Review Selective Recognition of Amino Acids and by Small Supramolecular Receptors

Joana N. Martins , João Carlos Lima and Nuno Basílio *

LAQV—REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; [email protected] (J.N.M.); [email protected] (J.C.L.) * Correspondence: [email protected]; Tel.: +351-212-948-355

Abstract: To this day, the recognition and high affinity binding of biomolecules in water by synthetic receptors remains challenging, while the necessity for systems for their sensing, transport and modu- lation persists. This problematic is prevalent for the recognition of peptides, which not only have key roles in many biochemical pathways, as well as having pharmacological and biotechnological applications, but also frequently serve as models for the study of . Taking inspiration in nature and on the that occur between several receptors and sequences, many researchers have developed and applied a variety of different synthetic receptors, as is the case of macrocyclic compounds, molecular imprinted polymers, organometallic cages, among others, to bind amino acids, small peptides and proteins. In this critical review, we present and discuss selected examples of synthetic receptors for amino acids and peptides, with a greater focus on supramolecular receptors, which show great promise for the selective recognition of these biomolecules in physio- logical conditions. We decided to focus preferentially on small synthetic receptors (leaving out of this review high molecular weight polymeric systems) for which more detailed and accurate molec- ular level information regarding the main structural and thermodynamic features of the receptor   biomolecule assemblies is available.

Citation: Martins, J.N.; Lima, J.C.; Keywords: amino acids; peptides; host-guest systems; molecular recognition; supramolecu- Basílio, N. Selective Recognition of lar receptors Amino Acids and Peptides by Small Supramolecular Receptors. Molecules 2021, 26, 106. https://doi.org/ 10.3390/ molecules26010106 1. Introduction

Academic Editor: Roland Pieters Peptides are fundamental molecules with several biological functions, acting as neu- Received: 17 November 2020 rotransmitters, neuromodulators and hormones in numerous biochemical processes, such Accepted: 22 December 2020 as quorum sensing, immune response, pain and metabolism, to name a few [1,2]. Given Published: 28 December 2020 their utmost importance in different biochemical contexts, the discovery of high affinity and selective synthetic receptors for peptide recognition holds a strong potential for en- Publisher’s Note: MDPI stays neu- abling new therapeutic agents, such as inhibitors/activity modulators or vehicles for drug tral with regard to jurisdictional clai- delivery, and important molecular components for advanced biological and diagnostic ms in published maps and institutio- tools [3–7]. Furthermore, the discovery of the fundamental binding interactions controlling nal affiliations. the recognition of small peptide sequences is expected to pave the way towards the rational design of synthetic receptors for surface recognition. [3,6,8–10]. However, selective and high-affinity receptors for peptides are recognized to be very difficult to design and synthesize in a rational, bottom-up manner. Being biological Copyright: © 2020 by the authors. Li- censee MDPI, Basel, Switzerland. molecules, these targets present a difficulty just by the fact that their recognition should This article is an open access article be made in water, imposing practical difficulties related to the solubility of synthetic distributed under the terms and con- organic receptors, which may require further functionalization to improve their solubility ditions of the Creative Commons At- in aqueous solution. Moreover, the solvation of these biomolecules by water and the need to tribution (CC BY) license (https:// remove the solvent molecules and destabilize the solvation sphere also imposes an energetic creativecommons.org/licenses/by/ penalty that must be overcome upon binding. Directional and strong interactions based on 4.0/). hydrogen bonding, which are frequently and successfully employed in organic solvents,

Molecules 2021, 26, 106. https://doi.org/10.3390/molecules26010106 https://www.mdpi.com/journal/molecules Molecules 2021, 26, 106 2 of 41

are completely or partially neutralized in aqueous solutions. Therefore, it is necessary to explore other mechanisms, such as the and multiple interactions acting cooperatively to achieve the high affinity required to bind the peptide targets at relevant µM concentration or below [11]. In addition, the flexible, rather ill-defined structure of peptide sequences also increases the difficulties associated with the rational design of small receptors [6,12–14]. While the challenges associated to the factors enumerated above may be common to other biological targets, peptide recognition, in its completeness, requires receptors that are sequence-selective, discriminating not only the type of amino acids residues composing the peptide but also the order by which the they are arranged. Although general methods for high affinity peptide recognition by synthetic receptors are still missing, several research groups have reported encouraging results that show that it is possible to bind specific peptide sequences with simple receptors, such as macrocyclic cavitands, molecular clips, and tweezers or self-assembled coordination cages. Other strate- gies based on templating methods, such as molecular imprinted polymers and micelles, also showed very promising results. In this review we will cover selected examples of cavitand-based receptors that have been shown to recognize amino acids and peptides in aqueous solution and use these examples to emphasize the noncovalent interactions and geometric factors that control binding stability and selectivity of the different complexes. For this reason, we focus on small receptors for which this type of information can be extrapolated in a more straightforward way and potentially used for the rational optimization of current receptors and design of new ones. Then we will also provide some selected examples of biological and technological applications based on the binding properties of the receptors discussed here to illustrate the potential of these molecules and approaches.

2. The Biological Targets: Amino Acids Amino acids are the basic building blocks of small peptides and proteins. Even though they are important targets by themselves, the development of supramolecular receptors that can bind specific amino acids with high affinity and selectivity constitutes a first step towards the development of multivalent, sequence specific receptors for peptides. However, the selective binding of free amino acids already presents a difficult task since some side chains of amino acids are very similar to each other. In this section we have divided the 20 common amino acids into four categories: basic, aromatic, acid, and neutral non- aromatic amino acids and discussed their properties. In the next section, their recognition by selected supramolecular receptors will be further discussed.

2.1. Basic Amino Acids L-Lysine (Lys) and L-Arginine (Arg) are amino acids that play very important roles in biological systems—these are sites of recognition by the enzyme trypsin [15] and also for methylation in several proteins, as is the case of histones, the proteins involved in the structure of chromatin in the nucleus [16]; both take part in protein–protein binding [17], among other functions [18,19]. Arginine has a very important role in the membrane penetrating properties of antimicrobial peptides, as well [20]. The positive charges on these two amino acids make them easier targets for the development of receptors in water, many of them relying on the formation of several ionic interactions to obtain the receptor-analyte complexes. This is the case of p-sulfonatocalix[n] arene [21–23], some examples of molecular tweezers [24,25], carboxylatopillar [5]arene [26], all of which have negative charges, to form ionic bonds with the charged amines of Lys and Arg. L-Histidine (His), despite the presence of two nitrogen atoms, is near equilibrium between its deprotonated and protonated form, at physiological pH, due to its pKa of 6.5 [27]. Due to this, it can act as a proton donor or acceptor, depending on the environment near the residue [27]. Despite this property, histidine is usually paired with the other two Molecules 2021, 26, 106 3 of 41

basic amino acids. Regardless, it presents a lower charge density than the other two and thus His is difficult to target selectively by ionic interactions, in favor of Lys and Arg [26,28].

2.2. Aromatic Amino Acids Aromatic amino acids—L-tryptophan (Trp), L-tyrosine (Tyr) and L-phenylalanine (Phe)—are essential for protein function having important roles in protein–protein interac- tion [29] and electron transport in peptides [30] and proteins [31]. Moreover, these amino acids are precursors of several neurotransmitters—dopamine, serotonin, epinephrine, etc. [32]. Besides these functions, tryptophan also has a role in the ability of antimicrobial peptides to form pores on membranes, through the formation of cation–π bonds with arginine [33]. In terms of protein detection and analytical studies, aromatic amino acids are im- portant because they give proteins the characteristic 280 nm absorption band and are the only amino acids that present fluorescence, specially tryptophan, with a higher quantum yield than the other aromatic amino acids [34]. Many strategies for the recognition of these amino acids are based on hydrophobic, π–π and ion dipole interactions, making use not only of the aromatic group’s characteristic properties but also of the bigger size of these amino acids’ side chain [35–45].

2.3. Acidic Amino Acids The two anionic amino acids L-aspartate (Asp) and L-glutamate (Glu) exert several roles beyond their function as protein monomers. Glutamate is well known for its role as excitatory neurotransmitter in vertebrates, as well as precursor for gamma-Aminobutyric acid, GABA, an inhibitory neurotransmitter [46]. This amino acid is also the precursor to several other biomolecules and takes part in the process of elimination of ammonium, in the urea cycle [47]. Like glutamate, aspartate is also an important precursor in several biosynthetic pathways and takes part in the urea cycle [48]. However, this anionic amino acid is also essential in energetical processes in eukaryotic cells, being part of the malate- aspartate shuttle [49] and also having a role in the gluconeogenesis pathway [50]. Unlike cationic and aromatic amino acids, none of the most used macrocycles or supramolecular systems show particular affinity towards these two amino acids, without the need for further functionalization. Nevertheless, the functionalization of macrocyclic compounds with cationic groups has been demonstrated to yield potential receptors for these amino acids [51–53].

2.4. Neutral Non-Aromatic Amino Acids Neutral non-aromatic amino acids are difficult to target selectively in aqueous solution due to their high solvation, similar structures, reduced hydrophobic surface and the lack overall positive or negative charge [3,54]. However, these amino acids still have key roles in proteins and peptides, providing sites for glycosylation [55,56] and other post translational modifications [7]. They are also fundamental in ammonia regulation [57], anabolic and catabolic processes in eukaryotic cells (acting as precursors to other macronutrients or sources of energy) [50] and redox homeostasis [58]. Despite the difficulty of targeting these molecules, some receptors were developed for several of these amino acids. Many of these, however, only reach mM affinities and many were not tested in physiological conditions due to low solubilities in water [54]. Furthermore, many receptors that bind to these biomolecules with higher affinity present very low selectivity and are frequently able to bind to other amino acids with similar or even higher affinities [35,41,43,59]. Thiols are particularly reactive and so, the recognition and detection of L-Cysteine is more commonly based on the creation of sensors that react with this redox biomolecule by nucleophilic substitution [60]. In terms of supramolecular recognition most of the existing systems are based on metal complexes [3]. Molecules 2021, 26, 106 4 of 44

by nucleophilic substitution [60]. In terms of supramolecular recognition most of the ex- isting systems are based on metal complexes [3].

Molecules 2021 26 , , 106 3. Receptors for Amino Acids 4 of 41 has had a fruitful contribution to the development of new, multifunctional organic molecules. In terms of molecular recognition, a variety of differ- ent3. Receptors types of receptors for Amino have Acids been developed, from polymeric complex structures to both acyclicSupramolecular and cyclic small chemistry molecules has [61]. had aMacrocyclic fruitful contribution and semi-rigid to the developmentreceptors have of been new, gainingmultifunctional importance organic due molecules.to their simplicity In terms and, of molecular at the same recognition, time, high a varietyaffinityof and different selec- tivitytypes towards of receptors a myriad have been of guests, developed, given from not only polymeric by the complex complementary structures noncovalent to both acyclic in- teractionsand cyclic that small can molecules be formed, [61]. but Macrocyclic also by the and size semi-rigid complementarity receptors havebetween been host gaining and guestimportance [61,62]. due to their simplicity and, at the same time, high affinity and selectivity towards a myriad of guests, given not only by the complementary noncovalent interactions 3.1.that Macrocycles can be formed, but also by the size complementarity between host and guest [61,62]. Macrocyclic receptors are a class of molecules that have a well-defined structure for 3.1. Macrocycles the binding of a target guest. The cavity in these is often more hydrophobic, while the portalsMacrocyclic usually present receptors relatively are a class polar of or molecules charged thatmoieties have [62]. a well-defined In this section, structure a select for groupthe binding of examples of a target of macrocyclic guest. The receptors, cavity in thesewhich is have often shown more hydrophobic,to bind to amino while acids, the willportals be presented. usually present relatively polar or charged moieties [62]. In this section, a select group of examples of macrocyclic receptors, which have shown to bind to amino acids, 3.1.1.will be p-Sulfonatocalix[n]arenes presented. 3.1.1.p-Sulfonatocalix[n]arenep-Sulfonatocalix[n]arenes (SCn) macrocycles (Figure 1) were initially synthesized by Shinkai and co-workers to improve the solubility of calixarenes in water and allow for the p-Sulfonatocalix[n]arene (SCn) macrocycles (Figure1) were initially synthesized by binding and detection of not only organic compounds, but also inorganic ions in aqueous Shinkai and co-workers to improve the solubility of calixarenes in water and allow for the conditions [21,63–67]. Owing to their high water solubility (> 0.1 M) and low toxicity, SCn binding and detection of not only organic compounds, but also inorganic ions in aqueous macrocycles have been widely investigated for their pharmaceutical and biological appli- conditions [21,63–67]. Owing to their high water solubility (>0.1 M) and low toxicity, cations [21,68–74]. SCn macrocycles have been widely investigated for their pharmaceutical and biological applications [21,68–74].

