, as it is a separation system with advantages over batch chromatographic systems and traditional processes (PAIS, 1999). Negawa and Shoji (1992) were the first authors to carry out an enantioresolution in an SMB chromatography, they resolved 1-phenylethanol on CHIRALCEL OD as CSP. Since then, there have been several enantioresolutions reported in the literature, as summarized in Table 11.
SMB simulates TMB counter-current flows and overcomes its intrinsic solid movement problems by keeping the solid phase fixed and switching the input and output streams cyclically and periodically (time switch) in a multi-column system, as shown in Figure 14, by simulating a bed movement [208]. SMB has four sections for which the function is analogous to TMB, as shown in Figure 13. The SMB system is fed with an eluent (stream E in Figure 14) and racemate to be separated (stream F in Figure 14). Two outlet streams remove the more and the less retained compounds, respectively, in the extract and raffinate streams (streams X and R, respectively in Figure 14). At the time instant t1, the inlet and outlet streams are set as shown in Figure 14. At time t2, they move clockwise from their current positions to the next ones. Next, they proceed to t3 and so on until they return to their original positions, thus finishing the cycle. The separation is promoted by adsorption of the enantiomer with greater chemical affinity onto the porous solid phase, as zoomed in Figure 14.
Optimal SMB operation depends upon several factors, such as reasonable operating conditions of its streams, switching time, eluent, and the chiral stationary phase. Engineering practices have shown that the scenario of SMB operating conditions is rather limited; a poor set of the pump flowrates may lead to contamination of the outlet streams, thus compromising the separation. The Simulated Moving Bed has been gaining prominence due to its advantageous features such as cleanliness, small size, security, and fast procedure when compared to other systems. Moreover, the method has presented great performance in the separation of enantiomeric drugs such as praziquantel [209], which is a drug frequently used to combat schistosomiasis.

6. Conclusions

Due to the importance of enantiomer in several fields, especially for pharmaceutical companies that strive to diminish drug side effects, this article approaches different features of enantiomers. The article defines what an enantiomer is, presents related nomenclature, optical activity, enantiomers actuation in human body, enantiomers in biomolecules, market and pharmaceutical demands, and enantioresolution methods such as membranes, crystallization, and chromatography, especially TMB and SMB. It also reports on the features of the most applied enantioresolution methods, main enantiomers, eluents, and chiral stationary phases, with an emphasis on the cyclic adsorption processes. Through the comprehension of the aspects related to enantiomers, the authors of this articles hope to raise the interest of incoming students of chemical engineering to enantioresolution, control, and optimization field.

Author Contributions

Conceptualization, I.B.R.N., K.V.P. and R.S.; writing—original draft preparation, I.B.R.N., K.V.P. and R.S.; writing—review and editing, I.B.R.N., K.V.P. and R.S.; supervision, I.B.R.N. and K.V.P. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financed in part by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior-Brasil (CAPES)-Finance Code 001. This work was also financially supported by: Base Funding-UIDB/50020/2020 of the Associate Laboratory LSRE-LCM-funded by national funds through FCT/MCTES (PIDDAC); FCT—Fundação para a Ciência e Tecnologia under CEEC Institutional program.

Conflicts of Interest

The authors declare no conflict of interest.