Figure 1. Structures of the p-sulfonatocalix[n]arene receptors, SCn, and corresponding schematizations of the macrocycles. Figure 1. Structures of the p-sulfonatocalix[n]arene receptors, SCn, and corresponding schematizations of the macrocycles. In early studies by Douteau-Guével et al. and Selkti et al. the complexation of p- sulfonatocalix[n]areneIn early studies by (n Douteau-Guével = 4, 6 and 8) with et Lys al. and and Arg Selkti was et explored al. the complexation by NMR spectroscopy of p-sul- fonatocalix[n]areneand microcalorimetry (n in= 4, solution 6 and 8) and with by Lys X-ray and diffraction Arg was explored in the solid by stateNMR [ 21spectroscopy,75,76]. The and1H NMRmicrocalorimetry studies carried in solution out at different and by X-ray pH values diffraction showed in the that solid the state calixarenes [21,75,76]. do The not 1interactH NMR withstudies these carried amino out acids at different at pH 13pH due values to the showed deprotonation that the calixarenes of the amino do andnot interactguanidino with groups, these amino this last acids one at being pH 13 only due partially to the deprotonation deprotonated of under the amino these conditionsand guan- idino(pKa =groups, 13.2) [75 this]. Atlast more one acidicbeing pHonly conditions partially deprotonated (pH 5 and pH under 1), the these authors conditions also pointed (pKa =out 13.2) the [75]. weaker At more interactions acidic pH between conditions these (pH amino 5 and acids pH and 1), thethe largerauthors SC6 also and pointed SC8 hosts, out theattributing weaker interactions the low affinity between to the these more amino flexible acids structure and the of larger these hosts.SC6 and It must SC8 hosts, be noted, at- tributinghowever, the that low the affinity larger SC8 to the can more form 1:1flexible and 1:2structure host:guest of these complexes hosts. withIt must Arg be and noted, Lys. however,SC4 was investigatedthat the larger in SC8 more can detail form at1:1 pH and 1 1:2 and host:guest 5, showing complexes that the with receptor Arg and displays Lys. SC4higher was affinity investigated for both in aminomore detail acids at at pH pH 1 5and and 5, that, showing independently that the receptor of the pH,displays Arg higher(K = 1.7 affinity× 103 Mfor− 1bothat pH amino 5, K acids= 2.0 at× pH102 5M and−1 that,at pH independently 1) binds more of strongly the pH, thanArg ( LysK = − − 1.7(K ×= 10 6.03 M×−110 at2 pHM 5,1 Kat =pH 2.0 × 5, 10K2 =M 1.0−1 at× pH10 1)2 Mbinds1 at more pH strongly 1). It was than also Lys noted (K = 6.0 that × the102 presence of high concentrations of metal cations inhibits the formation of the complexes, a phenomenon which was generalized to other guest molecules in later studies and assigned to the competitive binding of the cations (including hydronium) with SCn [77–85]. The

complexation induced chemical shifts suggests that the terminal amino/guanidino groups in the side chain are included in the cavity of SC4 while the α-amino group remains outside of the cavity, most probably due to repulsion arising from the carboxylate group. Molecules 2021,, 2626,, 106106 5 of 44

M−−11 atat pHpH 5,5, K == 1.01.0 ×× 101022 MM−−11 atat pHpH 1).1). ItIt waswas alsoalso notednoted thatthat thethe presencepresence ofof highhigh concen-concen- trationstrations ofof metalmetal cationscations inhibitsinhibits thethe formationformation ofof thethe complexes,complexes, aa phenomenonphenomenon whichwhich was generalized to other guest molecules in laterlater studiesstudies andand assignedassigned toto thethe competitivecompetitive binding of the cations (including hydronium) with SCn [77–85]. The complexation in- Molecules 2021, 26, 106 duced chemical shifts suggests that the terminal amino/guanidino groups in the side5 chain of 41 are included in the cavity of SC4 while the α-amino-amino groupgroup remainsremains outsideoutside ofof thethe cavity,cavity, most probably due to repulsion arising from the carboxylate group. TheThe microcalorimetry microcalorimetry studies studies showed showed that that the the binding binding of of both both amino amino acids acids with with SC4 SC4 is enthalpy driven (ΔHLys = −14.4 kJ/mol and ΔHArg = −20.3 kJ/mol), with only slight differ- isis enthalpy enthalpy driven driven ((∆ΔHHLysLys == −−14.414.4 kJ/mol kJ/mol and and ΔH∆ArgH Arg= −20.3= − 20.3kJ/mol), kJ/mol), with only with slight only slightdiffer- ences in entropy (TΔSLys = 2.0 kJ/mol and TΔSArg = −2.1 kJ/mol), yet with two times higher differencesences in entropy in entropy (TΔSLys (T ∆= S2.0Lys kJ/mol= 2.0 kJ/moland TΔS andArg =T −∆2.1SArg kJ/mol),= −2.1 yet kJ/mol), with two yet times with higher two timesaffinity higher of the affinity receptor of the towards receptor Arg towards than Lys Arg (Table than Lys 1), (Tablebeing in 1), line being with in line the withprevious the previousNMR data NMR [21,75]. data This [21,75 study]. This also study confirmed also confirmed thatthat SC6SC6 that andand SC6 SC8SC8 and areare SC8 inin factfact are weakerweaker in fact weaker bindersbinders bindersforfor thesethese for twotwo these targetstargets two targets but,but, contrarycontrary but, contrary toto whatwhat to whatwaswas reportedreported was reported basedbased based onon NMRNMR on NMR titrations,titrations, titrations, alsoalso alsosuggests suggests that that the thebinding binding affinity affinity of SC4 of SC4 increases increases in more in more acidic acidic conditions. conditions. TheThe thermodynamic thermodynamic information information given given by theby the microcalorimetry microcalorimetry studies studies indicates indicates that thethatthat more thethe more favorablemore favorablefavorable enthalpy enthalpyenthalpy of the Argofof thethe complex ArgArg complexcomplex is probably isis probablyprobably due to the duedue formation toto thethe formationformation of cation– ofof πcation–and π–ππ andandinteractions π–π interactionsinteractions between itsbetweenbetween guanidinium itsits guanidiniumguanidinium group and groupgroup the aromatic andand thethe rings aromaticaromatic in the ringsrings cavity inin ofthethe the cavitycavity calixarene ofof thethe (Figurecalixarenecalixarene2). Moreover,(Figure(Figure 2).2). thisMoreover,Moreover, further thisthis stabilization furtherfurther stabilizationstabilization can justify thecancan negativejustifyjustify thethe valuenegative of entropy, value of seeing entropy, as arginine seeing as binds arginine deeper bindsbinds in the deeperdeeper cavity inin of thethe the cavitycavity receptor ofof [thethe71] receptorreceptor and so, with[71][71] and reducedand so,so, withwith degrees reducedreduced of freedom degreesdegrees[ ofof21 ].freedomfreedom The affinity [21].[21]. TheThe of this affinityaffinity receptor ofof thisthis towards receptorreceptorL-Histidine towardstowards L-- (His)Histidine was also(His) tested, was also however, tested, thehowever, affinity the obtained affinity was obtained two times was two lower times than lower towards than Lysinetowardstowards and LysineLysine near theandandaffinity nearnear thethe towards affinityaffinityneutral towardstowards amino neutralneutral acids aminoamino [28 ]. acidsacids In addition [28].[28]. InIn additionaddition to the studies toto thethe madestudies in made solution, in solution, the crystallographic the crystallographic structures structures of the Lysof the (Figure Lys (Figure3)[ 76] 3) and [76] Arg and [ 71Arg] complexes[71][71] complexescomplexes further furtherfurther support supportsupport the recognition thethe recognitionrecognition of these ofof thesethese amino aminoamino acids acidsacids through throughthrough the inclusionthethe inclusioninclusion of theirof their side side chains chains in the in the cavity cavity of the of the SC4 SC4 receptor. receptor.

FigureFigure 2. 2.Structure Structure of of the the complex complex formed formed between between SC4 SC4 and andL L-Arginine,-Arginine,-Arginine, adapted adaptedadapted from fromfrom [ [21,71]21[21,71],71] (left) (left)(left) andand a a schematization schematization ofof thethe cation–π interactionsinteractions thatthat that areare are establishedestablished established inin in thethe the former,former, former, betweenbetween between thethe the aromaticaromatic rings rings of of the the receptor receptor and and the the guanidinium guanidinium moiety moiety of of the the analyte. analyte.

Figure 3. Crystallographic Figurestructure 3. ofCrystallographic the SC4-L-Lysine:-Lysine: structure thethe analyteanalyte of the (in(inSC4- bold)bold)L-Lysine: complexescomplexes the inin analyte aa parallelparallel (in positionposition bold) complexes toto thethe ca-ca- in a lixarenes,lixarenes, asas itit cancan bebe observedobservedparallel byby positionthethe twotwo toptop to the complexes;complexes; calixarenes, another as it can possible be observed conformation by the twois perpendicular top complexes; to anotherthe disposi- possible conformation is perpendicular to the disposition of the calixarenes, interacting not only with the calixarene at the bottom but having its ε-amino group, NZ1, interacting externally with the sulfonate groups of the top layer receptors. Reprinted with permission from [76]. Copyright (2000) The Royal Society of Chemistry.

Several more recent studies also measured the affinity of these complexes, using indicator displacement assays, in different conditions, which are summarized in Table 1. Many of these yielded higher affinities of the cationic amino acids towards the receptor Molecules 2021, 26, 106 6 of 44

tion of the calixarenes, interacting not only with the calixarene at the bottom but having its ε-amino group, NZ1, interact- ing externally with the sulfonate groups of the top layer receptors. Reprinted with permission from [76]. Copyright (2000) The Royal Society of Chemistry.

Several more recent studies also measured the affinity of these complexes, using in- Molecules 2021, 26, 106 dicator displacement assays, in different conditions, which are summarized in Table6 of 1. 41 Many of these yielded higher affinities of the cationic amino acids towards the receptor SC4, due to this smaller macrocycle having a more rigid structure in comparison to SC6 andSC4, SC8. due These to this studies smaller also macrocycle indicate that, having near a morephysiological rigid structure conditions, in comparison the binding to may SC6 vary,and SC8. considering These studies that the also presence indicate of that,other near cationic physiological molecules, conditions, such as buffer the bindingcounterions, may canvary, interfere considering extensively that the with presence the binding of other [21,75,86–88]. cationic molecules, such as buffer counterions, can interfereThe higher extensively affinity of with SC4 the towards binding methylat [21,75,86ed–88 lysine]. is also worth noting, an im- portantThe post-translational higher affinity of modification. SC4 towards methylatedHof and co-workers lysine is alsoobserved worth that noting, the affinity an impor- of thetant 1:1 post-translational complexes formed modification. by SC4 with Hof lysine and and co-workers its methylated observed derivatives that the increases affinity of with the the1:1 complexesdegree of methylation formed by SC4 reaching with lysine ca. 70-fold and its selectivity methylated to derivatives trimethyllysine increases over with lysine the [89].degree Based of methylation on NMR titration reaching experiments, ca. 70-fold the selectivity authors observed to trimethyllysine that while over the lysine side [89]. chainBased is on accommodated NMR titration to experiments, the SC4 pocket the authorsin a “side-on” observed binding that while mode, the in lysine trimethyllysine side chain theis accommodated NMe3+ group is to deeply the SC4 included pocket in in the a “side-on” cavity suggesting binding mode,an optimal in trimethyllysine charge and shape the + complementaryNMe3 group is between deeply includedthis group in and the cavitythe cali suggestingxarene receptor. an optimal In a following charge and example, shape thecomplementary same research between group this explored group and the the mo calixarenenofunctionalization receptor. In aof following trisulfonated example, ca- lix[4]arenesthe same research (Figure group 4) to explored tune their the affinity monofunctionalization and selectivity towards of trisulfonated trimethyllysine. calix[4]arenes Alt- hough(Figure most4) to tuneof the their different affinity receptors and selectivity showed towards lower binding trimethyllysine. affinities, Although SC4-Ar, mostthe one of withthe different the unsubstituted receptors showed aromatic lower panel binding (Z = H) affinities, directly SC4-Ar, connected the oneto the with calixarene the unsubsti- rim, wastuted found aromatic to display panel (Z the = H)higher directly binding connected affinity to thetowards calixarene trimethyllysine, rim, was found almost to display 2-fold largerthe higher than bindingthe one previously affinity towards observed trimethyllysine, for SC4 and an almost improved 2-fold selectivity larger than of 150-fold the one overpreviously unmethylated observed lysine for SC4 (Table and an1). improvedThis is presumably selectivity due of 150-fold to its higher over unmethylated hydrophobic characterlysine (Table and 1). additional This is presumably CH-π interactions due to its between higher hydrophobic the lysine side character chain andand additional the extra aromaticCH-π interactions wall [90]. between the lysine side chain and the extra aromatic wall [90].

Figure 4. Structures of the upper rim monofunctionalized sulfonatocalix[4]arene receptors synthesized by Hof, where the structure with an added aromatic ring withwith Z = H, corresponds to SC4-Ar [[90].90].

3.1.2. Pillar[n]arenes 3.1.2. Pillar[n]arenes Pillararenes are a relatively new class of macrocycles, that only recently have begun to be Pillararenesapplied for the are binding a relatively of analytes new class in ofwater macrocycles, [91–93]. Carboxylatopillar that only recently [5]arene have begun (CP5) to wasbe applied one of forthe thefirst binding water-soluble of analytes pillararenes in water to [91 be–93 synthesized]. Carboxylatopillar [91] and recently [5]arene it (CP5) has beenwas oneapplied of the as firsta receptor water-soluble for cationic pillararenes organic molecules. to be synthesized Similar to [91 SCn,] and these recently molecules it has presentbeen applied a rigid as electron a receptor rich for ca cationicvity lined organic with anionic molecules. groups Similar on both to SCn, sides these of moleculesthe cavity (Figurepresent 5) a rigid[26,94]. electron rich cavity lined with anionic groups on both sides of the cavity (Figure5)[26,94]. In line with its ability to bind positively charged guest molecules, CP5 was demon- strated to selectively bind basic amino acids, i.e., L-lysine, L-arginine and L-histidine, with mM affinity (Table 1). In contrast with SC4, the biomolecules penetrate completely into the cavity, with the amine group of the chiral center being stabilized by ionic interactions at one of the entrances, and the amine in the side group of the amino acid being stabilized by the opposite side. Although these interactions have a bigger role in stabilizing these complexes—as suggested by the selectivity towards cationic amino acids observed in the study—the hydrophobic effect also takes an important role, namely between the CP5 cavity and the aliphatic chains of the amino acids. Furthermore, CP5 has an even higher Molecules 2021, 26, 106 7 of 41

affinity towards arginine over the other two cationic amino acids, due to the presence of π π Molecules 2021, 26, 106 the guanidinium moiety. This group allows for cation– interactions with the orbitals7 of 44 of the cavity of the pillararene, as well as the possibility to form several H-bonds with the receptor due to the conjugation between the two nitrogen atoms [26,94].

OR OR OR

RO RO

RO OR RO

OR OR

R = Bodipy, N N N N B F N O F C, O PDA6 FigureFigure 5. 5. StructureStructure of of the the Pillar[5]arene Pillar[5]arene receptors, receptors, P5-Bodipy P5-Bodipy [ [95],95], carboxylatopillar[5]arene, carboxylatopillar[5]arene, C-P5 C-P5 [26], [26], and and dodecaamine dodecaamine pillarpillar [6]arene, [6]arene, PDA6 PDA6 [51]. [51].

InAnother line with example its ability of ato pillararenebind positively receptor, charged is dodecaamine guest molecules, pillar CP5 [6]arene, was demon- PDA6, stratedwhich hasto selectively 6 amine groups bind basic at each amino of its acids, entrances i.e., L (Figure-lysine,5 L)-arginine and has been and shownL-histidine, to bind with to mMacidic affinity amino (Table acids [1).51]. In The contrast principles with behind SC4, the the biomolecules affinity of PDA6 penetrate towards completely these two amino into theacids cavity, is the with same the as amine for CP5—the group of aminethe chiral groups center are being positively stabilized charged by ionic at physiological interactions atpH one and of the they entrances, can establish and the ionic amine interactions in the side with group the twoof the carboxylic amino acid groups being of stabilized both Glu byand the Asp. opposite Although side. thisAlthough is the mainthese driving interactio forcens have of the a bindingbigger role (with in affinitystabilizing in the theseµM complexes—asrange) the complex suggested can be by further the selectivity stabilized towards by cation– cationicπ interactions amino acids between observed the in cavity the study—theof the pillararene hydrophobic and the effect amine also group takes ofan the important amino acids.role, namely This not between only adds the attractiveCP5 cav- ityinteraction and the aliphatic between chains the molecules, of the amino but also acids. counterbalance Furthermore, the CP5 electrostatic has an even repulsion higher thataf- finitycould towards be stablished arginine between over the the other cationic two regionscationic of amino both receptoracids, due and to analytethe presence [51]. of the guanidiniumPillararenes moiety. can beThis applied group for allows the recognition for cation– ofπneutral interactions amino with acids the as well.π orbitals This hasof thebeen cavity explored of the by pillararene, Guler and co-workersas well as the [95 ],possibility who developed to form a pillar[5]arene-basedseveral H-bonds with sensor, the receptorP5-Bodipy, due which to the presentedconjugation high between selectivity the two towards nitrogenL-Asparagine. atoms [26,94].L-Asn is an analogue of L-Aspartate,Another example with an of amide a pillararene group in itsrecept sideor, chain is dodecaamine instead of a carboxylate pillar [6]arene, group. PDA6, Due to whichits zwitterionic has 6 amine character groups and at each neutral of its side entrances chain, the (Figure influence 5) and of has electrostatic been shown interactions to bind tobetween acidic amino receptor acids and [51]. analyte The principles on the receptor’s behind the selectivity affinity of is PDA6 negligible, towards being these mostly two aminodefined acids by theis the hydrogen same as bonds, for CP5—the hydrophobic amine and groups Van are der positively Waals interactions charged at [54 physio-,95]. P5- logicalBodipy pH is and composed they can by establish a pillar[5]arene ionic interactions macrocycle, with decoratedthe two carboxylic with BODIPY groups moieties of both Glu(Figure and5 Asp.). This Although receptor this detected is the Lmain-Asn driving selectively force by of fluorescence the binding spectroscopy, (with affinity showing in the 1 μanM increaserange) the in complex the Bodipy can fluorescence.be further stabilizedH-NMR by elucidated cation–π interactions the types of between interactions the cavitypresent, of showingthe pillararene that L-Asn and isthe inserted amine ingroup the pillararene of the amino cavity, acids. being This stabilized not only by adds several at- tractivetypes of electrostatic between interactions, the molecules, but with Vanbut also der Waalscounterbalance interactions the and electrostatic hydrogen repul- bonds sionbeing that the could most be relevant stablished for the between selectivity the ofcati theonic receptor. regions of both receptor and analyte [51].3.1.3. Cucurbit[n]urils Pillararenes can be applied for the recognition of neutral amino acids as well. This Cucurbit[n]urils (CB[n]) are macrocycles composed of n glycoluril units, that, owing has been explored by Guler and co-workers [95], who developed a pillar[5]arene-based to their outstanding binding properties, are largely applied for the recognition of several or- sensor, P5-Bodipy, which presented high selectivity towards L-Asparagine. L-Asn is an ganic molecules and biological analytes (Figure6)[ 44,96–100]. Thanks to their hydrophobic analogue of L-Aspartate, with an amide group in its side chain instead of a carboxylate group. Due to its zwitterionic character and neutral side chain, the influence of electro- static interactions between receptor and analyte on the receptor’s selectivity is negligible, being mostly defined by the hydrogen bonds, hydrophobic and Van der Waals interac- tions [54,95]. P5-Bodipy is composed by a pillar[5]arene macrocycle, decorated with BOD-

Molecules 2021, 26, 106 8 of 44

IPY moieties (Figure 5). This receptor detected L-Asn selectively by fluorescence spectros- copy, showing an increase in the Bodipy fluorescence. 1H-NMR elucidated the types of interactions present, showing that L-Asn is inserted in the pillararene cavity, being stabi- lized by several types of electrostatic interactions, but with Van der Waals interactions and hydrogen bonds being the most relevant for the selectivity of the receptor.

3.1.3. Cucurbit[n]urils Molecules 2021 26 , , 106 Cucurbit[n]urils (CB[n]) are macrocycles composed of n glycoluril units, that, 8owing of 41 to their outstanding binding properties, are largely applied for the recognition of several organic molecules and biological analytes (Figure 6) [44,96–100]. Thanks to their hydro- cavityphobic and cavity highly and electronegative highly electronegative carbonyl portals,carbonyl these portals, macrocyclic these macrocyclic containers arecontainers partic- ularlyare particularly suitable to suitable bind positively to bind chargedpositively and charged hydrophobic and hydrophobic guests. Buschmann guests. Buschmann et al. [35] performedet al. [35] performed earlier studies earlier on studies the complexation on the complexation of amino acidsof amino with acids CB6 whichwith CB6 showed which mMshowed affinity mM towards affinity thetowards Phe, Gly,the Phe, Ala, Gly, and Ala, Val, and in aqueous Val, in aqueous formic acidformic 50%( acidv/ 50%(v). Nauv/v). andNau co-workers and co-workers reported reported the complexation the complexation of lysine, of arginine lysine, arginine and histidine and histidine by CB7 with by CB7 all threewith aminoall three acids amino showing acids showing moderate moderate affinity inaffinity the mM in the range mM [45 range]. Later [45]. on, Later Macartney on, Mac- etartney al. investigated et al. investigated the recognition the recognition of lysine, of arginine lysine, arginine and their and methylated their methylated derivatives deriva- by CB6tives and by CB7CB6 [and101 ].CB7 Noteworthy, [101]. Noteworthy, the affinity the increases affinity increases significantly significantly upon lysine upon methy- lysine lation,methylation, reaching reaching a 3500-fold a 3500-fold selectivity selectivity for trimethyllysine for trimethyllysine over lysineover lysine as a result as a result of the of goodthe good complementarity complementarity of CB7 of and CB7 trimethylalkylammonium and trimethylalkylammonium motifs.On motifs. the other On the hand, other at neutralhand, pH,at neutral lysine seemspH, lysine to bind seems only to in bind the CB6 only portal, in the forming CB6 portal, an exclusion forming complex an exclusion that, nevertheless,complex that, is nevertheless, more stable thanis more the stable one formed than the with one the formed larger with CB7 the (1.1 larger× 10 4CB7M− 1(1.1vs. × − 5.210×4 M10−12 vs.M 5.21). × A 10 subsequent2 M−1). A subsequent systematic studysystematic on the study formation on the of formation host–guest of complexes host–guest betweencomplexes CB7 between and different CB7 and amino different acids revealedamino acid thats revealed their stability that increasestheir stability under increases acidic conditionsunder acidic [102 conditions]. The differences [102]. The in associationdifferences constantsin association obtained constants at different obtained pH at values differ- areent particularly pH values are relevant particularly for lysine relevant and argininefor lysine which and arginine show ca. which 3 order show of ca. magnitude 3 order of 2 −1 5 −1 increasesmagnitude from increases≈10 M fromat ≈ neutral102 M−1 pHat neutral to ≈10 pHM to at≈10 pH5 M 2.−1 Thisat pH observation 2. This observation puts in evidenceputs in evidence the repulsive the repulsive interactions interactions established established between thebetween carbonyl the portalscarbonyl of portals the receptor of the andreceptor carboxylate and carboxylate group of the group amino of acid.the amino This destabilizing acid. This destabilizing factor is more factor relevant is more in these rele- twovant cases, in these most two probably cases, most due toprobably the fact due that to their the fact side-chain that their must side-chain penetrate must deeply penetrate into thedeeply CB7 cavityinto the for CB7 optimal cavity binding for optimal interactions, binding interactions, which are counteracted which are counteracted by the negatively by the chargednegatively carboxylate charged groupscarboxylate at neutral groups or at slightly neutral acid or slightly conditions acid [102 conditions]. [102].

= = =

FigureFigure 6. 6.Structure Structure of of the the Cucurbit[n]uril Cucurbit[n]uril (CBn) (CBn) receptors receptors and and corresponding corresponding schemes schemes that that will will be be used used for for illustration illustration purposespurposes throughout throughout this this review. review.

DespiteDespite being being investigated investigated for for their their affinity affinity towards towards basic basic amino amino acids, acids, CB7, CB7, CB8 CB8 and and otherother CB[n] CB[n] derivatives derivatives were were found found to to be be more more successful successful at at targeting targeting aromatic aromatic amino amino acidsacids in in biologically biologically relevant relevant conditions conditions [ 44[44,102].,102]. Among Among the the different different CB[n] CB[n] homologues, homologues, CB6CB6 was was shown shown to to display display weak weak affinity affinity towards towards aromatic aromatic amino amino acids acids probably probably due todue the to small size of its cavity (Figure7e), while the ones with more than 6 glycoluril units have a the small size of its cavity (Figure 7e), while the ones with more than 6 glycoluril units large enough cavity to accommodate the aromatic side chain in its interior [36–39,44,45]. have a large enough cavity to accommodate the aromatic side chain in its interior [36– This is the case of both CB7 and CB8 that were reported to, respectively, form 1:1 complexes and 1:1/1:2 complexes with aromatic amino acids (Figure7a,c,d) [36–39,45]. CB7, for example, was shown to be selective for aromatic amino acids at pH 7 (see Table 1) displaying higher affinity for L-phenylalanine with binding constants in the order 5 −1 6 −1 of 10 M –10 M depending on the medium conditions, in particular on the presence of salts [102–104]. Amongst the different aromatic amino acids, CB7 is selective for Phe by a factor of approximately 10 over Tyr and by a factor of 100 over Trp [102]. Most thermody- namic studies show that the complexation process is enthalpy driven in great part due to the displacement of high energy water molecules from the cavity of the host [102,105–108]. This hydrophobic effect can be complemented by attractive ion-dipole interactions between the protonated amino group of the biomolecules and the carbonyl group of the cucurbituril (Figure7)[ 35,44,109]. In the case of the binding of aromatic amino acids by CB7, the process Molecules 2021, 26, 106 9 of 44

39,44,45]. This is the case of both CB7 and CB8 that were reported to, respectively, form 1:1 complexes and 1:1/1:2 complexes with aromatic amino acids (Figure 7a,c,d) [36–39,45]. CB7, for example, was shown to be selective for aromatic amino acids at pH 7 (see Table 1) displaying higher affinity for L-phenylalanine with binding constants in the order of 105 M−1–106 M−1 depending on the medium conditions, in particular on the presence of salts [102–104]. Amongst the different aromatic amino acids, CB7 is selective for Phe by a factor of approximately 10 over Tyr and by a factor of 100 over Trp [102]. Most thermody- namic studies show that the complexation process is enthalpy driven in great part due to the displacement of high energy water molecules from the cavity of the host [102,105– 108]. This hydrophobic effect can be complemented by attractive ion-dipole interactions between the protonated amino group of the biomolecules and the carbonyl group of the cucurbituril (Figure 7) [35,44,109]. In the case of the binding of aromatic amino acids by CB7, the process is also enthalpy driven with unfavorable negative entropy contributions. The enthalpic gain is only slightly higher for phenylalanine with respect to tyrosine and tryptophan, but the entropic loss is very low in first case (TΔS = −0.6 kJ.mol−1) becoming more important for tyrosine (TΔS = −3.7 kJ.mol−1) and tryptophan (TΔS = −11.3 kJ.mol−1) [102]. On the basis of these thermodynamic parameters, the observed selectivity trend for Molecules 2021, 26, 106 recognition of aromatic amino acids by CB7 was explained by taking into account9 ofthe 41 higher hydrophobic character of phenylalanine comparatively with tyrosine and the re- stricted motion of the indole side chain of tryptophan inside the CB7 cavity [102]. CB8, on the other hand, binds aromatic amino acids with 1:1 (Figure 7a) and 1:2 (Fig- is also enthalpy driven with unfavorable negative entropy contributions. The enthalpic ure 7c) stoichiometry, as mentioned above [110]. The ITC binding studies performed on gain is only slightly higher for phenylalanine with respect to tyrosine and tryptophan, but thesethe entropic systems loss did is not very allow low the in firstdetermination case (T∆S =of− the0.6 stepwise kJ.mol−1 binding) becoming constants more importantand ther- modynamicfor tyrosine parameters (T∆S = −3.7 for kJ.mol the 1:1−1 )and and 1:2 tryptophan complexes(T in∆ Ssepa= −rate11.3 which kJ.mol precludes−1) [102]. a On more the inbasis depth of these thermodynamic thermodynamic analysis. parameters, Nevertheless, the observed based selectivityon the obtained trend for overall recognition binding of constantsaromatic amino(i.e., K acids1:1K1:2) byone CB7 can was conclude explained that by CB8 taking is also into more account selective the higher for phenylalanine, hydrophobic butcharacter in contrast of phenylalanine with CB7, tryptophan comparatively is comp withlexed tyrosine with and only the slight restricted lower motionaffinity of than the phenylalanineindole side chain while of tryptophanthe affinity insideof tyrosine the CB7 seems cavity to be [102 too]. low to be measured [110].