Entry Link on the Encyclopedia Platform

References

  1. Johnson, M. Integrating Health Information: A Case Study of a Health Information Service for Thalidomide Survivors. Inform. Health Soc. Care 2007, 32, 27–33. [Google Scholar] [CrossRef]
  2. Maier, N.; Pilar, F.; Lindner, W. Separation of Enantiomers: Needs, Challenges, Perspectives. J. Chromatogr. A 2001, 906, 3–33. [Google Scholar] [CrossRef]
  3. Molbase. Available online: http://www.molbase.com/ (accessed on 28 January 2019).
  4. Pais, L.M.S. Chiral Separation by Simulated Moving Bed Chromatography; Universidade do Porto: Campo Alegre, Porto, 1999. [Google Scholar]
  5. Lough, W.J. Chiral Liquid Chromatography; Springer: New York, NY, USA, 1989. [Google Scholar]
  6. CAS. Available online: https://www.cas.org/ (accessed on 24 November 2021).
  7. Rang, H.; Ritter, J.M.; Flower, R.J.; Henderson, G. Farmacologia; Elsevier: Philadelphia, PA, USA, 2020. [Google Scholar]
  8. José, I.; De Veredas, V.; Antônio, M.; Costapinto, C.; José, M.; Carpes, S.; Duarte, R. Cromatografia em leito móvel simulado na produção de substâncias enantiomericamente puras ou enriquecidas em larga escala. Quim. Nova 2006, 29, 1027–1037. [Google Scholar]
  9. Aboul-Enein, H.Y.; Wainer, I.W. The Impact of Stereochemistry on Drug Development and Use; Winefordner, J.D., Ed.; Wiley-Interscience: Montreal, QC, Canada, 1997. [Google Scholar]
  10. Caldwell, J. Stereochemical Determinants of the Nature and Consequences. J. Chromatogr. A 1995, 694, 39–48. [Google Scholar] [CrossRef]
  11. Caldwell, J. Importance of Stereospecific Bioanalytical Monitoring in Drug Development. J. Chromatogr. A 1996, 719, 3–13. [Google Scholar] [CrossRef]
  12. Ariëns, E.J. Stereochemistry: A Source of Problems in Medicinal Chemistry. Med. Res. Rev. 1986, 6, 451–466. [Google Scholar] [CrossRef] [PubMed]
  13. Gal, J. The Discovery of Stereoselectivity at Biological Receptors: Arnaldo Piutti and the Taste of the Asparagine Enantiomers—History and Analysis on the 125th Anniversary. Eikasmos 2012, 24, 19. [Google Scholar] [CrossRef]
  14. Mannschreck, A.; Kiesswetter, R.; von Angerer, E. Unequal Activities of Enantiomers via Biological Receptors: Examples of Chiral Drug, Pesticide, and Fragrance Molecules. J. Chem. Educ. 2007, 84, 2012. [Google Scholar] [CrossRef]
  15. Kurt, J. Histology and Cell Biology, 2nd ed.; Elsevier, Ed.; Harwal Medical Publications: Baltimore, MD, USA, 1991. [Google Scholar]
  16. Sanz-Medel, A.; Blanco-González, E. Chiral Trace-Element Speciation in Biological Samples: Present Importance and Application to Speciation for Seleno-Amino Acids. Trends Anal. Chem. 2002, 21, 709–716. [Google Scholar] [CrossRef]
  17. Ali, I.; Alam, S.D.; Al-Othman, Z.A.; Farooqi, J.A. Recent Advances in SPE-Chiral-HPLC Methods for Enantiomeric Separation of Chiral Drugs in Biological Samples. J. Chromatogr. Sci. 2013, 51, 645–654. [Google Scholar] [CrossRef]
  18. Bhupinder Singh Sekhon. Enantioseparation of Chiral Drugs—An Overview. Int. J. PharmTech Res. 2010, 2, 1584–1594. [Google Scholar]
  19. Erb, S. Single-Enantiomer Drugs Poised for Further Market Growth. Pharm. Technol. 2006, 30, s14–s18. [Google Scholar]
  20. Technology Catalysts. Available online: https://technology-catalysts.com/industry-expertise/fine/ (accessed on 29 January 2019).
  21. De Camp, W.H. The FDA Perspective on the Development of Stereoisomers. Chirality 1989, 1, 2–6. [Google Scholar] [CrossRef]
  22. FDA. FDA’S Policy Statement for the Development of New Stereoisomeric Drugs; FDA: Silver Spring, MD, USA, 1992; Volume 4.
  23. Rekoske, J.E. Chiral Separations I Figure I. Distrib. Drugs Dev. Worldw. 2001, 47, 2–5. [Google Scholar]
  24. Rouhi, A.M. Fine Chemicals Companies Are Jockeying for Position to Deliver the Increasingly Complicated Chiral Small Molecules of the Future. Chem. Eng. News 2003, 81, 45–61. [Google Scholar] [CrossRef]
  25. Pharmacompass. Available online: https://www.pharmacompass.com/radio-compass-blog/top-drugs-by-sales-in-2017-who-sold-the-blockbuster-drugs (accessed on 19 December 2021).
  26. Hajos, Z.G.; Parrish, D.R. Asymmetric Synthesis of Bicyclic Intermediates of Natural Product Chemistry. J. Org. Chem. 1974, 39, 1615–1621. [Google Scholar] [CrossRef]
  27. Keith, J.M.; Larrow, J.F.; Jacobsen, E.N. Practical Considerations in Kinetic Resolution Reactions. Adv. Synth. Catal. 2001, 343, 5–26. [Google Scholar] [CrossRef]
  28. Trost, B.M. Atom Economy—A Challenge for Organic Synthesis: Homogeneous Catalysis Leads the Way. Angew. Chem. Int. Ed. Engl. 1995, 34, 259–281. [Google Scholar] [CrossRef]
  29. Carvalho, P.O.; Cass, Q.B.; Calafatti, S.A.; Contesini, F.J.; Bizaco, R. Review-Alternatives for the Separation of Drug Enantiomers: Ibuprofen as a Model Compound. Braz. J. Chem. Eng. 2006, 23, 291–300. [Google Scholar] [CrossRef]
  30. Dalgliesh, C.E. The Optical Resolution of Aromatic Amino-Acids on Paper Chromatograms. J. Chem. Soc. 1952, 3, 756. [Google Scholar] [CrossRef]
  31. Afonso, C.A.M.; Crespo, J.G. Recent Advances in Chiral Resolution through Membrane-Based Approaches. Angew. Chem. Int. Ed. Engl. 2004, 43, 5293–5295. [Google Scholar] [CrossRef] [PubMed]
  32. Pirkle, W.H.; Pochapsky, T.C. Considerations of Chiral Recognition Relevant to the Liquid Chromatographic Separation of Enantiomers. Chem. Rev. 1989, 89, 347–362. [Google Scholar] [CrossRef]
  33. Singh, K.; Ingole, P.G.; Bajaj, H.C.; Gupta, H. Preparation, Characterization and Application of β-Cyclodextrin-Glutaraldehyde Crosslinked Membrane for the Enantiomeric Separation of Amino Acids. Desalination 2012, 298, 13–21. [Google Scholar] [CrossRef]
  34. Francotte, E.R. Enantioselective Chromatography as a Powerful Alternative for the Preparation of Drug Enantiomers. J. Chromatogr. A 2001, 906, 379–397. [Google Scholar] [CrossRef]
  35. Ward, T.J.; Ward, K.D. Chiral Separations: A Review of Current Topics and Trends. Anal. Chem. 2012, 84, 626–635. [Google Scholar] [CrossRef] [PubMed]
  36. Adrjanowicz, K.; Kaminski, K.; Paluch, M.; Niss, K. Crystallization Behavior and Relaxation Dynamics of Supercooled S-Ketoprofen and the Racemic Mixture along an Isochrone. Cryst. Growth Des. 2015, 15, 3257–3263. [Google Scholar] [CrossRef]
  37. Collet, A.; Brienne, M.J.; Jacques, J. Optical Resolution by Direct Crystallization of Enantiomer Mixtures. Chem. Rev. 1980, 80, 215–230. [Google Scholar] [CrossRef]
  38. Lorenz, H.; Seidel-Morgenstern, A. Processes to Separate Enantiomers. Angew. Chem. Int. Ed. 2014, 53, 1218–1250. [Google Scholar] [CrossRef]
  39. Gervais, C.; Beilles, S.; Cardinaël, P.; Petit, S.; Coquerel, G. Oscillating Crystallization in Solution between (+)- and (-)-5-Ethyl-5-Methylhydantoin under the Influence of Stirring. J. Phys. Chem. B 2002, 106, 646–652. [Google Scholar] [CrossRef]
  40. Xie, R.; Chu, L.Y.; Deng, J.G. Membranes and Membrane Processes for Chiral Resolution. Chem. Soc. Rev. 2008, 37, 1243–1263. [Google Scholar] [CrossRef]
  41. Collet, A. Resolution of Racemates: Did You Say “Classical”? Angew. Chem. Int. Ed. 1998, 37, 3239–3241. [Google Scholar] [CrossRef]
  42. Rodrigo, A.A.; Lorenz, H.; Seidel-Morgenstern, A. Online Monitoring of Preferential Crystallization of Enantiomers. Chirality 2004, 16, 499–508. [Google Scholar] [CrossRef] [PubMed]
  43. Viedma, C. Enantiomeric Crystallization from DL-Aspartic and DL-Glutamic Acids. Implic. Biomol. Chirality Orig. Life 2001, 31, 501–509. [Google Scholar]
  44. Ge, S.H.; Li, X.G.; Hsing, I.M. Water Management in PEMFCs Using Absorbent Wicks. J. Electrochem. Soc. 2004, 151, B523. [Google Scholar] [CrossRef]
  45. Kmecz, I.; Simándi, B.; Székely, E.; Fogassy, E. Resolution of N-Methylamphetamine Enantiomers with Tartaric Acid Derivatives by Supercritical Fluid Extraction. Tetrahedron Asymmetry 2004, 15, 1841–1845. [Google Scholar] [CrossRef]
  46. Elsner, M.P.; Menéndez, D.F.; Muslera, E.A.; Seidel-Morgenstern, A. Experimental Study and Simplified Mathematical Description of Preferential Crystallization. Chirality 2005, 17, 183–195. [Google Scholar] [CrossRef] [PubMed]
  47. Lorenz, H.D.P.; Seidel-Morgenstern, A. Application of Preferential Crystallization to Resolve Racemic Compounds in a Hybrid Process. Chirality 2006, 18, 828–840. [Google Scholar] [CrossRef] [PubMed]
  48. Medina, D.D.; Goldshtein, J.; Margel, S.; Mastai, Y. Enantioselective Crystallization on Chiral Polymeric Microspheres. Adv. Funct. Mater. 2007, 17, 944–950. [Google Scholar] [CrossRef]
  49. D’Oria, E.; Karamertzanis, P.G.; Price, S.L. Spontaneous Resolution of Enantiomers by Crystallization: Insights from Computed Crystal Energy Landscapes. Cryst. Growth Des. 2010, 10, 1749–1756. [Google Scholar] [CrossRef]
  50. Springuel, G.; Leyssens, T. Innovative Chiral Resolution Using Enantiospecific Co-Crystallization in Solution. Cryst. Growth Des. 2012, 12, 3374–3378. [Google Scholar] [CrossRef]
  51. Gangadhararao, G.; Tulichala, R.N.P.; Swamy, K.C.K. Spontaneous Resolution upon Crystallization of Allenyl-Bis-Phosphine Oxides. Chem. Commun. 2015, 51, 7168–7171. [Google Scholar] [CrossRef]
  52. Manoj, K.; Takahashi, H.; Morita, Y.; Gonnade, R.G.; Iwama, S.; Tsue, H.; Tamura, R. Preferential Enrichment of DL-Leucine Using Cocrystal Formation With Oxalic Acid Under Nonequilibrium Crystallization Conditions. Chirality 2015, 27, 405–410. [Google Scholar] [CrossRef] [PubMed]
  53. Simon, M.; Donnellan, P.; Glennon, B.; Jones, R.C. Resolution via Diastereomeric Salt Crystallization of Ibuprofen Lysine: Ternary Phase Diagram Studies. Chem. Eng. Technol. 2018, 41, 921–927. [Google Scholar] [CrossRef]
  54. Rougeot, C.; Hein, J.E. Application of Continuous Preferential Crystallization to Efficiently Access Enantiopure Chemicals. Org. Process. Res. Dev. 2015, 19, 1809–1819. [Google Scholar] [CrossRef]
  55. Fogassy, E.; Nógrádi, M.; Kozma, D.; Egri, G.; Pálovics, E.; Kiss, V. Optical Resolution Methods. Org. Biomol. Chem. 2006, 4, 3011–3030. [Google Scholar] [CrossRef] [PubMed]
  56. Van Der Ent, E.M.; Van Riet, K.; Keurentjes, J.T.F.; Van Der Padt, A. Design Criteria for Dense Permeation-Selective Membranes for Enantiomer Separations. J. Membr. Sci. 2001, 185, 207–221. [Google Scholar] [CrossRef]
  57. Higuchi, A.; Higuchi, Y.; Furuta, K.; Yoon, B.O.; Hara, M.; Maniwa, S.; Saitoh, M.; Sanui, K. Chiral Separation of Phenylalanine by Ultrafiltration through Immobilized DNA Membranes. J. Memb. Sci. 2003, 221, 207–218. [Google Scholar] [CrossRef]
  58. Matsuoka, Y.; Kanda, N.; Lee, Y.M.; Higuchi, A. Chiral Separation of Phenylalanine in Ultrafiltration through DNA-Immobilized Chitosan Membranes. J. Memb. Sci. 2006, 280, 116–123. [Google Scholar] [CrossRef]
  59. Fernandes, C.; Tiritan, M.E.; Pinto, M.M.M. Chiral Separation in Preparative Scale: A Brief Overview of Membranes as Tools for Enantiomeric Separation. Symmetry 2017, 9, 206. [Google Scholar] [CrossRef]
  60. Kaner, R.B. Gas, Liquid and Enantiomeric Separations Using Polyaniline. Synth. Met. 2001, 125, 65–71. [Google Scholar] [CrossRef]
  61. Thoelen, C.; De Bruyn, M.; Theunissen, E.; Kondo, Y.; Vankelecom, I.F.J.; Grobet, P.; Yoshikawa, M.; Jacobs, P.A. Membranes Based on Poly(γ-Methyl-L-Glutamate): Synthesis, Characterization and Use in Chiral Separations. J. Memb. Sci. 2001, 186, 153–163. [Google Scholar] [CrossRef]
  62. Hadik, P.; Szabó, L.P.; Nagy, E. D,L-Lactic Acid and D,L-Alanine Enantioseparation by Membrane Process. Desalination 2002, 148, 193–198. [Google Scholar] [CrossRef]
  63. Donato, L.; Figoli, A.; Drioli, E. Novel Composite Poly(4-Vinylpyridine)/Polypropylene Membranes with Recognition Properties for (S)-Naproxen. J. Pharm. Biomed. Anal. 2005, 37, 1003–1008. [Google Scholar] [CrossRef]
  64. Gumí, T.; Valiente, M.; Palet, C. Elucidation of SR-Propranolol Transport Rate and Enantioselectivity through Chiral Activated Membranes. J. Memb. Sci. 2005, 256, 150–157. [Google Scholar]
  65. Higuchi, A.; Hayashi, A.; Kanda, N.; Sanui, K.; Kitamura, H. Chiral Separation of Amino Acids in Ultrafiltration through DNA-Immobilized Cellulose Membranes. J. Mol. Struct. 2005, 739, 145–152. [Google Scholar] [CrossRef]
  66. Ghazali, N.F.; Ferreira, F.C.; White, A.J.P.; Livingston, A.G. Enantiomer Separation by Enantioselective Inclusion Complexation-Organic Solvent Nanofiltration. Tetrahedron Asymmetry 2006, 17, 1846–1852. [Google Scholar] [CrossRef]
  67. Maximini, A.; Chmiel, H.; Holdik, H.; Maier, N.W. Development of a Supported Liquid Membrane Process for Separating Enantiomers of N-Protected Amino Acid Derivatives. J. Memb. Sci. 2006, 276, 221–231. [Google Scholar] [CrossRef]
  68. Wang, H.D.; Chu, L.Y.; Song, H.; Yang, J.P.; Xie, R.; Yang, M. Preparation and Enantiomer Separation Characteristics of Chitosan/β-Cyclodextrin Composite Membranes. J. Memb. Sci. 2007, 29, 262–270. [Google Scholar] [CrossRef]
  69. Xiao, Y.; Chung, T.S. Functionalization of Cellulose Dialysis Membranes for Chiral Separation Using Beta-Cyclodextrin Immobilization. J. Memb. Sci. 2007, 290, 78–85. [Google Scholar] [CrossRef]
  70. Xiong, W.W.; Wang, W.F.; Zhao, L.; Song, Q.; Yuan, L.M. Chiral Separation of (R,S)-2-Phenyl-1-Propanol through Glutaraldehyde-Crosslinked Chitosan Membranes. J. Memb. Sci. 2009, 328, 268–272. [Google Scholar] [CrossRef]
  71. Singh, K.; Bajaj, H.C.; Ingole, P.; Bhattacharya, A. Comparative Study of Enantioseparation of Racemic Tryptophan by Ultrafiltration Using BSA-Immobilized and BSA-Interpenetrating Network Polysulfone Membranes. Sep. Sci. Technol. 2010, 45, 346–354. [Google Scholar] [CrossRef]
  72. Wang, Z.; Cai, C.; Lin, Y.; Bian, Y.; Guo, H.; Chen, X. Enantioselective Separation of Ketoconazole Enantiomers by Membrane Extraction. Sep. Purif. Technol. 2011, 79, 63–71. [Google Scholar] [CrossRef]
  73. Sunsandee, N.; Leepipatpiboon, N.; Ramakul, P.; Pancharoen, U. The Selective Separation of (S)-Amlodipine via a Hollow Fiber Supported Liquid Membrane: Modeling and Experimental Verification. Chem. Eng. J. 2012, 180, 299–308. [Google Scholar] [CrossRef]
  74. Naksang, C.; Sunsandee, N.; Thamphiphit, N.; Pancharoen, U.; Ramakul, P.; Leepipatpiboon, N. Synergistic Enantioseparation of Rac-Phenylalanine via Hollow Fiber Supported Liquid Membrane. Sep. Sci. Technol. 2013, 48, 867–876. [Google Scholar] [CrossRef]
  75. Alizadeh, T. Synthesis of a Nano-Sized Chiral Imprinted Polymer and Its Use as an (S)-Atenolol Carrier in the Bulk Liquid Membrane. J. Sep. Sci. 2014, 37, 1887–1895. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, F.; He, L.; Sun, W.; Cheng, Y.; Liu, J.; Ren, Z. Chiral Liquid Membrane for Enantioselective Separation of Racemic Ibuprofen by L-Tartaric Acid Derivatives. RSC Adv. 2015, 5, 41729–41735. [Google Scholar] [CrossRef]
  77. Meng, C.; Sheng, Y.; Chen, Q.; Tan, H.; Liu, H. Exceptional Chiral Separation of Amino Acid Modified Graphene Oxide Membranes with High-Flux. J. Memb. Sci. 2017, 526, 25–31. [Google Scholar] [CrossRef]
  78. Huang, X.Y.; Pei, D.; Liu, J.F.; Di, D.L. A Review on Chiral Separation by Counter-Current Chromatography: Development, Applications and Future Outlook. J. Chromatogr. A 2018, 1531, 1–12. [Google Scholar] [CrossRef]
  79. Keating, J.J.; Bhattacharya, S.; Belfort, G. Separation of D, L-Amino Acids Using Ligand Exchange Membranes. J. Memb. Sci. 2018, 555, 30–37. [Google Scholar] [CrossRef]
  80. Anand, D.; Dhoke, G.V.; Gehrmann, J.; Garakani, T.M.; Davari, M.D.; Bocola, M.; Zhu, L.; Schwaneberg, U. Chiral Separation of d/l-Arginine with Whole Cells through an Engineered FhuA Nanochannel. Chem. Commun. 2019, 55, 5431–5434. [Google Scholar] [CrossRef]
  81. D’Orazio, G.; Fanali, C.; Gentili, A.; Tagliaro, F.; Fanali, S. Nano-Liquid Chromatography for Enantiomers Separation of Baclofen by Using Vancomycin Silica Stationary Phase. J. Chromatogr. A 2019, 1605, 6–13. [Google Scholar] [CrossRef]
  82. Hadjmohammadi, M.R.; Hashemi, M. Chiral Separation of Methadone Using Solid Membrane Extraction Based on Chiral Selector, Solid Membrane: Sheep Skin Leather. J. Iran. Chem. Soc. 2019, 16, 1611–1616. [Google Scholar] [CrossRef]
  83. Rodrigues, A.E.; Pereira, C.; Minceva, M.; Pais, L.S.; Ribeiro, A.M.; Ribeiro, A.; Silva, M.; Graça, N.; Santos, J.C. Simulated Moving Bed Technology: Principles, Design and Process Applications; Joe Hayton: Amsterdam, The Netherlands, 2015. [Google Scholar]
  84. Audebert, R. Direct Resolution of Enantiomers in Column Liouid Chromatography. J. Liq. Chromatogr. 1979, 2, 1063–1095. [Google Scholar] [CrossRef]
  85. Gil-Av, E.; Feibush, B.; Charles-Sigler, R. Separation of Enantiomers by Gas Liquid Chrimatography with an Optically Active Stationary Phase. Br. J. Psychiatry 1966, 10, 1009–1015. [Google Scholar]
  86. He, L.; Beesley, T.E. Applications of Enantiomeric Gas Chromatography: A Review. J. Liq. Chromatogr. Relat. Technol. 2005, 28, 1075–1114. [Google Scholar] [CrossRef]
  87. Zhang, Y.; Wu, D.R.; Wang-Iverson, D.B.; Tymiak, A.A. Enantioselective Chromatography in Drug Discovery. Drug Discov. Today 2005, 10, 571–577. [Google Scholar] [CrossRef]
  88. Schurig, V. Enantiomer Analysis by Complexation Gas Chromatography. J. Chromatogr. A 1988, 441, 135–153. [Google Scholar] [CrossRef]
  89. Schurig, V.; Kreidler, D. Gas-Chromatographic Enantioseparation of Unfunctionalized Chiral Hydrocarbons: An Overview. Methods Mol. Biol. 2013, 970, 45–67. [Google Scholar]
  90. Lang, J.C.; Armstrong, D.W. Chiral Surfaces: The Many Faces of Chiral Recognition. Curr. Opin. Colloid Interface Sci. 2017, 32, 94–107. [Google Scholar] [CrossRef]
  91. Kim, K.H.; Lee, J.H.; Ko, M.Y.; Hong, S.P.; Youm, J.R. Chiral Separation of β-Blockers after Derivatization with (-)-AMethoxy-α-(Trifluoromethyl)Phenylacetyl Chloride by Gas Chromatography. Arch. Pharm. Res. 2001, 24, 402–406. [Google Scholar] [CrossRef]
  92. Shellie, R.; Marriott, P.J. Comprehensive Two-Dimensional Gas Chromatography with Fast Enantioseparation. Anal. Chem. 2002, 74, 5426–5430. [Google Scholar] [CrossRef] [PubMed]
  93. Ding, J.; Welton, T.; Armstrong, D.W. Chiral Ionic Liquids as Stationary Phases in Gas Chromatography. Anal. Chem. 2004, 76, 6819–6822. [Google Scholar] [CrossRef] [PubMed]
  94. Paik, M.J.; Nguyen, D.T.; Kim, K.R. Enantioseparation of Flurbiprofen and Ketoprofen in Patches and in Urine Excretions by Achiral Gas Chromatography. Arch. Pharm. Res. 2004, 27, 1295–1301. [Google Scholar] [CrossRef] [PubMed]
  95. Pätzold, R.; Schieber, A.; Brückner, H. Gas Chromatographic Quantification of Free D-Amino Acids in Higher Vertebrates. Biomed. Chromatogr. 2005, 19, 466–473. [Google Scholar] [CrossRef]
  96. Petrović, M.; Debeljak, Ž.; Blažević, N. Optimization of Gas Chromatographic Method for the Enantioseparation of Arylpropionic Non-Steroidal Anti-Inflammatory Drug Methyl Esters. J. Pharm. Biomed. Anal. 2005, 39, 531–534. [Google Scholar] [CrossRef] [PubMed]