(a) (b) (c) (d) (e)

Figure 7. Examples of the CB[n] complexes: (a) CB8’s complex with one Phe molecule; (b) CB8’s heteroternary complex Figurewith Trp 7. andExamples Methyl of Viologen; the CB[n (c] )complexes: CB8’s homoternary (a) CB8′s complex complex with with two one Phe Phe molecules; molecule; ( d(b)) CB7 CB8 complex′s heteroternary with Phe; complex (e) CB6 withcomplex Trp withand Methyl Phe. Viologen; (c) CB8′s homoternary complex with two Phe molecules; (d) CB7 complex with Phe; (e) CB6 complex with Phe. CB8, on the other hand, binds aromatic amino acids with 1:1 (Figure7a) and 1:2 (FigureThe7c) larger stoichiometry, cavity of CB8 as mentioned allows the above binding [ 110 of]. amino The ITC acids binding in the studiespresence performed of auxiliary on gueststhese systems based on did electron not allow deficient the determination organic mole ofcules, the stepwise such as methyl binding viologen constants (MV) and [37], ther- 2,7-dimethyl-modynamic parameters diazaphenanthrenium for the 1:1 and (DPT) 1:2 complexes [38] and tetramethyl in separate whichbenzobis(imidazolium) precludes a more (MBBI)in depth [39]. thermodynamic The preformed analysis. 1:1 CB8:auxiliary Nevertheless, guest based complex on the can obtained be viewed overall as a bindingnew re- ceptorconstants capable (i.e., Kof1:1 formingK1:2) one 1:1:1 can heteroternary conclude that complexes CB8 is also with more electron selective rich for molecules phenylalanine, that, inbut some in contrast cases, does with not CB7, bind tryptophan significantly is complexed to CB8 alone with (Figure only 7b) slight [37–39]. lower Interestingly, affinity than phenylalanine while the affinity of tyrosine seems to be too low to be measured [110]. The larger cavity of CB8 allows the binding of amino acids in the presence of auxiliary guests based on electron deficient organic molecules, such as methyl viologen (MV) [37], 2,7-dimethyl- diazaphenanthrenium (DPT) [38] and tetramethyl benzobis(imidazolium) (MBBI) [39]. The preformed 1:1 CB8:auxiliary guest complex can be viewed as a new receptor capable of forming 1:1:1 heteroternary complexes with electron rich molecules that, in some cases, does not bind significantly to CB8 alone (Figure7b) [ 37–39]. Interestingly, the CB8:MV complex was shown to form 1:1:1 complexes only with the aromatic amino acids (except histidine). In the 1:2 complex, phenylalanine establishes strong contacts with the second Phe residue inside the cavity, as well as with CB8 itself [110]. Differently from what is observed for CB8 in the absence of auxiliary guest, the CB8:MV supramolecular receptor shows higher affinity for tryptophan (4.3 × 104 M−1) than for phenylalanine and tyrosine [110]. This selectivity was attributed to the charge transfer interactions between MV and the indole side chain of this amino acid inside the CB8 cavity. The nature of the auxiliary guest may also influence the recognition of the second guest. CB8:MBBI displays binding affinities for tryptophan (3.4 × 104 M−1)[39] similar to the one observed for CB8:MV, while CB8:DPT forms 1:1:1 complexes with this amino acid that are one order of magnitude more stable (4.2 × 105 M−1)[38]. Cucurbiturils can also be modified covalently to enhance their affinity towards amino acids. One example of this is the work done by Isaacs and co-workers, which incorporate Molecules 2021, 26, 106 10 of 44

the CB8:MV complex was shown to form 1:1:1 complexes only with the aromatic amino acids (except histidine). In the 1:2 complex, phenylalanine establishes strong contacts with the second Phe residue inside the cavity, as well as with CB8 itself [110]. Differently from what is observed for CB8 in the absence of auxiliary guest, the CB8:MV supramolecular receptor shows higher affinity for tryptophan (4.3 × 104 M−1) than for phenylalanine and tyrosine [110]. This selectivity was attributed to the charge transfer interactions between MV and the indole side chain of this amino acid inside the CB8 cavity. The nature of the auxiliary guest may also influence the recognition of the second guest. CB8:MBBI displays binding affinities for tryptophan (3.4 × 104 M−1) [39] similar to the one observed for

Molecules 2021, 26, 106 CB8:MV, while CB8:DPT forms 1:1:1 complexes with this amino acid that are one order10 of 41of magnitude more stable (4.2 × 105 M−1) [38]. Cucurbiturils can also be modified covalently to enhance their affinity towards amino acids. One example of this is the work done by Isaacs and co-workers, which in- twocorporate aromatic two rings aromatic within rings the CB6 within macrocycle. the CB6 macrocycle. This study yielded This study a water-soluble yielded a water-sol- receptor withuble anreceptor elongated with cavity,an elongated CB6Ar cavity, (Figure CB6Ar8), which (Figure is able 8), which to form is ableπ–π tointeractions form π–π withinter- includedactions with analytes, included while analytes, maintaining while CB[n]’smaintain capacitying CB[n]’s to form capacity ion-dipole to form interactions ion-dipole atin- theteractions macrocycle at the entrances. macrocycle The entrances. addition The of theadditionπ–π interaction of the π–π rendersinteraction a receptor renders with a re- 6 −1 micromolarceptor with affinitymicromolar towards affinity tryptophan towards ( Ktryptophana = 3.2 × 10 (KMa = 3.2) and × 10 ca.6 M 100-fold−1) and ca. selectivity 100-fold forselectivity this amino for acidthis amino over phenylalanine acid over phenylalanine and tyrosine and [40 tyrosine]. [40].

CB6Ar =

FigureFigure 8. 8.Structure Structure ofof thethe modifiedmodified CB6CB6 receptor,receptor, CB6Ar, CB6Ar, and and a a schematization schematization of of the the complex complex formed formed between between the the receptor receptor andandL L-Trp,-Trp, with with the the establishment establishment of of two twoπ π–π–πinteractions interactions [40 [40].].

3.1.4. Cyclodextrins 3.1.4. Cyclodextrins Cyclodextrins (CD) are macrocycles formed by glucose units that can bind a variety of differentCyclodextrins small molecules. (CD) are macrocycles The most commo formedn byones glucose are those units composed that can bind of 6, a 7, variety and 8 ofglucose different units, small corresponding molecules. respectively The most common to α-CD, ones β- CD, are and those γ-CD composed (Figure of9) [111]. 6, 7, and Their 8 glucosehost–guest units, complexes corresponding with respectivelybiomolecules to areα-CD, usuallyβ- CD, low and inγ specificity,-CD (Figure but9)[ are111 ].slightly Their Molecules 2021, 26, 106 host–guest complexes with biomolecules are usually low in specificity, but11 are of 44 slightly more stable with aromatic or small, less polar molecules moreα stable-cyclodextrin with aromatic was shown or small, to bind less L polar-Phe moleculesin a 2003 study by Buschmann et al., but the binding is rather unspecific, only driven by hydrophobic interactions, yielding the same affinities for this amino acid and for a series of other less polar amino acids, as well as other non-polar analytes tested [35].

Figure 9. Structure of the receptors α-Cyclodextrin (α-CD) and β-Cyclodextrin (β-CD) and correscheme 41. recently synthe- − sized aFigure single 9. substitutedStructure of analoguethe receptors of this α-Cyclodextrin macrocycle, (αβ-CD)-CDU, and with β-Cyclodextrin high affinity (β towards-CD) and Lcorrescheme-tryptophan 41.(K recently= 5.2 × syn-104 M 1), thanksthesized to the addeda single “arm” substituted with analogue hydrogen of bondingthis macrocycle, groups β as-CDU, well with as a high hydrophobic affinity towards region L (Figure-tryptophan 10). ( DespiteK = 5.2 × the104 larger M−1), thanks to the added “arm” with hydrogen bonding groups as well as a hydrophobic region (Figure 10). Despite the cavity of β-cyclodextrin, structural studies showed that there was no inclusion complex, contrary to what is usual for these larger cavity of β-cyclodextrin, structural studies showed that there was no inclusion complex, contrary to what is usual systems,for these but insteadsystems,the but complexinstead the was complex formed was at formed the entrance at the entrance of the of receptor. the receptor. This This binding binding was was shown shown to to happenhappen in 2:1 host:guestin 2:1 host:guest stoichiometry stoichiometry with the with receptor the receptor forming forming a dimer, a dimer, with with the the analyte analyte in in the the createdcreated pocket, pocket, being being stabilized stabilized by hydrophobicby hydrophobic interactions interactions and hydrogen and hydrogen bonds bonds [41]. [41].

Figure 10. Structure of the receptor β-CDxU (left) and structure of the dimer that is formed upon complexation of L-Phe (right), reprinted with permission from [41]. Copyright (2017) Elsevier.

3.1.5. Cavitands Cavitands is a term that encompasses several types of rigid macrocycles and it was first coined by Cram and co-workers in 1982 [112]. These constitute a class of receptors that present a “enforced cavity” where a small guest can bind, being the most widely studied ones those based on resorcinarenes macrocycles [62,112]. The Dalcanale group has reported several cavitand derivatives (Figure 11) that were found to recognize amino ac- ids and amino acid metabolites [113,114], and were able to obtain the crystallographic structure of the complexes between some of these cavitand receptors and all basic amino acids, Arg, Lys, and His. The 1:2 complexes were shown to be mainly stabilized by direct and water-mediated hydrogen bonds between a guest molecule and two host molecules [114]. These receptors were also shown to be particularly adequate to selectively recognize sarcosine, a potential prostate cancer biomarker, over glycine in water and in biological fluids [113]. The crystal structures of the complexes showed that while glycine methyl ether is localized at the receptor’s portal, sarcosine forms an inclusion complex with the N-methyl group deeply included in the cavity. This structural arrangement allows the stabilization of the complex by a combination of non-covalent interactions that include CH-π contacts between the N-methyl residue and the aromatic pocket, along with cation-

Molecules 2021, 26, 106 11 of 44

Figure 9. Structure of the receptors α-Cyclodextrin (α-CD) and β-Cyclodextrin (β-CD) and correscheme 41. recently syn- thesized a single substituted analogue of this macrocycle, β-CDU, with high affinity towards L-tryptophan (K = 5.2 × 104 M−1), thanks to the added “arm” with hydrogen bonding groups as well as a hydrophobic region (Figure 10). Despite the larger cavity of β-cyclodextrin, structural studies showed that there was no inclusion complex, contrary to what is usual Moleculesfor 2021these, 26 systems,, 106 but instead the complex was formed at the entrance of the receptor. This binding was shown to happen11 of 41 in 2:1 host:guest stoichiometry with the receptor forming a dimer, with the analyte in the created pocket, being stabilized by hydrophobic interactions and hydrogen bonds [41].

Figure 10. Structure of the receptor β-CDxU ( left) and structure of the dimer that is formedformed upon complexationcomplexation of L-Phe (right), reprinted with permission fromfrom [[41].41]. Copyright (2017) Elsevier.Elsevier.