  97. Shi, X.; Guo, H.; Wang, M. Enantioseparation of Chiral Epoxides Using Four New Cyclodextrin Derivatives as Chiral Stationary Phases of Capillary Gas Chromatography. Anal. Chim. Acta 2005, 553, 43–49. [Google Scholar] [CrossRef]
  98. Sicoli, G.; Jiang, Z.; Jicsinsky, L.; Schurig, V. Modified Linear Dextrins (“acyclodextrins”) as New Chiral Selectors for the Gas-Chromatographic Separation of Enantiomers. Angew. Chem. Int. Ed. 2005, 44, 4092–4095. [Google Scholar] [CrossRef]
  99. Takahisa, E.; Engel, K.H. 2,3-Di-O-Methoxymethyl-6-O-Tert-Butyldimethylsilyl-γ-Cyclodextrin: A New Class of Cyclodextrin Derivatives for Gas Chromatographic Separation of Enantiomers. J. Chromatogr. A 2005, 1063, 181–192. [Google Scholar] [CrossRef]
  100. Levkin, P.A.; Ruderisch, A.; Schurig, V. Combining the Enantioselectivity of a Cyclodextrin and a Diamide Selector in a Mixed Binary Gas-Chromatographic Chiral Stationary Phase. Chirality 2006, 18, 49–63. [Google Scholar] [CrossRef]
  101. Paik, M.J.; Nguyen, D.T.; Kim, K.R. N-Menthoxycarbonylation Combined with Trimethylsilylation for Enantioseparation of β-Blockers by Achiral Dual-Column Gas Chromatography. J. Chromatogr. A 2006, 1103, 177–181. [Google Scholar] [CrossRef]
  102. Zheng, R.C.; Zheng, Y.G.; Shen, Y.C. Enantioseparation and Determination of 2,2-Dimethylcyclopropanecarboxamide and Corresponding Acid in the Bioconversion Broth by Gas Chromatography. Biomed. Chromatogr. 2007, 21, 610–615. [Google Scholar] [CrossRef] [PubMed]
  103. Paik, M.J.; Lee, J.; Kim, K.R. N-Ethoxycarbonylation Combined with (S)-1-Phenylethylamidation for Enantioseparation of Amino Acids by Achiral Gas Chromatography and Gas Chromatography-Mass Spectrometry. J. Chromatogr. A 2008, 1214, 151–156. [Google Scholar] [CrossRef] [PubMed]
  104. Forró, E. New Gas Chromatographic Method for the Enantioseparation of β-Amino Acids by a Rapid Double Derivatization Technique. J. Chromatogr. A 2009, 1216, 1025–1029. [Google Scholar] [CrossRef] [PubMed]
  105. Grisales, J.O.; Lebed, P.J.; Keunchkarian, S.; González, F.R.; Castells, C.B. Permethylated β-Cyclodextrin in Liquid Poly(Oxyethylene) as a Stationary Phase for Capillary Gas Chromatography. J. Chromatogr. A 2009, 1216, 6844–6851. [Google Scholar] [CrossRef]
  106. Sicoli, G.; Kreidler, D.; Czesla, H.; Hopf, H.; Schurig, V. Gas Chromatographic Enantioseparation of Unfunctionalized Chiral Alkanes: A Challenge in Separation Science (Overview, State of the Art, and Perspectives). Chirality 2009, 21, 182–198. [Google Scholar] [CrossRef] [PubMed]
  107. Stephany, O.; Dron, F.; Tisse, S.; Martinez, A.; Nuzillard, J.M.; Peulon-Agasse, V.; Cardinaël, P.; Bouillon, J.P. (L)- or (d)-Valine Tert-Butylamide Grafted on Permethylated β-Cyclodextrin Derivatives as New Mixed Binary Chiral Selectors. Versatile Tools for Capillary Gas Chromatographic Enantioseparation. J. Chromatogr. A 2009, 1216, 4051–4062. [Google Scholar] [CrossRef]
  108. Kühnle, M.; Kreidler, D.; Holtin, K.; Czesla, H.; Schuler, P.; Schurig, V.; Albert, K. Online Coupling of Enantioselective Capillary Gas Chromatography with Proton Nuclear Magnetic Resonance Spectroscopy. Chirality 2010, 22, 808–812. [Google Scholar] [CrossRef]
  109. Schurig, V. Utilisation Des Cyclodextrines Dérivées Comme Sélecteurs de Séparation Énantiomérique Par Chromatographie Gazeuse. Ann. Pharm. Fr. 2010, 68, 82–98. [Google Scholar] [CrossRef]
  110. Drake, S.; Morrison, C.; Smith, F. Simultaneous Chiral Separation of Methylamphetamine and Common Precursors Using Gas Chromatography/Mass Spectrometry. Chirality 2011, 23, 593–601. [Google Scholar] [CrossRef]
  111. Xie, S.M.; Zhang, Z.J.; Wng, Z.Y.; Yuan, L.M. Chiral Metal-Organic Frameworks for High-Resolution Gas Chromatographic Separations. J. Am. Chem. Soc. 2011, 133, 11892–11895. [Google Scholar] [CrossRef]
  112. Mohr, S.; Weiß, J.A.; Spreitz, J.; Schmid, M.G. Chiral Separation of New Cathinone- and Amphetamine-Related Designer Drugs by Gas Chromatography-Mass Spectrometry Using Trifluoroacetyl-l-Prolyl Chloride as Chiral Derivatization Reagent. J. Chromatogr. A 2012, 1269, 352–359. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, L.; McDonald, J.A.; Khan, S.J. Enantiomeric Analysis of Polycyclic Musks in Water by Chiral Gas Chromatography-Tandem Mass Spectrometry. J. Chromatogr. A 2013, 1303, 66–75. [Google Scholar] [CrossRef] [PubMed]
  114. Xie, S.M.; Zhang, X.H.; Zhang, Z.J.; Yuan, L.M. Porous Chiral Metal-Organic Framework InH(D-C10H14O4)2 with Anionic-Type Diamond Network for High-Resolution Gas Chromatographic Enantioseparations. Anal. Lett. 2013, 46, 753–763. [Google Scholar] [CrossRef]
  115. Liu, H.; Xie, S.M.; Ai, P.; Zhang, J.H.; Zhang, M.; Yuan, L.M. Metal-Organic Framework Co(D-Cam)1/2(Bdc)1/2(Tmdpy) for Improved Enantioseparations on a Chiral Cyclodextrin Stationary Phase in Gas Chromatography. Chempluschem 2014, 79, 1103–1108. [Google Scholar] [CrossRef]
  116. Myrgorodska, I.; Meinert, C.; Martins, Z.; Le Sergeant d’Hendecourt, L.; Meierhenrich, U.J. Quantitative Enantioseparation of Amino Acids by Comprehensive Two-Dimensional Gas Chromatography Applied to Non-Terrestrial Samples. J. Chromatogr. A 2016, 1433, 131–136. [Google Scholar] [CrossRef] [PubMed]
  117. Henry, M.C.; Road, G.S.; Prussia, K.; Yonker, C.R.; Pacific, B.; National, N.; Box, L.P.O.; Richland, K. Supercritical Fluid Chromatography, Pressurized Liquid Extraction, and Supercritical Fluid Extraction. J. Chromatogr. A 2006, 78, 3909–3916. [Google Scholar] [CrossRef] [PubMed]
  118. De Klerck, K.; Mangelings, D.; Vander Heyden, Y. Supercritical Fluid Chromatography for the Enantioseparation of Pharmaceuticals. J. Pharm. Biomed. Anal. 2012, 69, 77–92. [Google Scholar] [CrossRef]
  119. Miller, L. Preparative Enantioseparations Using Supercritical Fluid Chromatography. J. Chromatogr. A 2012, 1250, 250–255. [Google Scholar] [CrossRef]
  120. Klesper, E.; Iber, K.; Clark, M. High Pressure Gas Chromatography above Critical Temperatures. J. Org. Chem. 1962, 27, 700–706. [Google Scholar]
  121. Taylor, L.T. Supercritical Fluid Chromatography for the 21st Century. J. Supercrit. Fluids 2009, 47, 566–573. [Google Scholar] [CrossRef]
  122. Pasquali, I.; Bettini, R. Are Pharmaceutics Really Going Supercritical? Int. J. Pharm. 2008, 364, 176–187. [Google Scholar] [CrossRef]
  123. Chester, T.L.; Pinkston, J.D.; Raynie, D.E. Supercritical Fluid Chromatography and Extraction. Anal. Chem. 1994, 66, 106–130. [Google Scholar] [CrossRef] [PubMed]
  124. Williams, K.L.; Sander, L.C. Enantiomer Separations on Chiral Stationary Phases in Supercritical Fluid Chromatography. J. Chromatogr. A 1997, 785, 149–158. [Google Scholar] [CrossRef]
  125. Mourier, P.A.; Eliot, E.; Caude, M.H.; Rosset, R.H.; Bouchet, L. Supercritical and Subcritical Fluid Chromatography on a Chiral Stationary Phase for the Resolution of Phosphine Oxide Enantiomers. Anal. Chem. 1985, 8, 2819–2823. [Google Scholar] [CrossRef]
  126. Macaudière, P.; Caude, M.; Rosset, R. Chiral Resolutions in SFC: Mechanisms and Applications with Various Chiral Stationary Phases. J. Chromatogr. Sci. 1989, 27, 583–591. [Google Scholar] [CrossRef]
  127. Stringham, R.W.; Lynam, K.G.; Grasso, C.C. Application of Subcritical Fluid Chromatography to Rapid Chiral Method Development. Anal. Chem. 1994, 66, 1949–1954. [Google Scholar] [CrossRef]
  128. Johannsen, M. Separation of Enantiomers of Ibuprofen on Chiral Stationary Phases by Packed Column Supercritical Fluid Chromatography. J. Chromatogr. A 2001, 937, 135–138. [Google Scholar] [CrossRef]
  129. Toribio, L.; Bernal, J.L.; Nozal, M.J.; Jimenez, J.J.; Nieto, E.M. Applications of the Chiralpak AD and Chiralcel OD Chiral Columns in the Enantiomeric Separation of Several Dioxolane Compounds by Supercritical Fluid Chromatography Q. J. Chromatogr. A 2001, 921, 305–313. [Google Scholar] [CrossRef]
  130. Wang, T.; Barber, M.; Hardt, I.; Kassel, D.B. Mass-Directed Fractionation and Isolation of Pharmaceutical Compounds by Packed-Column Supercritical Fluid Chromatography/Mass Spectrometry. Rapid Commun. Mass Spectrom. 2001, 15, 2067–2075. [Google Scholar] [CrossRef]
  131. Garzotti, M.; Hamdan, M. Supercritical Fluid Chromatography Coupled to Electrospray Mass Spectrometry: A Powerful Tool for the Analysis of Chiral Mixtures. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2002, 770, 53–61. [Google Scholar] [CrossRef]
  132. Liu, Y.; Berthod, A.; Mitchell, C.R.; Xiao, T.L.; Zhang, B.; Armstrong, D.W. Super/Subcritical Fluid Chromatography Chiral Separations with Macrocyclic Glycopeptide Stationary Phases. J. Chromatogr. A 2002, 978, 185–204. [Google Scholar] [CrossRef]
  133. Nozal, M.J.; Toribio, L.; Bernal, J.L.; Nieto, E.M.; Jime, J.J. Separation of Albendazole Sulfoxide Enantiomers by Chiral Supercritical-Fluid Chromatography. J. Biochem. Biophys. Methods 2002, 54, 339–345. [Google Scholar] [CrossRef]
  134. Nozal, M.J.; Toribio, L.; Bernal, J.L.; Castano, N.S. Eparation of Triadimefon and Triadimenol Enantiomers and Diastereoisomers by Supercritical Fluid Chromatography. J. Chromatogr. A. 2003, 986, 135–141. [Google Scholar] [CrossRef]
  135. Toribio, L.; Nozal, M.J.; Bernal, J.L.; Nieto, E.M. Use of Semipreparative Supercritical Fluid Chromatography to Obtain Small Quantities of the Albendazole Sulfoxide Enantiomers. J. Chromatogr. A 2003, 1011, 155–161. [Google Scholar] [CrossRef]
  136. Toribio, L.; Nozal, M.J.; Bernal, J.L.; Jimenez, J.J.; Alonso, C. Chiral Separation Ofsome Triazole Pesticides by Supercritical Fluid Chromatography. J. Chromatogr. A 2004, 1046, 249–253. [Google Scholar]
  137. del Nozal, M.J.; Toribio, L.; Bernal, J.L.; Alonso, C.; Jiménez, J.J. Chiral Separation of Omeprazole and Several Related Benzimidazoles Using Supercritical Fluid Chromatography. J. Sep. Sci. 2004, 27, 1023–1029. [Google Scholar] [CrossRef] [PubMed]
  138. Maftouh, M.; Granier-loyaux, C.; Chavana, E.; Marini, J.; Pradines, A.; Vander, Y.; Picard, C. Screening Approach for Chiral Separation of Pharmaceuticals Part III. Supercritical Fluid Chromatography for Analysis and Purification in Drug Discovery. J. Chromatogr. A 2005, 1088, 67–81. [Google Scholar] [CrossRef]
  139. Toribio, L.; Del Nozal, M.J.; Bernal, J.L.; Alonso, C.; Jiménez, J.J. Comparative Study of the Enantioselective Separation of Several Antiulcer Drugs by High-Performance Liquid Chromatography and Supercritical Fluid Chromatography. J. Chromatogr. A 2005, 1091, 118–123. [Google Scholar] [CrossRef] [PubMed]
  140. Yang, Y.; Su, B.; Yan, Q.; Ren, Q. Separation of Naproxen Enantiomers by Supercritical/Subcritical Fluid Chromatography. J. Pharm. Biomed. Anal. 2005, 39, 815–818. [Google Scholar] [CrossRef]
  141. Coe, R.A.; Rathe, J.O.; Lee, J.W. Supercritical Fluid Chromatography-Tandem Mass Spectrometry for Fast Bioanalysis of R/S-Warfarin in Human Plasma. J. Pharm. Biomed. Anal. 2006, 42, 573–580. [Google Scholar] [CrossRef]
  142. Toribio, L.; Alonso, C.; del Nozal, M.J.; Bernal, J.L.; Jiménez, J.J. Enantiomeric Separation of Chiral Sulfoxides by Supercritical Fluid Chromatography. J. Sep. Sci. 2006, 29, 1363–1372. [Google Scholar] [CrossRef] [PubMed]
  143. Toribio, L.; del Nozal, M.J.; Bernal, J.L.; Alonso, C.; Jiménez, J.J. Enantiomeric Separation of Several Antimycotic Azole Drugs Using Supercritical Fluid Chromatography. J. Chromatogr. A 2007, 1144, 255–261. [Google Scholar] [CrossRef]
  144. Wang, Z.; Jonca, M.; Lambros, T.; Ferguson, S.; Goodnow, R. Exploration of Liquid and Supercritical Fluid Chromatographic Chiral Separation and Purification of Nutlin-3-A Small Molecule Antagonist of MDM2. J. Pharm. Biomed. Anal. 2007, 45, 720–729. [Google Scholar] [CrossRef]
  145. West, C.; Bouet, A.; Gillaizeau, I.; Coudert, G.; Lafosse, M.; Lesellier, E. Chiral Separation of Phospine-Containing a -Amino Acid Derivatives Using Two Complementary Cellulosic Stationary Phases in Supercritical Fluid Chromatography. Chirality 2010, 251, 242–251. [Google Scholar] [CrossRef] [PubMed]
  146. Toribio, L.; Nozal, M.J.; Bernal, J.L.; Bernal, J.; Martín, M.T. Study of the Enantiomeric Separation of an Acetamide Intermediate by Using Supercritical Fluid Chromatography and Several Polysaccharide Based Chiral Stationary Phases. J. Chromatogr. A 2011, 1218, 4886–4891. [Google Scholar] [CrossRef] [PubMed]
  147. West, C.; Guenegou, G.; Zhang, Y.; Morin-Allory, L. Insights into Chiral Recognition Mechanisms in Supercritical Fluid Chromatography. II. Factors Contributing to Enantiomer Separation on Tris-(3,5-Dimethylphenylcarbamate) of Amylose and Cellulose Stationary Phases. J. Chromatogr. A 2011, 1218, 2033–2057. [Google Scholar] [CrossRef] [PubMed]
  148. Hamman, C.; Schmidt, D.E.; Wong, M.; Hayes, M. The Use of Ammonium Hydroxide as an Additive in Supercritical Fluid Chromatography for Achiral and Chiral Separations and Purifications of Small, Basic Medicinal Molecules. J. Chromatogr. A 2012, 1218, 7886–7894. [Google Scholar] [CrossRef]
  149. West, C.; Bouet, A.; Routier, S.; Lesellier, E. Effects of Mobile Phase Composition and Temperature on the Supercritical Fluid Chromatography Enantioseparation of Chiral Fluoro-Oxoindole-Type Compounds with Chlorinated Polysaccharide Stationary Phases. J. Chromatogr. A 2012, 1269, 325–335. [Google Scholar] [CrossRef]
  150. Tao, Y.; Dong, F.; Xu, J.; Liu, X.; Cheng, Y.; Liu, N.; Chen, Z.; Zheng, Y. Green and Sensitive Supercritical Fluid Chromatographic-Tandem Mass Spectrometric Method for the Separation and Determination of Flutriafol Enantiomers in Vegetables, Fruits, and Soil. J. Agric. Food Chem. 2014, 62, 11457–11464. [Google Scholar] [CrossRef] [PubMed]
  151. Regalado, E.L.; Welch, C.J. Pushing the Speed Limit in Enantioselective Supercritical Fluid Chromatography. J. Sep. Sci. 2015, 38, 2826–2832. [Google Scholar] [CrossRef]
  152. Khater, S.; Lozac’h, M.A.; Adam, I.; Francotte, E.; West, C. Comparison of Liquid and Supercritical Fluid Chromatography Mobile Phases for Enantioselective Separations on Polysaccharide Stationary Phases. J. Chromatogr. A 2016, 1467, 463–472. [Google Scholar] [CrossRef] [PubMed]
  153. Jung, M.; Schurig, V. Extending the Scope of Enantiomer Separation by Capillary Supercritical Fluid Chromatography on Immobilized Poly. J. High. Resolut. Chromatogr. 1993, 16, 215–223. [Google Scholar] [CrossRef]
  154. Armstrong, D.W. Optical Isomer Separation by Liquid Chromatography. Anal. Chem. 1987, 59, 84A–97A. [Google Scholar] [CrossRef] [PubMed]
  155. Däppen, R.; Arm, H.; Meyer, V.R. Applications and Limitations of Commercially Available Chiral Stationary Phases for High-Performance Liquid Chromatography. J. Chromatogr. A 1986, 373, 1–20. [Google Scholar] [CrossRef]
  156. Pham-Huy, C.; Villain-Pautet, G.; Hua, H.; Chikhi-Chorfi, N.; Galons, H.; Thevenin, M.; Claude, J.R.; Warnet, J.M. Separation of Oxazepam, Lorazepam, and Temazepam Enantiomers by HPLC on a Derivatized Cyclodextrin-Bonded Phase: Application to the Determination of Oxazepam in Plasma. J. Biochem. Biophys. Methods 2002, 54, 287–299. [Google Scholar] [CrossRef]
  157. Boatto, G.; Nieddu, M.; Faedda, M.V.; De Caprariis, P. Enantiomeric Separation by HPLC of 1,4-Dihydropyridines with Vancomycin as Chiral Selector. Chirality 2003, 15, 494–497. [Google Scholar] [CrossRef]
  158. Narayana, C.; Suresh, T.; Mahender Rao, S.; Dubey, P.K.; Moses Babu, J. A Validated Chiral HPLC Method for the Enantiomeric Separation of Linezolid on Amylose Based Stationary Phase. J. Pharm. Biomed. Anal. 2003, 32, 21–28. [Google Scholar] [CrossRef]
  159. Putkonen, T.; Tolvanen, A.; Jokela, R.; Caccamese, S.; Parrinello, N. Total Synthesis of (±)-Tangutorine and Chiral HPLC Separation of Enantiomers. Tetrahedron 2003, 59, 8589–8595. [Google Scholar] [CrossRef]
  160. Zhang, X.; Ouyang, J.; Baeyens, W.R.G.; Zhai, S.; Yang, Y.; Huang, G. Enantiomeric Separation of β-Blockers by HPLC Using (R)-1-Naphthylglycine and 3,5-Dinitrobenzoic Acid as Chiral Stationary Phase. J. Pharm. Biomed. Anal. 2003, 31, 1047–1057. [Google Scholar] [CrossRef]
  161. Kumar, Y.R.; Ramulu, G.; Vevakanand, V.V.; Vaidyanathan, G.; Srinivas, K.; Kumar, M.K.; Mukkanti, K.; Reddy, M.S.; Venkatraman, S.; Suryanarayana, M.V. A Validated Chiral HPLC Method for the Enantiomeric Separation of Tolterodine Tartarate. J. Pharm. Biomed. Anal. 2004, 35, 1279–1285. [Google Scholar] [CrossRef]
  162. Lämmerhofer, M.; Gyllenhaal, O.; Lindner, W. HPLC Enantiomer Separation of a Chiral 1,4-Dihydropyridine Monocarboxylic Acid. J. Pharm. Biomed. Anal. 2004, 35, 259–266. [Google Scholar] [CrossRef]
  163. Caccamese, S.; Caruso, C.; Parrinello, N.; Savarino, A. High-Performance Liquid Chromatographic Separation and Chiroptical Properties of the Enantiomers of Naringenin and Other Flavanones. J. Chromatogr. A 2005, 1076, 155–162. [Google Scholar] [CrossRef]
  164. Ali, I.; Naim, L.; Ghanem, A.; Aboul-Enein, H.Y. Chiral Separations of Piperidine-2,6-Dione Analogues on Chiralpak IA and Chiralpak IB Columns by Using HPLC. Talanta 2006, 69, 1013–1017. [Google Scholar] [CrossRef] [PubMed]
  165. Gazić, I.; Bosak, A.; Šinko, G.; Vinković, V.; Kovarik, Z. Preparative HPLC Separation of Bambuterol Enantiomers and Stereoselective Inhibition of Human Cholinesterases. Anal. Bioanal. Chem. 2006, 385, 1513–1519. [Google Scholar] [CrossRef]
  166. Sun, P.; Wang, C.; Armstrong, D.; Péter, A.; Forró, E. Separation of Enantiomers of β-Lactams by HPLC Using Cyclodextrin-Based Chiral Stationary Phases. J. Liq. Chromatogr. Relat. Technol. 2006, 29, 1847–1860. [Google Scholar] [CrossRef]
  167. Sun, P.; Krishnan, A.; Yadav, A.; Singh, S.; MacDonnell, F.M.; Armstrong, D.W. Enantiomeric Separations of Ruthenium (II) Polypyridyl Complexes Using HPLC With Cyclofructan Chiral Stationary Phases. Inorg. Chem. 2007, 46, 10312–10320. [Google Scholar] [CrossRef] [PubMed]
  168. Hoffmann, C.V.; Pell, R.; Lämmerhofer, M.; Lindner, W. Synergistic Effects on Enantioselectivity of Zwitterionic Chiral Stationary Phases for Separations of Chiral Acids, Bases, and Amino Acids by HPLC. Anal. Chem. 2008, 80, 8780–8789. [Google Scholar] [CrossRef] [PubMed]
  169. Ali, I.; Gaitonde, V.D.; Aboul-Enein, H.Y.; Hussain, A. Chiral Separation of β-Adrenergic Blockers on CelluCoat Column by HPLC. Talanta 2009, 78, 458–463. [Google Scholar] [CrossRef] [PubMed]
  170. Zhou, Y.; Li, L.; Lin, K.; Zhu, X.; Liu, W. Enantiomer Separation of Triazole Fungicides by High-Performance Liquid Chromatography. Chirality 2009, 21, 421–427. [Google Scholar]
  171. Ye, J.; Yu, W.; Chen, G.; Shen, Z.; Zeng, S. Enantiomeric Separation of 2-Arylpropionic Acid Nonsteroidal Anti-Inflammatory Drugs by HPLC with Hydroxypropyl-β-Cyclodextrin as Chiral Mobile Phase Additive. Biomed. Chromatogr. 2010, 24, 799–807. [Google Scholar] [CrossRef]
  172. Ali, I.; Al-Othman, Z.A.; Hussain, A.; Saleem, K.; Aboul-Enein, H.Y. Chiral Separation of β-Adrenergic Blockers in Human Plasma by SPE-HPLC. Chromatographia 2011, 73, 251–256. [Google Scholar] [CrossRef]