3.1.5.α Cavitands-cyclodextrin was shown to bind L-Phe in a 2003 study by Buschmann et al., but the binding is rather unspecific, only driven by hydrophobic interactions, yielding the same Cavitands is a term that encompasses several types of rigid macrocycles and it was affinities for this amino acid and for a series of other less polar amino acids, as well as other first coined by Cram and co-workers in 1982 [112]. These constitute a class of receptors non-polar analytes tested [35]. that present a “enforced cavity” where a small guest can bind, being the most widely studied3.1.5. Cavitands ones those based on resorcinarenes macrocycles [62,112]. The Dalcanale group has reported several cavitand derivatives (Figure 11) that were found to recognize amino ac- Cavitands is a term that encompasses several types of rigid macrocycles and it was idsfirst and coined amino by Cramacid metabolites and co-workers [113,114], in 1982 and [112 were]. These able constitute to obtain a classthe crystallographic of receptors that structurepresent a of “enforced the complexes cavity” between where a some small of guest these can cavitand bind, being receptors the most and widelyall basic studied amino acids,ones those Arg, basedLys, and on resorcinarenesHis. The 1:2 complexes macrocycles were [62 shown,112]. The to be Dalcanale mainly stabilized group has by reported direct andseveral water-mediated cavitand derivatives hydrogen (Figure bonds 11between) that were a guest found molecule to recognize and two amino host molecules acids and [114].amino These acid metabolitesreceptors were [113 also,114 shown], and were to be able particularly to obtain adequate the crystallographic to selectively structure recognize of sarcosine,the complexes a potential between prostate some ofcancer these biomarker, cavitand receptors over glycine and allin water basic aminoand in acids,biological Arg, fluidsLys, and [113]. His. The The crystal 1:2 complexes structures were of shownthe complexes to be mainly showed stabilized that while by direct glycine and methyl water- ethermediated is localized hydrogen at the bonds receptor’s between portal, a guest sarcosine molecule forms and two an inclusion host molecules complex [114 with]. These the Nreceptors-methyl weregroup also deeply shown included to be particularly in the cavity adequate. This structural to selectively arrangement recognize sarcosine,allows the a stabilizationpotential prostate of thecancer complex biomarker, by a combination over glycine of non-covalent in water and interactions in biological that fluids include [113]. CH-Theπ crystal contacts structures between of the the N complexes-methyl residue showed and that the while aromatic glycine pocket, methyl along ether with is localized cation- at the receptor’s portal, sarcosine forms an inclusion complex with the N-methyl group deeply included in the cavity. This structural arrangement allows the stabilization of the complex by a combination of non-covalent interactions that include CH-π contacts between the N-methyl residue and the aromatic pocket, along with cation-dipole and hydrogen bonding between the ammonium and the phosphonate groups. The selective complexation of sarcosine in solution was confirmed by ITC experiments in methanol, by water:chloroform extraction and solubilization assays. The recognition of sarcosine in water and in urine was achieved by grafting the cavitands to a silicon surface. The authors showed that the receptor held its recognition properties at these interfaces and devised a sensor based on a luminescence indicator displacement assay using (9-anthrylmethyl) methyl ammonium chloride as probe. More recently, a comprehensive study on recognition of amino acids by tetraphos- phonate cavitands was performed to dissect the thermodynamic factors responsible for the observed selectivity of these receptors towards N-methyl amino acids [115]. The com- plexation was investigated both in methanol and in aqueous solutions, showing that the stability of the resulting complexes significantly decreases (almost 2 order of magnitude) in water. The ITC results demonstrated that while the association process is both enthalpy and entropy-driven in methanol, it becomes entropically unfavorable in water. The authors attributed this observation to the non-classical enthalpy driven hydrophobic effect and to the enhanced solvation of the complex in aqueous solution. Despite of the decreased affinity, the selectivity of these receptors toward N-methylated amino acids improves in aqueous solution resulting in their exclusive complexation. Molecules 2021, 26, 106 12 of 44

dipole and hydrogen bonding between the ammonium and the phosphonate groups. The selective complexation of sarcosine in solution was confirmed by ITC experiments in methanol, by water:chloroform extraction and solubilization assays. The recognition of sarcosine in water and in urine was achieved by grafting the cavitands to a silicon surface. The authors showed that the receptor held its recognition properties at these interfaces and devised a sensor based on a luminescence indicator displacement assay using (9-an- thrylmethyl) methyl ammonium chloride as probe. More recently, a comprehensive study on recognition of amino acids by tetraphos- phonate cavitands was performed to dissect the thermodynamic factors responsible for the observed selectivity of these receptors towards N-methyl amino acids [115]. The com- plexation was investigated both in methanol and in aqueous solutions, showing that the stability of the resulting complexes significantly decreases (almost 2 order of magnitude) in water. The ITC results demonstrated that while the association process is both enthalpy and entropy-driven in methanol, it becomes entropically unfavorable in water. The au- thors attributed this observation to the non-classical enthalpy driven hydrophobic effect

Molecules 2021, 26, 106 and to the enhanced solvation of the complex in aqueous solution. Despite of the de- 12 of 41 creased affinity, the selectivity of these receptors toward N-methylated amino acids im- proves in aqueous solution resulting in their exclusive complexation.

Figure 11.Figure General 11. General structure structure of the Dalcanale of the Dalcanale cavitand cavitand receptors, receptors, Cav[R1,R Cav[R2,R3] 1[113–115].,R2,R3][113 –115].

3.1.6. Other3.1.6. Macrocyclic Other Macrocyclic Receptors Receptors A variety ofA different variety of macrocycles different macrocycles have been haveused beenfor the used binding for the of bindinga myriad of of a myriad of molecules. molecules.Some less known Some lessexamples known that examples are only that now are being only nowapplied being in appliedbiomolecule in biomolecule recognitionrecognition or that present or that more present complex more complex molecular molecular structures structures are described are described below, below, show- showcasingcasing the versatility the versatility and diverse and diverse design design that macrocycles that macrocycles can present. can present. Early workEarly by Kaifer work et by al. Kaifer showed et al. that showed the tetracationic that the tetracationic cyclophane cyclophane cyclobis(para- cyclobis(paraquat- quat-p-phenylene),p-phenylene), CPQ, CPQ,commonly commonly known known as the asblue-box, the blue-box, forms forms charge charge transfer transfer com- complexes with aromatic amino acids in aqueous solution (Figure 12). This receptor is selective for plexes with aromatic amino acids in aqueous solution (Figure 12). This receptor is selec- tryptophan (K = 1.0 × 103 M−1) showing ca. 2 and >10 fold lower affinity for tyrosine tive for tryptophan (K = 1.0 × 103 M−1) showing ca. 2 and >10 fold lower affinity for tyrosine and phenylalanine, respectively [116]. Cao and co-workers more recently explored the and phenylalanine, respectively [116]. Cao and co-workers more recently explored the af- affinity of two distinct tetracationic cyclophane derivatives towards anionic and aromatic finity of two distinct tetracationic cyclophane derivatives towards anionic and aromatic biomolecules. The TeCPh, the cyclophane analogue with a narrower electron-deficient biomolecules. The TeCPh, the cyclophane analogue with a narrower electron-deficient cavity described in this study (Figure 12), showed a high specificity towards tryptophan cavity described in this study (Figure 12), showed a high specificity towards tryptophan Molecules 2021, 26, 106 (K = 1.2 × 103 M−1) in relation to the other amino acids, forming π–π bonds with13 of the 44 (K = 1.2 × 103 M−1) in relation to the other amino acids, forming π–π bonds with the elec- electron-rich aromatic side chain of the analytes, and possibly being further stabilized by tron-rich aromatic side chain of the analytes, and possibly being further stabilized by ionic ionic interactions with the carboxylate group of Trp [42]. interactions with the carboxylate group of Trp [42].

CPQ TeCPh

FigureFigure 12. 12. StructureStructure of of the the cyclophane cyclophane receptors receptors CPQ [116] [116] and TeCPh [[42].42].

AlfonsoAlfonso et et al. al. synthesized synthesized two two novel novel oxaazamacrocycles, oxaazamacrocycles, which which presented presented enantiose- enantiose- lectivelective binding binding with with both both aspartate aspartate and and glutam glutamate—OAate—OA (R,R) e OA (S,S,S,S) [53]. [53]. Only the latterlatter showed considerableconsiderable selectivity selectivity towards towards the the L isomersL isomers of theseof these anionic anionic biomolecules, biomole- cules,but with butrelatively with relatively low affinities. low affinities. The former The former showed showed higher higher affinity affinity towards towards the D-isomer the D- isomerof aspartate. of aspartate. This specificity This specificity is given is by given the positioningby the positioning of the positive of the positive groups groups and oxygen and oxygenon the receptoron the receptor in relation in relation to the analyteto the analyte that it that has it affinity has affinity towards towards (Figure (Figure 13). This13). Thisdistinct distinct approach approach in receptor in receptor design design allowed allowed for the for positioning the positioning of its of charged its charged and polar and polarmoieties moieties in proximity in proximity to guests’, to guests’, in a complementaryin a complementary manner. manner. The positiveThe positive ammonium ammo- nium groups turned towards the cavity of the receptor and the two oxygens, free to form two hydrogen bonds each, facing in the same general direction, is the ideal conformation for the binding of D-Ac-Asp to OA (R,R), allowing the establishment of ionic interactions and one without the need for many spatial rearrangements of the receptor [53].

OA (R,R) OA (S,S,S,S)

Figure 13. Structure of the oxaazamacrocycle receptors OA(R,R) and OA(S,S,S,S) [53].

Much like the previous examples, calixpyridinium, C4Pyr is a supramolecular sys- tem that has not been target of much attention in biomolecule binding applications (Figure 14). However, this macrocycle presents many similarities to calixarenes, with high solu- bility and with added cationic groups allowing for the creation of a binding site for small anionic biomolecules. Ren and co-workers tested this receptor for its affinity towards an- ionic amino acids—it showed to be highly selective for these, when compared to the rest of the proteinogenic amino acids, binding aspartate and glutamate (K ≈ 103 M−1) solely by ionic interactions [52].

Molecules 2021, 26, 106 13 of 44

CPQ TeCPh

Figure 12. Structure of the cyclophane receptors CPQ [116] and TeCPh [42].

Alfonso et al. synthesized two novel oxaazamacrocycles, which presented enantiose- lective binding with both aspartate and glutamate—OA (R,R) e OA (S,S,S,S) [53]. Only the latter showed considerable selectivity towards the L isomers of these anionic biomole- cules, but with relatively low affinities. The former showed higher affinity towards the D- isomer of aspartate. This specificity is given by the positioning of the positive groups and Molecules 2021, 26, 106 oxygen on the receptor in relation to the analyte that it has affinity towards13 of(Figure 41 13). This distinct approach in receptor design allowed for the positioning of its charged and polar moieties in proximity to guests’, in a complementary manner. The positive ammo- nium groups turned towards the cavity of the receptor and the two oxygens, free to form groups turnedtwo hydrogen towards bonds the cavity each, offacing the receptor in the same and general the two direction, oxygens, is free the to ideal form conformation two hydrogen bonds each, facing in the same general direction, is the ideal conformation for for the binding of D-Ac-Asp to OA (R,R), allowing the establishment of ionic interactions the bindingand ofoneD -hydrogenAc-Asp to bond OA (R,R), without allowing the need the for establishment many spatial of rearrangements ionic interactions of andthe receptor one hydrogen[53]. bond without the need for many spatial rearrangements of the receptor [53].

OA (R,R) OA (S,S,S,S)

FigureFigure 13. 13. StructureStructure of of the the oxaazamac oxaazamacrocyclerocycle receptors receptors OA(R,R OA (R,R)) and and OA(S,S,S,S) OA (S,S,S,S) [53]. [53 ]. Much like the previous examples, calixpyridinium, C4Pyr is a supramolecular system Much like the previous examples, calixpyridinium, C4Pyr is a supramolecular sys- that has not been target of much attention in biomolecule binding applications (Figure 14). tem that has not been target of much attention in biomolecule binding applications (Figure However, this macrocycle presents many similarities to calixarenes, with high solubility 14). However, this macrocycle presents many similarities to calixarenes, with high solu- and with added cationic groups allowing for the creation of a binding site for small anionic bility and with added cationic groups allowing for the creation of a binding site for small biomolecules. Ren and co-workers tested this receptor for its affinity towards anionic anionic biomolecules. Ren and co-workers tested this receptor for its affinity towards an- amino acids—it showed to be highly selective for these, when compared to the rest of the ionic amino acids—it showed to be highly selective for these, when compared to the rest Molecules 2021, 26, 106 proteinogenic amino acids, binding aspartate and glutamate (K ≈ 103 M−1) solely by14 ionic of 44 of the proteinogenic amino acids, binding aspartate and glutamate (K ≈ 103 M−1) solely by interactions [52]. ionic interactions [52].

FigureFigure 14. 14.Structure Structure of of the the receptor receptor calixpyridinium, calixpyridinium, C4Pyr. C4Pyr.

3.2.3.2. Open-ChainOpen-Chain ReceptorsReceptors DespiteDespite thethe greatgreat variety variety of of macrocycles macrocycles here here presented presented with with affinities affinities reaching reaching the the µμMM range,range, somesome open-chain receptors receptors can can presen presentt rigid rigid binding binding sites sites with with affinities affinities rival- ri- vallingling those those of of macrocycles macrocycles [24] [24.]. On On the the other other hand, hand, less less rigidrigid structuresstructures cancan bringbring other other advantages,advantages, e.g., e.g., the the flexibility flexibility to to bind bind larger larger guests guests [ 117[117].]. MolecularMolecular tweezerstweezers areare open-chain open-chain receptors receptors comprising comprising semi-rigid semi-rigid structures structures that that cancan bind bind complementary complementary analytes analytes [ 24[24,25,118],25,118]. Two. Two studies studies reported reported rigid rigid molecular molecular tweez- twee- erszers with with the the ability ability to to bind bind Arg Arg and and Lys, Lys, which which were were designed designed with with nine nine adjoining adjoining six-six- memberedmembered ringsrings andand a hydroquinone group group at at the the center. center. The The two two hydroxyl hydroxyl groups groups in the in thehydroquinone hydroquinone were were modified modified to to incorporat incorporatee different different anionic moieties—phosphonate,moieties—phosphonate, phosphate,phosphate, sulfate sulfate and and carboxylate—creatingcarboxylate—creating four four differently-substituteddifferently-substituted receptors, receptors, phos-phos- phonatephonate (MPnT),(MPnT), phosphatephosphate (MPT),(MPT), sulfatesulfate (MST)(MST) and and carboxylate carboxylate (MCT), (MCT), respectively respectively (Figure(Figure 15 15))[24 [24,25].,25]. These These receptors receptors showed showed different different affinities affinities towards towards Arg andArg Lys,and asLys, well as as the acylated and O-methylated modified residues, as shown in Table1. well as the acylated and O-methylated modified residues, as shown in Table 1.