  173. Bi, W.; Tian, M.; Row, K.H. Chiral Separation and Determination of Ofloxacin Enantiomers by Ionic Liquid-Assisted Ligand-Exchange Chromatography. Analyst 2011, 136, 379–387. [Google Scholar] [CrossRef] [PubMed]
  174. Li, X.; Liu, Y.; Hu, C.; Bai, L.; Gao, B.; Huang, K. Direct Optical Resolution of Chiral Pesticides by High Performance Liquid Chromatography. Chin. J. Chem. Eng. 2011, 19, 603–609. [Google Scholar] [CrossRef]
  175. Jibuti, G.; Mskhiladze, A.; Takaishvili, N.; Karchkhadze, M.; Chankvetadze, L.; Farkas, T.; Chankvetadze, B. HPLC Separation of Dihydropyridine Derivatives Enantiomers with Emphasis on Elution Order Using Polysaccharide-Based Chiral Columns. J. Sep. Sci. 2012, 35, 2529–2537. [Google Scholar] [CrossRef]
  176. Matarashvili, I.; Chankvetadze, L.; Fanali, S.; Farkas, T.; Chankvetadze, B. HPLC Separation of Enantiomers of Chiral Arylpropionic Acid Derivatives Using Polysaccharide-Based Chiral Columns and Normal-Phase Eluents with Emphasis on Elution Order. J. Sep. Sci. 2013, 36, 140–147. [Google Scholar] [CrossRef]
  177. Padivitage, N.L.T.; Dodbiba, E.; Breitbach, Z.S.; Armstrong, D.W. Enantiomeric Separations of Illicit Drugs and Controlled Substances Using Cyclofructan-Based (LARIHC) and Cyclobond I 2000 RSP HPLC Chiral Stationary Phases. Drug Test. Anal. 2014, 6, 542–551. [Google Scholar] [CrossRef]
  178. Shu, Y.; Breitbach, Z.S.; Dissanayake, M.K.; Perera, S.; Aslan, J.M.; Alatrash, N.; MacDonnell, F.M.; Armstrong, D.W. Enantiomeric Separations of Ruthenium (II) Polypyridyl Complexes Using HPLC With Cyclofructan Chiral Stationary Phases. Chirality 2015, 27, 64–70. [Google Scholar] [CrossRef]
  179. Broughton, D.B.; Gerhold, C.G. Continuous Sorption Process Employing Fixed Bed of Sorbent and Moving Inlets and Outlets. U.S. Patent US2985589A, 23 May 1961. [Google Scholar]
  180. Nicoud, R.M. Chromatographic Processes; Cambridge University Press: Cambridge, UK, 2015. [Google Scholar]
  181. Negawa, M.; Shoji, F. Optical Resolution by Simulated Moving-Bed Adsorption Technology. J. Chromatogr. A 1992, 590, 113–117. [Google Scholar] [CrossRef]
  182. Cavoy, E.; Deltent, M.F.; Lehoucq, S.; Miggiano, D. Laboratory-Developed Simulated Moving Bed for Chiral Drug Separations. J. Chromatogr. A 1997, 769, 49–57. [Google Scholar] [CrossRef]
  183. Devant, R.M.; Jonas, R.; Schulte, M.; Keil, A.; Charton, F. Enantiomer Separation of a Novel Ca-Sensitizing Drug by Simulated Moving Bed (SMB)—Chromatography. J. Prakt. Chem. 1997, 339, 315–321. [Google Scholar] [CrossRef]
  184. Francotte, E.R.; Richert, P. Applications of Simulated Moving-Bed Chromatography to the Separation of the Enantiomers of Chiral Drugs. J. Chromatogr. A 1997, 769, 101–107. [Google Scholar] [CrossRef]
  185. Pais, L.S.; Loureiro, J.M.; Rodrigues, A.E. Separation of 1,1′-Bi-2-Naphthol Enantiomers by Continuous Chromatography in Simulated Moving Bed. Chem. Eng. Sci. 1997, 52, 245–257. [Google Scholar] [CrossRef]
  186. Pais, L.S.; Loureiro, J.M.; Rodrigues, A.E. Modeling, Simulation and Operation of a Simulated Moving Bed for Continuous Chromatographic Separation of 1,1′-Bi-2-Naphthol Enantiomers. J. Chromatogr. A 1997, 769, 25–35. [Google Scholar] [CrossRef]
  187. Francotte, E.; Richert, P.; Mazzotti, M.; Morbidelli, M. Simulated Moving Bed Chromatographic Resolution of a Chiral Antitussive. J. Chromatogr. A 1998, 796, 239–248. [Google Scholar] [CrossRef]
  188. Heuer, C.; Küsters, E.; Plattner, T.; Seidel-Morgenstern, A. Design of the Simulated Moving Bed Process Based on Adsorption Isotherm Measurements Using a Perturbation Method. J. Chromatogr. A 1998, 827, 175–191. [Google Scholar] [CrossRef]
  189. Pais, L.S.; Loureiro, J.M.; Rodrigues, A.E. Modeling Strategies for Enantiomers Separation by SMB Chromatography. AIChE J. 1998, 44, 561–569. [Google Scholar] [CrossRef]
  190. Khattabi, S.; Cherrak, D.E.; Mihlbachler, K.; Guiochon, G. Enantioseparation of 1-Phenyl-1-Propanol by Simulated Moving Bed under Linear and Nonlinear Conditions. J. Chromatogr. A 2000, 893, 307–319. [Google Scholar] [CrossRef]
  191. Huthmann, E.; Juza, M. Modification of a Commercial Chiral Stationary Phase: Influences on Enantiomer Separations Using Simulated Moving Bed Chromatography. J. Chromatogr. A 2001, 908, 185–200. [Google Scholar] [CrossRef]
  192. Francotte, E.; Leutert, T.; Vecchia, L.L.; Ossola, F.; Richert, P.; Schmidt, A. Preparative Resolution of the Enantiomers of Tert-Leucine Derivatives by Simulated Moving Bed Chromatography. Chirality 2002, 14, 313–317. [Google Scholar] [CrossRef]
  193. Lee, K.B.; Chin, C.Y.; Xie, Y.; Cox, G.B.; Wang, N.L. Standing-Wave Design of a Simulated Moving Bed under a Pressure Limit for Enantioseparation of Phenylpropanolamine. Ind. Eng. Chem. Res. 2005, 44, 3249–3267. [Google Scholar] [CrossRef]
  194. Amanullah, M.; Grossmann, C.; Mazzotti, M.; Morari, M.; Morbidelli, M. Experimental Implementation of Automatic “cycle to Cycle” Control of a Chiral Simulated Moving Bed Separation. J. Chromatogr. A 2007, 1165, 100–108. [Google Scholar] [CrossRef] [PubMed]
  195. Choi, Y.J.; Han, S.K.; Chung, S.T.; Row, K.H. Separation of Racemic Bupivacaine Using Simulated Moving Bed with Mathematical Model. Biotechnol. Bioprocess. Eng. 2007, 12, 625–633. [Google Scholar] [CrossRef]
  196. Zhang, L.; Gedicke, K.; Kuznetsov, M.A.; Staroverov, S.M.; Seidel-Morgenstern, A. Application of an Eremomycin-Chiral Stationary Phase for the Separation of Dl-Methionine Using Simulated Moving Bed Technology. J. Chromatogr. A 2007, 1162, 90–96. [Google Scholar] [CrossRef] [PubMed]
  197. Araújo, J.M.M.; Rodrigues, R.C.R.; Eusébio, M.F.J.; Mota, J.P.B. On-Line Enantiomeric Analysis Using High-Performance Liquid Chromatography in Chiral Separation by Simulated Moving Bed. J. Chromatogr. A 2008, 1189, 292–301. [Google Scholar] [CrossRef]
  198. Zabka, M.; Minceva, M.; Gomes, P.S.; Rodrigues, A.E. Chiral Separation of R,S-α- Tetralol by Simulated Moving Bed. Sep. Sci. Technol. 2008, 43, 727–765. [Google Scholar] [CrossRef]
  199. Acetti, D.; Langel, C.; Brenna, E.; Fuganti, C.; Mazzotti, M. Intermittent Simulated Moving Bed Chromatographic Separation of (RS,RS)-2-(2,4-Difluorophenyl)Butane-1,2,3-Triol. J. Chromatogr. A 2010, 1217, 2840–2846. [Google Scholar] [CrossRef] [PubMed]
  200. Langel, C.; Grossmann, C.; Jermann, S.; Mazzotti, M.; Morari, M.; Morbidelli, M. Experimental Optimizing Control of the Simulated Moving Bed Separation of Tröger’s Base Enantiomers. Ind. Eng. Chem. Res. 2010, 49, 11996–12003. [Google Scholar] [CrossRef]
  201. Lee, E.; Park, M.B.; Kim, J.M.; Kim, W.S.; Kim, I.H. Simulated Moving-Bed for Separation of Mandelic Acid Racemic Mixtures. Korean J. Chem. Eng. 2010, 27, 231–234. [Google Scholar] [CrossRef]
  202. Katsuo, S.; Mazzotti, M. Intermittent Simulated Moving Bed Chromatography: 2. Separation of Tröger’s Base Enantiomers. J. Chromatogr. A 2010, 1217, 3067–3075. [Google Scholar] [CrossRef]
  203. Katsuo, S.; Langel, C.; Sandré, A.L.; Mazzotti, M. Intermittent Simulated Moving Bed Chromatography: 3. Separation of Tröger’s Base Enantiomers under Nonlinear Conditions. J. Chromatogr. A 2011, 1218, 9345–9352. [Google Scholar] [CrossRef] [PubMed]
  204. Ribeiro, A.E.; Gomes, P.A.; Pais, L.; Rodrigues, A.E. Chiral Separation of Flurbiprofen Enantiomers by Preparative and Simulated Moving Bed Chromatography. Chirality 2011, 23, 602–611. [Google Scholar] [CrossRef] [PubMed]
  205. Ribeiro, A.E.; Gomes, P.S.; Pais, L.S.; Rodrigues, A.E. Separation Science and Technology Chiral Separation of Ketoprofen Enantiomers by Preparative and Simulated Moving Bed Chromatography Chiral Separation of Ketoprofen Enantiomers by Preparative and Simulated Moving Bed Chromatography. Sep. Sci. Technol. 2011, 4611, 1726–1739. [Google Scholar] [CrossRef]
  206. Gong, R.; Lin, X.; Li, P.; Yu, J.; Rodrigues, A.E. Experiment and Modeling for the Separation of Guaifenesin Enantiomers Using Simulated Moving Bed and Varicol Units. J. Chromatogr. A 2014, 1363, 242–249. [Google Scholar] [CrossRef] [PubMed]
  207. Cunha, F.C.; Secchi, A.R.; de Souza, M.B.; Barreto, A.G. Separation of Praziquantel Enantiomers Using Simulated Moving Bed Chromatographic Unit with Performance Designed for Semipreparative Applications. Chirality 2019, 31, 583–591. [Google Scholar] [CrossRef] [PubMed]
  208. Blehaut, J.; Nicoud, R.M. Recent Aspects in Simulated Moving Bed. Analusis 1998, 26, 60–70. [Google Scholar] [CrossRef]
  209. Cunha, F.C. Chromatographic Separation of Praziquantel Racemate Using Simulated Moving Bed: From Unit Design to Dynamic Studies with Online Measurements; Universidade Federal do Rio de Janeiro: Rio de Janeiro, Brazil, 2021. [Google Scholar]
Figure 1. Chiral carbon (*) bound to four different groups in different representations.
Figure 1. Chiral carbon (*) bound to four different groups in different representations.
Encyclopedia 02 00011 g001
Figure 2. Pairs of enantiomers of (a) chiral silicon, (b) chiral nitrogen, (c) chiral sulfur, (d) chiral phosphorus, (e) cobalt complex, (f) allenes and (g) atropisomers.
Figure 2. Pairs of enantiomers of (a) chiral silicon, (b) chiral nitrogen, (c) chiral sulfur, (d) chiral phosphorus, (e) cobalt complex, (f) allenes and (g) atropisomers.
Encyclopedia 02 00011 g002
Figure 3. Number of enantiomers related to the numbers of chiral carbons (*) for (a) lactic acid and (b) isoleucine.
Figure 3. Number of enantiomers related to the numbers of chiral carbons (*) for (a) lactic acid and (b) isoleucine.
Encyclopedia 02 00011 g003
Figure 4. Tartaric acid presents (a) two enantiomers and (b) a meso isomer.
Figure 4. Tartaric acid presents (a) two enantiomers and (b) a meso isomer.
Encyclopedia 02 00011 g004
Figure 5. (a) Amino acids L and D -Alanine and (b) monosaccharides L and D -Fructose.
Figure 5. (a) Amino acids L and D -Alanine and (b) monosaccharides L and D -Fructose.
Encyclopedia 02 00011 g005
Figure 6. Deoxyribose (a) and ribose (b) monosaccharides structural formulas from DNA and RNA and their chiral carbons.
Figure 6. Deoxyribose (a) and ribose (b) monosaccharides structural formulas from DNA and RNA and their chiral carbons.
Encyclopedia 02 00011 g006
Figure 7. Perfect (a) and failed (b) connection between enantiomers and biological receptor.
Figure 7. Perfect (a) and failed (b) connection between enantiomers and biological receptor.
Encyclopedia 02 00011 g007
Figure 8. Enantiomers obtained by Chiral Route (a) Racemic Route (b).
Figure 8. Enantiomers obtained by Chiral Route (a) Racemic Route (b).
Encyclopedia 02 00011 g008
Figure 9. Main methods for enantiomer separation.
Figure 9. Main methods for enantiomer separation.
Encyclopedia 02 00011 g009
Figure 10. Illustration of conglomerate (a) and racemic compound (b) crystals.
Figure 10. Illustration of conglomerate (a) and racemic compound (b) crystals.
Encyclopedia 02 00011 g010
Figure 11. Chiral recognition sites in a porous solid membrane selecting a specific enantiomer.
Figure 11. Chiral recognition sites in a porous solid membrane selecting a specific enantiomer.
Encyclopedia 02 00011 g011
Figure 12. Adsorbed Enantiomer 1 (E1) onto the chiral environment according to Dalgliesh’s three-point rule and non-absorbed Enantiomer 2 (E2).
Figure 12. Adsorbed Enantiomer 1 (E1) onto the chiral environment according to Dalgliesh’s three-point rule and non-absorbed Enantiomer 2 (E2).
Encyclopedia 02 00011 g012
Figure 13. Schematic diagram of a TMB chromatograph.
Figure 13. Schematic diagram of a TMB chromatograph.
Encyclopedia 02 00011 g013
Figure 14. Schematic diagram of an SMB chromatograph and its operation where the more retained compound is adsorbed onto the solid porous phase.
Figure 14. Schematic diagram of an SMB chromatograph and its operation where the more retained compound is adsorbed onto the solid porous phase.
Encyclopedia 02 00011 g014
Table 1. Types of isomers and their structural formula [3].
Table 1. Types of isomers and their structural formula [3].
IsomerismChemical FormulaStructural Formula
StructuralFunction C 3 H 6 O Encyclopedia 02 00011 i001 Encyclopedia 02 00011 i002
PropanalPropanone
Chain C 4 H 10 Encyclopedia 02 00011 i003 Encyclopedia 02 00011 i004
n-ButaneIsobutane
Position C 5 H 10 O Encyclopedia 02 00011 i005 Encyclopedia 02 00011 i006
2-Pentanone3-Pentanone
Metamerism C 4 H 10 O Encyclopedia 02 00011 i007 Encyclopedia 02 00011 i008
1-MethoxypropaneEthoxyethane
Tautomerism C 6 H 6 O Encyclopedia 02 00011 i009 Encyclopedia 02 00011 i010 Encyclopedia 02 00011 i011
OxepinBenzene oxide
StereoDiastereomer
(Geometric)
C 2 H 2 Cl 2 Encyclopedia 02 00011 i012 Encyclopedia 02 00011 i013
cis-1,2-dichloroethenetrans-1,2-dichloroethene
Enantiomer
(Optical)
C 3 H 6 O 3 Encyclopedia 02 00011 i014 Encyclopedia 02 00011 i015
(S)-Lactic acid(R)-Lactic acid
Table 2. Rules for enantiomer nomenclature.
Table 2. Rules for enantiomer nomenclature.
RuleIllustration
1st Ranking of atoms in descending order of atomic number Encyclopedia 02 00011 i062
2nd Pose the least atomic number atoms at the rear Encyclopedia 02 00011 i063
3rd Draw a circle arrow from the first to the third position. Encyclopedia 02 00011 i064
Table 3. Difference in biological activities between enantiomers [4].
Table 3. Difference in biological activities between enantiomers [4].
Compounds(S) Enantiomer Actuation(R) Enantiomer Actuation
Limonene Encyclopedia 02 00011 i016
Lemon odor
Encyclopedia 02 00011 i017
Orange odor
Carvone Encyclopedia 02 00011 i018
Caraway flavor
Encyclopedia 02 00011 i019
Spearmint flavor
Asparagine Encyclopedia 02 00011 i020
Bitter taste
Encyclopedia 02 00011 i021
Sweet taste
Aspartame Encyclopedia 02 00011 i022
Sweet taste
Encyclopedia 02 00011 i023
Bitter taste
Ethambutol Encyclopedia 02 00011 i024
Tuberculostatic
Encyclopedia 02 00011 i025
Causes blindness
Thalidomide Encyclopedia 02 00011 i026
Teratogen
Encyclopedia 02 00011 i027
Sedative
Penicillamine Encyclopedia 02 00011 i028
Antiarthritic
Encyclopedia 02 00011 i029
Mutagen
Ketamine Encyclopedia 02 00011 i030
Anesthetic
Encyclopedia 02 00011 i031
Hallucinogen
Dopa Encyclopedia 02 00011 i032
Anti-Parkinson
Encyclopedia 02 00011 i033
Serious side effects
Chloramphenicol Encyclopedia 02 00011 i034
Inactive
Encyclopedia 02 00011 i035
Antibacterial
Propranolol Encyclopedia 02 00011 i036
Antihypertensive, antiarrhythmic
Encyclopedia 02 00011 i037
Contraceptive
Paclobutrazol Encyclopedia 02 00011 i038
Plant growth regulator
Encyclopedia 02 00011 i039
Fungicide
Table 4. Top seller drugs worldwide in 2017 with chiral active ingredients (PHARMACOMPASS, 2019).
Table 4. Top seller drugs worldwide in 2017 with chiral active ingredients (PHARMACOMPASS, 2019).
Product NameChiral Active IngredientIndicationRevenue (in
Millions of Dollars)
Revlimid Encyclopedia 02 00011 i040
Lenalidomide
Oncology8187
Xarelto Encyclopedia 02 00011 i041
Rivaroxaban
Cardiovascular Diseases6590
Lyrica Encyclopedia 02 00011 i042
Pregabalin
Neurological/Mental Disorders5317
Imbruvica Encyclopedia 02 00011 i043
Ibrutinib
Oncology4466
Harvoni Encyclopedia 02 00011 i044
Ledipasvir
Encyclopedia 02 00011 i045
Sofosbuvir
Infectious Diseases (HIV, Hepatitis, etc.)4370
Symbicort Pulmicort Encyclopedia 02 00011 i046
Budesonide
Encyclopedia 02 00011 i047
Formoterol
Respiratory Disorders4360
Januvia Encyclopedia 02 00011 i048
Sitagliptin
Diabetes3737
Epclusa Encyclopedia 02 00011 i049
Sofosbuvir
Infectious Diseases (HIV, Hepatitis, etc.)3510
Triumeq Encyclopedia 02 00011 i050
Abacavir
Encyclopedia 02 00011 i051
Dolutegravir
Encyclopedia 02 00011 i052
Lamivudine
Infectious Diseases (HIV, Hepatitis, etc.)3470
Latuda Encyclopedia 02 00011 i053
Lurasidone
Neurological/Mental Disorders3350
Truvada Encyclopedia 02 00011 i054
Emtricitabine
Infectious Diseases (HIV, Hepatitis, etc.)3134
Nexium Encyclopedia 02 00011 i055
Esomeprazol
Gastrointestinal Disorders2795
Invega Sustenna
Encyclopedia 02 00011 i056
Paliperidone Palmitate
Neurological/Mental Disorders2569
Zytiga Encyclopedia 02 00011 i057
Abiraterone Acetate
Oncology2505
Table 6. Experimental studies of enantioresolutions applying membranes.
Table 6. Experimental studies of enantioresolutions applying membranes.
EnantiomersMembraneRef.
PhenylalanineImmobilized DNA membranes[57]
PhenylalanineDNA-immobilized chitosan membranes[58]
PhenylalaninePolyaniline[60]
Tryptophan, tyrosine, and phenylalaninepoly(γ-methyl-l-glutamate) membranes[61]
Lactic acid and alaninePolypropylene hollow-fiber module liquid membrane[62]
NaproxenPoly(4-vinylpyridine) /polypropylene membranes[63]
PropranololChiral derivatized polysulfone[64]
Tryptophan, henylglycine and phenylalanineImmobilized DNA membranes[65]
1-phenylethanol(R,R)-TADDOL[66]
N-protected amino acid derivativesAdamantyl-carbamoyl-11-
octadecylthioether-quinine/-quinidine
[67]
TryptophanChitosan/-cyclodextrin composite membranes[68]
TryptophanCellulose dialysis membranes[69]
2-phenyl-1-propanolGlutaraldehyde-crosslinked chitosan membranes[70]
TryptophanBSA-Immobilized and
BSA-Interpenetrating Network Polysulfone Membranes
[71]
KetoconazoleHydrophobic l-isopentyl tartrate and hydrophilic sulfobutylether--cyclodextrin[72]
AmlodipineHollow fiber supported liquid membrane[73]
PhenylalanineHollow fiber supported liquid membrane[74]
AtenololNano-sized chiral imprinted polymers[75]
IbuprofenL-tartaric acid derivatives[76]
DOPAL-Glutamic acid-Graphene oxide
based membranes
[77]
Tyrosine, phenylalanine and tryptophanD-penicillamine-modified membrane and N-acetyl-L-cysteine-modified membrane[78]
PhenylalanineRegenerated cellulose membranes[79]
ArginineChiral channel protein (FhuAF4)[80]
BaclofenSilica-based vancomycin-chiral stationary phase[81]
MethadoneChiral (2-hydroxypropyl)-β-cyclodextrin[82]
Table 7. Experimental studies of enantioresolutions applying gas chromatography.
Table 7. Experimental studies of enantioresolutions applying gas chromatography.
EnantiomersCSPGas CarrierRef.
β-Blockers(-)-α-methoxy-α-(trifluoromethyl)
phenylacetyl chloride
Helium[91]
β-pinene, sabinene, limonene, linalool, terpinen-4-ol, α-terpineol, linalyl acetateEtTBS-βCD and DB-5Hydrogen[92]
Chiral alcohols, chiral sulfoxides,
chiral epoxides and acetylated amines
Chiral
ionic liquid stationary phases
Helium[93]
Flurbiprofen and ketoprofenAgilent 6890
gas chromatograph
Helium[94]
Alanina, prolina, serina, asparagine, glutamine, lisine, ornitinaChirasil-l-Val capillary columnsHelium[95]
Obuprofen, fenoprofen
and ketoprofen methyl esters
Heptakis-(2,3-di-Omethyl-
6-O-t-butyldimethyl-silyl)-β-cyclodextrin
Hydrogen[96]
Chiral epoxidesCyclodextrin derivativesNitrogen[97]
α-amino acidsModified Linear DextrinsHydrogen[98]
Methyl branched compounds2,3-Di-O-methoxymethyl-6-O-tert-butyldimethylsilyl-γ-cyclodextrinHydrogen[99]
Hydrocarbons, underivatized alcohols, ketones, and proteinogenic amino acid
derivatives
Permethylated-βcyclodextrin
and resorcinarene with pendant L- or D-valine diamide groups
Hydrogen[100]
β-BlockersDB-5 and DB-17 dual-columnsHelium[101]