R

R

MPnT: R=OP(CH3)O2 2 MPT: R=OPO3

MST: R=OSO3 MCT: R=OCH2CO2 Figure 15. Structure of the Molecular Tweezers reported in Schradder et al. functionalized with phosphonate (MPnT), phosphate (MPT), sulfate (MST), and carboxylate (MCT).

The first of these receptors to be tested was the phosphonate tweezer, which showed se- lectivity towards Lys, with even higher affinity towards its α-N/C-Protected form. NMR and ITC studies elucidated the interactions formed in these complexes where all the analytes tested showed a full incorporation of their side chain near the hydroquinone group (Figure 16). The binding process is aided by cation–π interactions between the positively charged groups of the amino acids and the electron-rich interior region of the tweezer, as well as by hydrophobic interactions between the alkyl chains of the analytes and the aromatic groups. However, the

Molecules 2021, 26, 106 14 of 44

Figure 14. Structure of the receptor calixpyridinium, C4Pyr.

3.2. Open-Chain Receptors Despite the great variety of macrocycles here presented with affinities reaching the μM range, some open-chain receptors can present rigid binding sites with affinities rival- ling those of macrocycles [24]. On the other hand, less rigid structures can bring other advantages, e.g., the flexibility to bind larger guests [117]. Molecular tweezers are open-chain receptors comprising semi-rigid structures that can bind complementary analytes [24,25,118]. Two studies reported rigid molecular twee- zers with the ability to bind Arg and Lys, which were designed with nine adjoining six- membered rings and a hydroquinone group at the center. The two hydroxyl groups in the hydroquinone were modified to incorporate different anionic moieties—phosphonate, phosphate, sulfate and carboxylate—creating four differently-substituted receptors, phos- Molecules 2021, 26, 106 phonate (MPnT), phosphate (MPT), sulfate (MST) and carboxylate (MCT), respectively14 of 41 (Figure 15) [24,25]. These receptors showed different affinities towards Arg and Lys, as well as the acylated and O-methylated modified residues, as shown in Table 1.

R

R

MPnT: R=OP(CH3)O2 2 MPT: R=OPO3

MST: R=OSO3 MCT: R=OCH2CO2

FigureFigure 15.15. Structure of of the the Molecular Molecular Tw Tweezerseezers reported reported in in Schradder Schradder et etal. al. functionalized functionalized with with phosphonate (MPnT), phosphate (MPT), sulfate (MST), and carboxylate (MCT). phosphonate (MPnT), phosphate (MPT), sulfate (MST), and carboxylate (MCT).

The first of these receptors to be tested was the phosphonate tweezer, which showed se- Table 1. Binding constants (Ka) for the formation of host-guest complexes between amino acids and synthetic supramolecu- lectivity towards Lys, with even higher affinity towards its α-N/C-Protected form. NMR and lar receptors. ITC studies elucidated the interactions formed in these complexes where all the analytes tested −1 Host Guest showedKa a(M full incorporation) of their Conditionsside chain near the hydroquinone group Method (Figure 16). Ref. The binding1.52 process× 103 is aidedPhosphate by cation– Bufferπ interactions 10 mM pH between 8, 20 ◦C the positively ITC charged groups [21] of the amino acids and the electron-rich interior region of the tweezer, as well as by hydrophobic 1.7 × 103 95% water/5% deuterium oxide pH 5 interactions between the alkyl chains of the analytes and the aromatic groups. However,[75 ]the 2.0 × 102 95% water/5% deuterium oxide pH 1 1.55 × 103 95% water/5% deuterium oxide pH 8, 20 ◦C[22] L-Arginine 1H-NMR 6.40 × 103 Deuterium oxide unbuffered, 25 ◦C[88]

1.30 × 104 Deuterium oxide unbuffered, 25 ◦C [86] 2.8 × 104 Acetate buffer 5 mM pD 4.5 Deuterium oxide 40 mM phosphate buffer 3.30 × 102 [89] pD 7, 25 ◦C SC4 7.4 × 102 Phosphate Buffer 10 mM pH 8, 20 ◦C ITC [21] 6.0 × 102 95% water/5% Deuterium oxide pH 5 [75] 1.0 × 102 95% water/5% Deuterium oxide pH 1 1H-NMR 1.36 × 103 95% water/5% Deuterium oxide pH 8, 20 ◦C[22] L-Lysine Fluorescence <1 × 103 Ammonium acetate buffer 10 mM, pH 6, 25 ◦C [87] Spectroscopy 4.60 × 103 Deuterium oxide unbuffered 1H-NMR [86] 3.90 × 103 Acetate buffer 5 mM pD 4.5 5.20 × 102

L-LysMe 4.00 × 103 ◦ 4 Phosphate buffer 40 mM, pH 7.4, 30 C ITC [89] L-LysMe2 1.60 × 10 4 L-LysMe3 3.70 × 10 Molecules 2021, 26, 106 15 of 41

Table 1. Cont.

−1 Host Guest Ka (M ) Conditions Method Ref. ◦ L-Histidine 5.00 × 102 95% water/5% Deuterium oxide pH 8, 20 C [22]

L-Arginine 1.90 × 102 ◦ L-Lysine 90 Phosphate buffer pH 8, 20 C [21] 1 L-Arginine 3.50 × 102 H-NMR 2 L-Lysine 4.2 × 10 ◦ SC4-Ar Phosphate buffer 40 mM pD 7.0, 25 C [90] 4 L-LysMe3 6.4 × 10 ◦ L-Lysine 1.40 × 102 Phosphate Buffer pH 8, 20 C

C4Pyr L-Glutamate 1.76 × 103 Water pH 6 [52] L-Aspartate × 3 1.16 10 Fluorescence ◦ P5 Bodipy L-Asparagine 2.5 × 105 Dimethyl formamide/water 1:1, 25 C[Spectroscopy 95] 5.24 × 103 HEPES buffer pH 7.4, 25 ◦C[94] L-Arginine 5.90 × 103 Deuterium oxide pD 7.2, 25 ◦C 1H-NMR [26] Fluorescence 1.12 × 103 HEPES buffer pH 7.4, 25 ◦C [94] Spectroscopy CP5 L-Lysine 1.80 × 103

L-Histidine 1.50 × 103 Deuterium oxide pD 7.2, 25 ◦C 1 [26] TriMe-Lys 1.30 × 103 H-NMR Ac-Lys 1.90 × 102 L-Glutamate 1.00 × 106 Fluorescence PDA6 Water pH 6, 25 ◦C [51] L-Aspartate 9.80 × 105 Spectroscopy L-Lysine 1.40 × 103 Deuterium oxide, Sodium Ac-His-OMe 7.00 × 102 Dihydrogenophosphate 25 mM pH 4.4 1H-NMR [24] MPnT 4.40 × 103 Ac-Lys-OMe Deuterium oxide unbuffered 2.30 × 104

L-Lysine 1.14 × 103

L-Lysine 4.76 × 104 Phosphate buffer 200 mM pH 7.6 Ac-Arg-OMe 1.67 × 104 MPT Ac-Lys-OMe 5.88 × 104 Ac-Arg-OMe 5.00 × 104 Ac-Lys-OMe 1.11 × 105 Phosphate buffer 10 mM pH 7.6 L-Lysine 4.41 × 103

L-Arginine 1.43 × 103 MST Fluorescence [25] Ac-Arg-OMe 1.30 × 104 Spectroscopy Phosphate buffer 10 mM pH 7.2 Ac-Lys-OMe 5.26 × 104

L-Lysine 8.60 × 102

L-Arginine 1.64 × 103 Phosphate buffer 10 mM pH 7.2 MCT Ac-Arg-OMe 3.56 × 103 Ac-Lys-OMe 1.56 × 103 Ac-Lys-OMe 4.42 × 103 Phosphate buffer 200 mM pH 7.6 Molecules 2021, 26, 106 16 of 41

Table 1. Cont.

−1 Host Guest Ka (M ) Conditions Method Ref. 3.16 × 104 Methanol 3.98 × 104 Water with 1.6% Methanol CEAT Lys-OMe [118] HEPES buffer 50 mM pH 7.5 with 7.0 × 101 1.6% Methanol ◦ L-Lysine 1.10 × 104 Deuterium oxide 0.1 M NaCl, 25 C 1H-NMR [101] Glycine 4.70 × 103 × 3 CB6 L-Alanine 1.00 10 ◦ Aqueous Formic Acid 50%(v/v), 25 C [35] L-Valine 1.40 × 103 3 L-Phenylalanine 1.40 × 10 ITC 1.80 × 106 Water, 25 ◦C[36] 1.80 × 105 Phosphate Buffer pH 7, 25 ◦C [102] L-Phenylalanine 1.20 × 106 Phosphoric acid buffer pH 2, 25 ◦C 1.50 × 105 Water, 25 ◦C 1H-NMR 5 8.20 × 10 UV-Visible [44] nd Spectroscopy 2.30 × 105 1.60 × 104 Phosphate Buffer pH 7, 25 ◦C ITC [102] L-Tyrosine 1.80 × 105 Phosphoric acid buffer pH 2, 25 ◦C Fluorescence 2.40 × 104 Ammonium Acetate Buffer pH 6, 30 ◦C Spectroscopy [45] 2.20 × 104 ITC UV-Visible 3.70 × 105 nd [44] Spectroscopy CB7 1.20 × 103 Phosphate Buffer pH 7, 25 ◦C ITC [102] L-Tryptophan 7.40 × 103 Phosphoric acid buffer pH 2, 25 ◦C Fluorescence 1.60 × 103 Ammonium Acetate Buffer 10 mM pH 6, 25 ◦C Spectroscopy 1.90 × 103 Ammonium Acetate Buffer 10 mM pH 6, 30 ◦C ITC Fluorescence [45] 8.70 × 102 Ammonium Acetate Buffer 10 mM pH 6, 25 ◦C Spectroscopy 8.00 × 102 Ammonium Acetate Buffer 10 mM pH 6, 30 ◦C L-Lysine 2.10 × 102 Phosphate Buffer pH 7, 25 ◦C ITC [102] 3.10 × 105 Phosphoric acid buffer pH 2, 25 ◦C 5.30 × 102 Lys-NMe 1.80 × 103 Sodium Acetate Buffer 50 mM, deuterium 1 [101] 4 ◦ H-NMR Lys-NMe2 6.00 × 10 oxide pD 4.7, 25 C 6 Lys-NMe3 1.90 × 10 Fluorescence 3.10 × 102 Ammonium Acetate Buffer 10 mM pH 6, 25 ◦C [45] Spectroscopy L-Arginine 1.40 × 105 Phosphoric acid buffer pH 2, 25 ◦C [102] ITC ◦ 3.27 × 102 Ammonium Acetate Buffer 10 mM pH 6, 30 C [45] Molecules 2021, 26, 106 17 of 41

Table 1. Cont.

−1 Host Guest Ka (M ) Conditions Method Ref. Fluorescence 4.00 × 102 Ammonium Acetate Buffer 10 mM pH 6, 25 ◦C L-Histidine Spectroscopy 2.40 × 103 Phosphoric acid buffer pH 2, 25 ◦C ITC [102]

L-Tyrosine a <1.00 × 103 a 8 Sodium Phosphate 10 mM pH 7 L-Phenylalanine 1.10 × 10 ◦ [110] CB8-AA2, 27 C L-Tryptophan a 6.90 × 107

L-Tyrosine 2.20 × 103 ITC

L-Phenylalanine 5.30 × 103 CB8 Sodium Phosphate 10 mM pH 7, co-binding [37] ◦ L-Tryptophan 4.30 × 104 with MV, 27 C

L-Trp-OMe 6.30 × 104 UV-Visible 4.20 × 105 Co-binding with DPT [38] Spectroscopy L-Tryptophan Sodium Phosphate 10 mM pH 7, co-binding 3.40 × 104 ITC [39] with MBBI, 27 ◦C

L-Phenylalanine 4.20 × 104 ◦ Fluorescence L-Tyrosine 5.70 × 104 Sodium Acetate 50 mM pH 4.74, 22 C [40] CB6Ar Spectroscopy L-Tryptophan 3.20 × 106 Glycine 5.60 × 102

L-Alanine 1.12 × 103 ◦ α-CDx Aqueous Formic Acid 50%(v/v), 25 C ITC [35] L-Valine 1.62 × 103

L-Phenylalanine 2.6 × 102

◦ Fluorescence β-CDx L-Tyrosine 5.0 × 101 Phosphate Buffer 0.01 M pH 7.0, 20 C [119] Spectroscopy

L-Tryptophan 5.24 × 104

L-Phenylalanine 2.24 × 104 ◦ β-CDU L-Tyrosine 1.33 × 104 Phosphate buffer 50 mM, 25 C ITC [120]

L-Alanine 5.00 × 103

L-Serine 1.97 × 104

L-Tryptophan 1.00 × 103 ◦ CPQ L-Tyrosine 4.54 × 102 Phosphate buffer 50 mM pH 7, 25 C 1H-NMR [116] L-Phenylalanine 1.06 × 102 N-Me- 1.70 × 105 Leucine·HCl N-Me- 1.40 × 105 Cav[(CH2)2 Alanine·HCl CH3, · 4 ◦ Proline HCl 1.20 × 10 Methanol, 20 C [115] CH2CH3] ITC Threonine·HCl 2.00 × 104 Alanine·HCl 1.10 × 104 Tyrosine·HCl 1.10 × 104 Cysteine·HCl 7.30 × 103 Molecules 2021, 26, 106 18 of 41

Table 1. Cont.

−1 Host Guest Ka (M ) Conditions Method Ref. N-Me- 1.07 × 106 Lysine·HCl 1:1 Methanol, 20 ◦C (amino acid:cavitand) N-Me- 1.45 × 103 Lysine·HCl 1:2 4 ◦ N-Me-Glycine- 6.80 × 10 Methanol, 20 C methyl 3.43 × 103 Water, 20 ◦C ester·HCl Cav[(CH2)2 1.89 × 103 Phosphate Buffer Saline 0.1 M pH 7, 20 ◦C + − CH3Py Cl , 3 CH CH ] Sarcosine 1.02 × 10 2 3 Water, 20 ◦C 1.49 × 103 N-Me-Lysine·HCl 1.13 × 103 Phosphate Buffer Saline 0.1 M pH 7, 20 ◦C ◦ TeCPh L-Tryptophan 1.21 × 103 Phosphate Buffer 10 mM pH 7.4, 25 C[42]

N-Ac-D-Aspartate 1.62 × 104 OA (R,R)-1 × 3 N-Ac-D-Glutamate 3.31 10 ◦ pH-metric [53] Tetramethylammonium chloride 0.1 M, 20 C titrations OA N-Ac-L-Aspartate 3.40 × 102 (S,S,S,S)-2 N-Ac-L-Glutamate 6.30 × 102

N-Ac-L-Alanine 2.10 × 103 1:1 90% water/10% Dimethyl sulfoxide GP3cat ITC [59] N-Ac-L-Glutamate 1.52 × 106 2:1 90% water/10% Dimethyl sulfoxide a 1:2 host-guest stoichiometry. Overall binding constant in M−2. The overall binding constant corresponds to the product of the stepwise binding constants for the 1:1 and 1:2 complexes, i.e., K11.K12.

The first of these receptors to be tested was the phosphonate tweezer, which showed selectivity towards Lys, with even higher affinity towards its α-N/C-Protected form. NMR and ITC studies elucidated the interactions formed in these complexes where all the analytes tested showed a full incorporation of their side chain near the hydroquinone group (Figure 16). The binding process is aided by cation–π interactions between the positively charged groups of the amino acids and the electron-rich interior region of the tweezer, as well as by hydrophobic interactions between the alkyl chains of the analytes and the aromatic groups. However, the formation of the complexes are mainly driven by ionic interactions between the receptor’s anionic groups and the positive charges in the analyte—explaining the higher affinity towards Lys, seeing as arginine presents a more delocalized charge, diminishing the stability of this latter complex [24]. This was supported by later studies with molecular tweezers comprising different anionic moieties, in which the phosphate and sulfonate containing tweezers present the highest affinities, in contrast to phosphonate and carboxylate which present lower charge density. The lower affinity of most of these tweezers to non-protected forms of the amino acids is most likely due to the presence of repulsive forces between the receptor and the carboxylate group of the amino acids [25]. Another application of this concept was developed by Mandl and König, in which they made a receptor with two modified crown ether moieties, CEAT [118] (Figure 17). These are capable of binding O-methylated Lys, with an affinity of 4.0 × 104 M−1, namely by the formation of H-bonds between the crown ether ring and the amines in the amino acid. However, these receptors have rather small affinities when in aqueous buffered solutions, while also needing the presence of a small percentage of methanol, which indicates that they are less suitable for physiological applications [118]. Molecules 2021, 26, 106 15 of 44

formation of the complexes are mainly driven by ionic interactions between the receptor’s an- ionic groups and the positive charges in the analyte—explaining the higher affinity towards Lys, seeing as arginine presents a more delocalized charge, diminishing the stability of this latter complex [24].

Molecules 2021, 26, 106 15 of 44

formation of the complexes are mainly driven by ionic interactions between the receptor’s an- Molecules 2021, 26, 106 ionic groups and the positive charges in the analyte—explaining the higher affinity19 of 41towards Lys, seeing as arginine presents a more delocalized charge, diminishing the stability of this latter complex [24].

Figure 16. (a) Proposed structure adapted from Fokkens et al. and Monte Carlo simulations of the complexes between MPnT and Ac-Lys-OMe (b) and Ts-Arg-OEt (c)—MacroModel 7.1, Amber*, water, 5000 steps. Reprinted from [24] with permission. Copyright (2005) American Chemical Society.

This was supported by later studies with molecular tweezers comprising different anionic moieties, in which the phosphate and sulfonate containing tweezers present the highest affinities, in contrast to phosphonate and carboxylate which present lower charge density. The lower affinity of most of these tweezers to non-protected forms of the amino acids is most likely due to the presence of repulsive forces between the receptor and the carboxylate group of the amino acids [25]. Another application of this concept was developed by Mandl and König, in which they made a receptor with two modified crown ether moieties, CEAT [118] (Figure 17).

These are capable of binding O-methylated Lys, with an affinity of 4.0 × 104 M−1, namely Figure 16. (a) ProposedFigureby structurethe 16. formation(a) adapted Proposed of from H-bonds structure Fokkens adaptedbetween et al. froman thed Monte Fokkenscrown Carlo ether et al. simulations andring Monteand the of Carlo theamines simulationscomplexes in the between ofamino the MPnT and Ac-Lys-OMecomplexesacid. (b )However, and between Ts-Arg-OEt these MPnT (receptorsc and)—MacroModel Ac-Lys-OMe have rather 7.1, (b) andAmber*, sm Ts-Arg-OEtall affinitieswater, ( c5000)—MacroModel when steps. in Reprinted aqueous 7.1, Amber*, from buffered [24] water, with so- permission. Copyright5000lutions, (2005) steps. Am while Reprintederican also Chemical fromneeding [24 Society.] withthe presence permission. of Copyright a small percentage (2005) American of methanol, Chemical Society.which indi- cates that they are less suitable for physiological applications [118]. This was supported by later studies with molecular tweezers comprising different anionic moieties, in which the phosphate and sulfonate containing tweezers present the highest affinities, in contrast to phosphonate and carboxylate which present lower charge density. The lower affinity of most of these tweezers to non-protected forms of the amino acids is most likely due to the presence of repulsive forces between the receptor and the carboxylate group of the amino acids [25]. Another application of this concept was developed by Mandl and König, in which they made a receptor with two modified crown ether moieties, CEAT [118] (Figure 17).

These are capable of binding O-methylated Lys, with an affinity of 4.0 × 104 M−1, namely FigureFigureby 17. 17. theStructure Structure formation of of the the of molecular molecular H-bonds tweezer, tweezer, between CEAT, CEAT, the developed crow developedn ether by by Mandl ring Mandl and and and the König König amines [118 [118].]. in the amino acid. However, these receptors have rather small affinities when in aqueous buffered so- Molecules 2021, 26, 106 SchmuckSchmuck and and Geiger Geiger also also developed developed an an open open chain chain receptor receptor for for amino amino acids, acids, based based16 of 44 lutions, while also needing the presence of a small percentage of methanol, which indi- on a guanidinium pyrrole unit, with 2 added cationic groups, GP3cat (Figure 18). This on a guanidiniumcates that they pyrrole are less unit, suitable with for 2 added physiological cationic applications groups, GP3cat [118]. (Figure 18). This receptorreceptor showed showed a remarkablea remarkable increase increase in affinityin affinity towards towards acetylated acetylatedL-Ala, L-Ala, in comparison in compari- toson other to monoother mono or di-cationic or di-cationic forms offorms the receptor.of the receptor. This 1:1 This complex 1:1 complex is mainly is stabilized mainly stabi- by ioniclized interactions by ionic interactions between thebetween receptor the andreceptor the carboxylateand the carboxylate of the analyte, of the analyte, as well as well by multipleas by multiple hydrogen hydrogen bonds that bonds are formedthat are atform theed same at the time same with time the samewith groups.the same Despite groups. the extra carboxylate of glutamate, the 1:1 complex is more stable with alanine, although Despite the extra carboxylate of glutamate, the 1:1 complex is more stable with alanine, two receptors can bind to 1 molecule of glutamate, which increases the affinity towards this although two receptors can bind to 1 molecule of glutamate, which increases the affinity amino acid. Although promising, this system was not studied solely in water, still needing towards this amino acid. Although promising, this system was not studied solely in water, 10% DMSO in order to increase its solubility [59]. still needing 10% DMSO in order to increase its solubility [59].

Figure 17. Structure of the molecular tweezer, CEAT, developed by Mandl and König [118].

Schmuck and Geiger also developed an open chain receptor for amino acids, based on a guanidinium pyrrole unit, with 2 added cationic groups, GP3cat (Figure 18). This receptor showed a remarkable increase in affinity towards acetylated L-Ala, in compari-

FigureFigure 18. 18.Structure Structure of of the the guanidinium guanidinium pyrrole pyrrole receptor, receptor, GP3cat GP3cat [59 [59].].

Table 1. Binding constants (Ka) for the formation of host-guest complexes between amino acids and synthetic supramolecular receptors.

Host Guest Ka (M−1) Conditions Method Ref. Phosphate Buffer 10 1.52 × 103 ITC [21] mM pH 8, 20 °C 95% water/5% deu- 1.7 × 103 terium oxide pH 5 [75] 95% water/5% deu- 2.0 × 102 terium oxide pH 1 95% water/5% deu- 1.55 × 103 terium oxide pH 8, [22] L-Arginine 20 °C Deuterium oxide 1 6.40 × 103 H-NMR [88] unbuffered, 25 °C Deuterium oxide 1.30 × 104 unbuffered, 25 °C [86] Acetate buffer 5 mM 2.8 × 104 pD 4.5 SC4 Deuterium oxide 40 3.30 × 102 mM phosphate [89] buffer pD 7, 25 °C Phosphate Buffer 10 7.4 × 102 ITC [21] mM pH 8, 20 °C 95% water/5% Deu- 6.0 × 102 terium oxide pH 5 [75] 95% water/5% Deu- 1.0 × 102 1 L-Lysine terium oxide pH 1 H-NMR 95% water/5% Deu- 1.36 × 103 terium oxide pH 8, [22] 20 °C Ammonium acetate Fluores- < 1 × 103 buffer 10 mM, pH 6, cence Spec- [87] 25 °C troscopy

Molecules 2021, 26, 106 20 of 41

4. Recognition of Peptides: Taking Inspiration in Nature One of the main targets for protein detection is the recognition of key elements, most often on the surface of the protein, seeing as this is the region most easily available for binding. For small peptides most of their residues are easily available for binding and is also easier to select a target region for detection. In either case, the best way to achieve this is by the recognition of the peptide side-chains or C and N-terminal amino acid residues [7]. In this section, the focus will be on the previously mentioned receptors, and other, more complex structures, applied in the selective recognition of target amino acid sequences in peptides [22,24–26,35,52,121–123].

4.1. Macrocyclic Receptors From the receptors presented before, there are a few that have received a larger focus and that their affinity towards certain residues and groups is well defined. Cyclodextrins, for example, have been shown to have affinity for hydrophobic amino acids like phenylala- nine, but still bind weakly to the peptides studied in most works, while cucurbit[n]urils, which are selective for hydrophobic and basic amino acids form complexes with peptides and proteins that are significantly more stable, reaching nanomolar affinities in specific cases [37,108,124,125]. Charged macrocyclic receptors, as can be expected, bind oppositely charged residues, being this, together with the hydrophobic effect, the main driving forces for peptide binding in aqueous solutions while other non-covalent interactions may con- tribute in a small extent for overall binding energy [7,9,33]. However, it must be stressed that these secondary interactions may play a decisive role in the overall selectivity and structure of the complexes. The simplest and non-modified forms of these macrocycles can be used for selective binding of peptides, which is reported in many studies [21,35,41]. Using these as a starting point, many other receptors have been developed, modifying the original structures to suit the type of amino acids and peptides that are being targeted [40,52]. There are also other types of macrocycles that, while less used for biological applications, have been adapted and show promise in the binding and detection of peptides, as was touched upon in the previous chapters [51,94,95].