2,2-dimethylcyclopropane-carboxamideg-cyclodextrinHelium[102]
12 amino acidsN-Ethoxycarbonylation was combined with (S)-1-phenylethylamidationHelium[103]
β-amino acidCP-Chirasil-Dex CB and CP-Chirasil L-ValNitrogen[104]
1-phenylethanolPermethylated -cyclodextrinNitrogen[105]
3-methylhexane, 2,3-dimethylpentane, 3-methyl-heptane, 3,4-dimethylhexane, 2,4-dimethylhexane, 2,3-
dimethylhexane, 2,2,3-trimethylpentane
octakis(6-O-methyl-2,3-di-O-pentyl)-g-cyclodextrinHydrogen[106]
Amino acid derivatives(l)- or (d)-Valine tert-butylamide grafted on permethylated -cyclodextrinHelium[107]
2,4-dimethylhexaneoctakis(6-O-methyl-2,3-di-O-pentyl)-γ-cyclodextrinNitrogen[108]
α- and β-pinene, cis- and trans-pinane, 2,3 butanediol, γ-valerolacton, 1-phenylethyl-lamine, 1-phenylethanol, 2-ethyl-exanoic acidDerivatized cyclodextrinsHelium[109]
Methylamphetamineγ-cyclodextrinHelium[110]
Citronellal, camphor,
alanine, leucine, valine, isoleucine, 1-phenyl-1,2-ethandiol, phenylsuccinic
acid, and 1-phenyl-ethanol
Chiral Metal-Organic FrameworksNitrogen[111]
Cathinone- and amphetamine-related designer drugsTrifluoroacetyl-l-prolyl chlorideHelium[112]
Galaxolide, tonalide, phantolide, traseolide and cashmeranChiral heptakis(2,3- di-O-methyl-6-O-t-butyl dimethylsilyl)--cyclodextrinHelium[113]
Citronellal, 1-phenyl-1,2-ethandiol, 1-Phenyl-ethanol, 2-amino-1-butanol, limonene, methionine, prolinePorous Chiral Metal-Organic FrameworkNitrogen[114]
2-hexanol, linalool, citronellal, methyl l-b-hydroxyisobutyrate,
limonene, rose oxide, dihydrocarvyl
acetate, menthol, valine, and leucine
Metal–Organic Framework on a Chiral CyclodextrinNitrogen[115]
30 amino acidsPress-Tight© connected Varian-Chrompack Chirasil-l-ValHelium[116]
Table 8. Experimental studies of enantioresolutions applying Supercritical Fluid Chromatography.
Table 8. Experimental studies of enantioresolutions applying Supercritical Fluid Chromatography.
EnantiomersCSPRef.
IbuprofenKromasil CHI-TBB, Kromasil CHI-DMB, Chirobiotik T, Chiracel OBH and Chiralpal AD[128]
Dioxolane compoundsChiralpak AD and Chiralcel OD[129]
Enantiomeric pharmaceuticalsChiralpak AD[130]
Enantiomeric pharmaceuticalsChiralcel OD, Chiralcel OJ and Chiralcel AD[131]
A set of 111 chiral compoundsChirobiotic T, Chirobiotic TAG and Chirobiotic R[132]
Albendazole sulfoxideChiralpak AD and Chiralcel OD[133]
Triadimefon and triadimenolChiralpak AD[134]
Albendazole sulfoxideChiralpak AD[135]
Omeprazole and several related benzimidazolesChiralpak AD[136]
Triazole pesticidesChiralpak AD[137]
Enantiomeric pharmaceuticalsChirlapak AD and AS, and Chiralcel OD and OJ[138]
Antiulcer drugsChiralpak AD[139]
NaproxenKromasil CHI-TBB[140]
WarfarinChiralpak AD-H[141]
Chiral sulfoxidesChiralpak AD[142]
Antimycotic azole drugsChiralpak AD[143]
Nutlin-3Chiralcel OD, Chiralcel AD, Chiralcel OJ, Chirobiotic T, Chirobiotic V[144]
Phospine-Containing α-Amino Acid DerivativesLux Cellulose-1 and -2[145]
Acetamide intermediateChiralcel OD-H, Chiralpak AD, Lux Cellulose-2 and Lux Amylose-2[146]
Tris-(3,5-dimethylphenylcarbamate) of amyloseChiralcel OD-H and Chiralpak AD-H[147]
MianserinChiralcel OJ[148]
Chiral fluoro-oxoindole-type compoundsLux Cellulose-1, Lux Cellulose-
2 and Lux Amylose-2
[149]
FlutriafolChiralpak IA-3[150]
Enantiomeric pharmaceuticalsChiralpak IC and Chiralpak AD-3[151]
Troeger’s base, binaphthol, mandelic methylester, trans-stilbene oxide, flavanone and guaifenesineChiralcel AD-H and Chiralpak IC[152]
Table 9. Experimental studies of enantioresolutions applying HPLC Chromatography.
Table 9. Experimental studies of enantioresolutions applying HPLC Chromatography.
EnantiomersCSPEluentRef.
Antifungal chiral drugsPolysaccharide derivativesHexane-ethanol and hexane-2-propanol[133]
Fungicide EnantiomersAmylopectin Based Chiraln-hexane and isopropanol[144]
Oxazepam, lorazepam, and temazepamDerivatized cyclodextrin-bondedAcetonitrile[156]
1,4-DihydropyridinesVancomycinMethanol/acetic acid/TEA[157]
LinezolidAmylose basedHexane, 2-propanol and trifluoro acetic acid[158]
TangutorineChiralcel OD and Chiralpak ADn-hexane/2-propanol[159]
β-blockers(R)-1-naphthylglycine and 3,5-dinitrobenzoic acidn-hexane, 1,2-dichloroethane and methanol[160]
Tolterodine tartarateChiralcel OD-Hn-hexane and isopropyl[161]
1,4-dihydropyridinemonocarboxylic acidTert-butylcarbamoylquinineMethanol and ammonium acetate buffer[162]
Naringenin and other flavanonesChiralcel OD-H and Chiralpak AS-Hn-hexane/alcohol[163]
Piperidine-2,6-dione analoguesChiralpak IA and Chiralpak IBMethyl-tert-butyl ether-THF[164]
BambuterolChiralpak ADHexane/2-propanol[165]
β-LactamsCyclodextrin-Based ChiralIsopropanol-heptane[166]
Ruthenium(II) Polypyridyl ComplexesCyclodextrin ChiralMethanol and acetonitrile[167]
Chiral acids, bases, and amino acidsZwitterionic ion-exchange-typeAcetic acid, formic acid, diethylamine, and ammonium acetate[168]
10 β-adrenergic blockersCelluCoat columnn-heptane–ethanol–diethylamine[169]
Triazole FungicidesChrialcel OD and Chrialcel OJHexane/2-propanol[170]
2-arylpropionic
acid nonsteroidal anti-infl ammatory drugs
Hydroxypropyl-β-cyclodextriMethanol and NaH2PO4 buffer[171]
4 β-adrenergic blockersSPE-Chiraln-Heptane:ethanol:diethylamine[172]
OfloxacinIonic liquid-assisted ligand-exchangeMethanol/water[173]
Chiral PesticidesCellulose tris-(3,5-dimethylphenyl-carbamate)-coated chiralEthanol, n-propanol, iso-propanol, n-butanol, and iso-butanol[174]
Dihydropyridine derivativesPolysaccharide-based chiralFormic acid[175]
Arylpropionic acid derivativesChiralpak ADn-hexane modified either with 2-propanol or ethanol[176]
Illicit drugsCyclofructan-based and cyclobond I 2000 RSPHeptane with ethanol or isopropanol[177]
Ruthenium (II) Polypyridyl ComplexesCyclofructanAcetonitrile and methanol[178]
Table 10. Functions of the TMB sections.
Table 10. Functions of the TMB sections.
SectionFunctionIllustration
IThe more retained compound moves upward desorbed with the eluent, so that it leaves the system in the Extract stream (X). The eluent cleans the solid that is regenerated prior to being recycled in Section IV. Encyclopedia 02 00011 i058
IIThe more retained compound moves downward and is adsorbed on the solid whereas the less retained compound is desorbed with the eluent. This prevents contamination of the less retained compound in the Extract stream (X); the less retained compound moves upward to the Raffinate stream (R). Encyclopedia 02 00011 i059
IIIThe more retained compound moves downward adsorbed on the solid and the less retained compound is desorbed with the eluent. This prevents contamination in the Raffinate stream (R); the more retained compound moves downward to the Extract steam (X). Encyclopedia 02 00011 i060
IVThe less retained compound moves downward adsorbed with the solid flow, so that it leaves the system in the Raffinate stream (R). The solid phase cleans the liquid that is regenerated prior to be recycled to Section I. Encyclopedia 02 00011 i061
Table 11. Experimental studies of enantioresolutions applying SMB chromatography.
Table 11. Experimental studies of enantioresolutions applying SMB chromatography.
EnantiomersCSPEluentRef.
TramadolCHIRALPAK AD 202-propanol, hexane and diethylamine[182]
EMD 53986Cellulose-tri-(p-methyl-benzoate) and polymeric silica basedEthylacetate and ethanol[183]
Guaifenesin, aminoglutetimida, and formoterolCHIRALCEL OJ and CHIRALCEL ODHeptane/ethanol[184]
1,1′-bi-2-naphthol3,5-dinitrobenzoyl phenylglycine bonded to silica gelHeptane and isopropanol[185]
1,1′-bi-2-naphthol3,5-dinitrobenzoyl phenylglycine bonded to silica gelHeptane and isopropanol[186]
GuaifenesinCHIRALCEL ODHeptane/ethanol[187]
1-phenoxy-2-propanolCHIRALCEL ODHexane and isopropanol[188]
1,1′-bi-2-naphthol3,5-dinitrobenzoyl phenylglycine bonded to silica gelHeptane and isopropanol[189]
1-phenyl-1-propanolCHIRACEL OBEthyl acetate and heptane[190]
Trans-stilbene oxide and Tröger’s BaseCHIRALPAK AS and CHIRALPAK AS-VHexane/isopropanol[191]
N-carbobenzoxy-tert-leucine and N-Boc-tert-leucine-benzylesterCHIRALCEL OD and CHIRALPAK ADHeptane/ethanol and Heptane/2-propanol[192]
PhenylpropanolamineCHIRALPAK ADMethanol[193]
GuaifenesinCHIRALCEL ODEthanol[194]
BupivacaineKromasil CHI-TBBIso-propanol, hexane, and acetic acid[195]
DL-methionineEremomycinMethanol and water[196]
Tröger’s baseCHIRALPAK ADMethanol[197]
α-TetralolCHIRALPAK ADHeptane/2-propanol[198]
(RS,RS)-2-(2,4-difluorophenyl)butane-1,2,3-triolCHIRALCEL OJ and CHIRALPAK ADHexane, ethanol, and methanol[199]
Tröger’s baseCHIRALPAK ADEthanol[200]
Mandelic acidKromasil TBBHexane and ter-butylmethylether[201]
Tröger’s BaseCHIRALPAK ADEthanol[202]
Tröger’s BaseCHIRALPAK ADEthanol[203]
KetoprofenChiralpak AD1Ethanol, hexane and trifluoroacetic acid[204]
KetoprofenChiralpak AD1Ethanol, hexane and trifluoroacetic acid[205]
GuaifenesinCHIRALCEL ODHexane/ethanol[206]
PraziquantelChiralcel OZMethanol[207]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Santos, R.; Pontes, K.V.; Nogueira, I.B.R. Enantiomers and Their Resolution. Encyclopedia 2022, 2, 151-188. https://doi.org/10.3390/encyclopedia2010011

AMA Style

Santos R, Pontes KV, Nogueira IBR. Enantiomers and Their Resolution. Encyclopedia. 2022; 2(1):151-188. https://doi.org/10.3390/encyclopedia2010011

Chicago/Turabian Style

Santos, Rodrigo, Karen V. Pontes, and Idelfonso B. R. Nogueira. 2022. "Enantiomers and Their Resolution" Encyclopedia 2, no. 1: 151-188. https://doi.org/10.3390/encyclopedia2010011

Article Metrics

Back to TopTop