4.1.1. Calixarenes p-Sulfonatocalix[4]arene (SC4) binds not only cationic amino acids but also small cationic peptides. Complexes with polycationic peptides are, in general, more stable due to the formation multivalent ionic interactions. One of the first studies with polycationic peptides and SC4 was reported by Morel-Desrosiers and co-workers [22], in which they observe some changes in the way that peptides bind to this receptor, in comparison to the single amino acid. For Lys-Lys, Arg-Arg and Lys-Arg, the N-terminal amino acid is enclosed within the cavity of the receptor, with the C-terminal amino acid staying only near the entrance of the macrocycle. NMR studies showed that, between Lys-Lys and Arg-Arg, the latter binds deeper in the SC4 cavity, most likely due to the stabilization of the complex by cation–π interactions. The two other peptides differ slightly in the mode of binding—while Lys-Arg seems to bind with the N-terminal amino acid, lysine, in the cavity and arginine remaining outside the macrocycle, the Arg-Lys peptide binds with the C-terminal deeper in the cavity. The tripeptides tested in this study showed higher stabilization than the remainder of the analytes. This is most likely due to two main factors: the multivalent ionic interactions, due to the presence of an added cationic group, increasing the sites of interaction and, so, increasing the stability of the complex [126]; a greater distance between the C-terminal and the sulfonate groups of the receptor diminishes the electrostatic repulsion between these two negatively charged groups and allows for more stable binding with the N-terminal amino acid, deeper in the cavity, and the middle amino acid, at the entrance of the cavity. This study also showed that, in general, the complexes formed with the amino acid L-Arg and Arginine peptides are slightly more stable than their L-Lys counterparts, due to the Molecules 2021, 26, 106 21 of 41

presence of the guanidinium moiety that may undergo π–π interactions with the calixarene cavity, as explained in Section 3.1.1.[22]. Ghale et al. [127] also used p-sulfonatocalix[4]arene for the detection of arginine-rich peptides, using an indicator displacement method. In this study, this receptor was applied for the detection of several cationic analytes in liposomes (with more focus on protamine, a natural occurring peptide with 21 arginine residues) and to study their transport across membranes, mediated by the membrane protein OmpF. The association constants presented in Table2 further establish that, in general, this macrocycle forms more stable complexes with peptides with higher number of positively charged residues due multivalent ionic interactions, as commented above. Later studies show the same effect with an amphiphilic SC4 and its complexation with heptaarginine and other small arginine-rich cell-penetrating peptides [121]. Molecules 2021, 26, 106 The high affinities towards multivalent cationic peptides make this receptor a promis-23 of 44

ing one, for in vitro study of several types of peptides—antimicrobial peptides [33], pro- tamines [128], and mitochondrial signaling peptides [129]—but also make it a viable tool forinclude applications the use of in SC4 sensing as a promoter of proteins of andprot theirein crystallization, modulation. Selectedprotein assembly examples and of thisas a includemasking the agent, use ofto SC4alter asproteins a promoter’ surface of protein properties crystallization, [130–135]. protein assembly and as a masking agent, to alter proteins’ surface properties [130–135]. This is the case of studies with the protein cytochrome c, with characteristic positively This is the case of studies with the protein cytochrome c, with characteristic positively charged regions at the surface, composed of several lysine residues, which have a very charged regions at the surface, composed of several lysine residues, which have a very important role in the protein–protein interactions that Cyt c can establish. McGovern et important role in the protein–protein interactions that Cyt c can establish. McGovern al. have shown that SC4 binds this protein at three different lysine residues (Figure 19), et al. have shown that SC4 binds this protein at three different lysine residues (Figure 19), forming ionic and cation–π interactions with the residue, but being further stabilized by forming ionic and cation–π interactions with the residue, but being further stabilized by hydrogen bonds with the nearest polar residues and groups, as well as by the hydropho- hydrogen bonds with the nearest polar residues and groups, as well as by the hydrophobic bic effect. The binding of SC4 results in a change in the overall charge at these sites, con- effect. The binding of SC4 results in a change in the overall charge at these sites, conceal- cealing the lysine residues and altering the protein–protein recognition properties of these ing the lysine residues and altering the protein–protein recognition properties of these biomolecules. Moreover, the changes at the protein surface enhanced the protein’s ability biomolecules. Moreover, the changes at the protein surface enhanced the protein’s ability to form crystals, indicating possible uses of SC4, not only in physiological conditions, for to form crystals, indicating possible uses of SC4, not only in physiological conditions, for function modulation purposes,purposes, butbut alsoalso asas anan enhancerenhancer ofof crystallizationcrystallization [[131].131].

(a) (b)

Lys5

Lys22 Lys4 Lys89

Figure 19. Image of 3TYI (structure obtained by McGovern et al. [131]) created with the software Discovery Studio 2020 Client (BIOVIA, Dessault Systèmes, San Diego: Dessault Systèmes, 2020) (a) Asymmetric unit of the crystal structure of the Figure 19. Image of 3TYI (structure obtained by McGovern et al. [131]) created with the software Discovery Studio 2020 cytochromeClient (BIOVIA, c-SC4 Dessault complex, Systèmes showing, San three Diego: interact Dessault Table 89. Systèmes, (red), Lys22 2020) (orange), (a) Asymmetric and Lys4 unit (yellow), of the atcrystal the surface structure of the of protein,the cytochrome with respective c-SC4 complex, interactions showing in bright three orange. interacTable (b) Side 89. view (red), of oneLys22 of (orange) the proteins, and in Lys4 theasymmetric (yellow), at unitthe surface for better of visualizationthe protein, with of the respective Lys4 binding interactions site; Lys5 in is bright also represented orange. (b)in Side yellow, view for of itone could of the form proteins complementary in the asymmetric interactions unit with for thebetter SC4 visualization molecule in of its the proximity. Lys4 binding site; Lys5 is also represented in yellow, for it could form complementary interac- tions with the SC4 molecule in its proximity.

4.1.2. Calixpyridinium This macrocycle was tested not only with amino acids, as previously mentioned, but with a glutamate dipeptide, to evaluate if the high affinity was maintained or if there were any changes in the binding mode. The same study showed that the calixpyridinium-pep- tide complex presented very similar stability as the one with only glutamate, which might indicate that the binding occurs mostly in the same way (Table 2) [52]. In the case of the dipeptide it is possible that the N-terminal residue is also inside the macrocycle’s cavity but that the electrostatic repulsion between the positive N-terminal hinders the binding, resulting in equal affinity to that of L-Glu. Despite the possible use of this system in the detection of peptides, it remains that calixpyridinium presents higher affinity constants towards nucleosides, in particular to- wards ATP, and so, being more interesting as a receptor for this anionic molecule [136].

4.1.3. Pillararenes Much like calixpyridinium, pillararenes previously mentioned either were not fur- ther tested for their affinity towards peptides or were only tested with a couple of small peptides, yielding very similar results as for the binding of the respective amino acids.

Molecules 2021, 26, 106 22 of 41

4.1.2. Calixpyridinium This macrocycle was tested not only with amino acids, as previously mentioned, but with a glutamate dipeptide, to evaluate if the high affinity was maintained or if there were any changes in the binding mode. The same study showed that the calixpyridinium- peptide complex presented very similar stability as the one with only glutamate, which might indicate that the binding occurs mostly in the same way (Table2)[ 52]. In the case of the dipeptide it is possible that the N-terminal residue is also inside the macrocycle’s cavity but that the electrostatic repulsion between the positive N-terminal hinders the binding, resulting in equal affinity to that of L-Glu. Despite the possible use of this system in the detection of peptides, it remains that cal- ixpyridinium presents higher affinity constants towards nucleosides, in particular towards ATP, and so, being more interesting as a receptor for this anionic molecule [136].

4.1.3. Pillararenes Much like calixpyridinium, pillararenes previously mentioned either were not further tested for their affinity towards peptides or were only tested with a couple of small peptides, yielding very similar results as for the binding of the respective amino acids. Carboxylatopillar[5]arene (CP5) was tested for two peptides—Ala-Arg-Ala and Ala-Lys- Ala—in the same study mentioned before [26]. NMR studies showed that, in this case, the CP5 receptor only encapsulated the side chain of the middle basic amino acids, stabilizing both by ionic interactions and the hydrophobic effect. Similar to the amino acid results, this macrocycle has five times higher affinity for the peptide with Arginine, most likely due to the formation of cation–π interactions, giving it a rather high selectivity for the presence of this amino acid.

4.1.4. Cucurbiturils The binding of peptides to CB[n] constitutes a remarkable success story in the field of host-guest systems comprising this type of biological molecules. As previously mentioned, CB[n] generally display higher affinity for hydrophobic aromatic amino acids being the release of high energy water molecules from the macrocycle’s cavity the main thermo- dynamic driving force contributing for the stability of the complexes [105]. Ion-dipole and hydrogen bonding interactions between carbonyls portals of the receptor and specific functional groups present in the peptide are believed to contribute in a less extension for the overall driving force but may have a decisive role in the observed selectivity. The high affinity interactions of N-terminal aromatic residues, in particular phenylalanine, with CB7 and CB8 constitutes a paradigmatic target for the selective binding behavior of this class of hosts [7,37,108,122,124]. For example, Phe-Gly and CB7 form a 1:1 com- plex with a K = 3.0 × 107 M−1 while the complex formed with Gly-Phe is much weaker (K = 1.3 × 103 M−1), as is summarized in Table2[ 122]. In both cases the phenylalanine residue is included in the CB7 cavity. However, while in Phe-Gly this binding mode favors attractive ion-dipole interactions between the carbonyl portals and the ammonium group and minimizes the repulsion of the portal with the carboxylate group, for Gly-Phe the carboxylate is closer to the carbonyl group precluding optimal inclusion by electrostatic repulsion. Furthermore, the attractive interaction between the ammonium group in the Gly residue with the carbonyl portals of the receptor imposes a significant entropic penalty due to conformational restriction of the guest [122]. Although displaying slightly lower binding constant, both tryptophan- and tyrosine-glycine dipeptides display similar behavior to that described for Phe-Gly/Gly-Phe [122]. It is also worth noting that the smaller CB6, in contrast to CB7 and CB8 (see below), does not significantly bind peptides with terminal aromatic residues, showing much higher affinity for those comprising terminal lysine [122]. Urbach and co-workers [37,108] focus their earlier work on the binding of N-terminal amino acids to the larger CB8. As described above for CB7, CB8 also binds preferentially peptides containing aromatic amino acids at the N-terminal. These receptor-analyte pairs can form 1:1:1 complexes in the presence of methyl viologen as an auxiliary guest [37], but Molecules 2021, 26, 106 23 of 41

can also assist the dimerization N-terminal aromatic peptides in the absence of methyl viologen by forming 1:2 host–guest complexes. CB8 shows selectivity towards Trp-Gly-Gly and Phe-Gly-Gly (Figure 20)[108], forming CB8-peptide2 complexes with overall binding constant up to 3 × 1011 M−2 (Table2)[ 108,137,138]. While the arguments described above Molecules 2021, 26, 106 to rationalize the selective binding of N-terminal aromatic residues to CB7 are also likely25 toof 44

hold here, the crystal structures shown in Figure 20, besides showing those interactions, also demonstrate that hydrogen bonding between the amide protons and the oxygen atoms in the carbonyl portals may also contribute to increase the stability of these complexes [108].

(a) (b)

FigureFigure 20. 20.Crystal Crystal structure structure of of the the complexes complexes with with CB8 CB8 (structure (structure in in white) white) and and the th peptidese peptides (structures (structures in in black)—( black)—(a)a CB8-) CB8- (Trp-Gly-Gly)(Trp-Gly-Gly) and and (b) ( CB8-(Phe-Gly-Gly)2.b) CB8-(Phe-Gly-Gly)2. Despite Despite the the formation formation of 1:2 of complexes1:2 complexes for bothfor both these these peptides, peptides, Heitmann Heitmann et al. et wereal. were only ableonly to able obtain to obtain this complex this complex structure structure for Phe-Gly-Gly. for Phe-Gly-Gly. Reprinted Reprinted with permission with perm fromission [108 from]. Copyright [108]. Copyright (2006) (2006) American Chemical Society. American Chemical Society.

MoreMore studies studies have have been been done done with with CB8, CB8, screening screening for for any any specificity specificity among among peptides peptides withwithN -terminalN-terminal aromatic aromatic residues residues [125 [125].]. Most Most of of these these were were performed performed with with tripeptides, tripeptides, inin which which the theN -terminalN-terminal residue residue was was aromatic, aromatic, and and the twothe two others others varied, varied, in order in order to assess to as- theirsess importance their importance in the in binding the binding process. process. It was It discovered was discovered that, even that, thougheven though most ofmost the of peptidesthe peptides bind inbind a 2:1 in ratioa 2:1 toratio CB8, to theCB8, sequence the sequence Tyr-Leu-Ala Tyr-Leu-Ala showed showed high affinityhigh affinity (in the (in nMthe range) nM range) for this for macrocycle. this macrocycle. However, However, it bonded it inbonded a 1:1 ratio in a instead 1:1 ratio of theinstead 2:1 observed of the 2:1 previously.observed previously. This was not This only was true not for only Tyr-Leu-Ala; true for Tyr-Leu-Ala; when tyrosine when was tyrosine the N -terminalwas the N- aminoterminal acid, amino some acid, peptides some formed peptides a 1:1formed complex a 1:1 withcomplex CB8, with because CB8, the because adjacent the aminoadjacent 0 acidamino could acid also could be encapsulated also be encapsulated in the CB8 ins cavity the CB8 along′s cavity with Lalong-Tyr, leavingwith L-Tyr, the C-terminal leaving the atC-terminal the entrance at ofthe the entrance macrocycle of the (Figure macrocycle 21). These (Figure peptides 21). These were peptides of the Tyr-X-Ala were of the kind Tyr- (withX-Ala X kind being (with either X being Leu, Lys,either Phe Leu, or Lys, Tyr), Phe and or didTyr), not and bind did innot this bind way in this to CB8 way when to CB8 Xwhen and Ala X and were Ala switched. were switched. This highThis high affinity affinity towards towards a Tyr-Leu-Ala a Tyr-Leu-Ala peptide peptide is ais stepa step towardstowards developing developing simple simple supramolecular supramolecular systems systems that that mimic mimic the the specificity specificity displayed displayed byby -based antibody-based receptors. receptors. TheThe same same researchers researchers proceeded proceeded toto dodo further screening, screening, namely namely with with a alibrary library of of144 144tripeptides, tripeptides, X1-X2-Ala. X1-X2-Ala. X1 X1 consisted consisted of of amino amino acids withwith largelargehydrophobic hydrophobic or or cationic cationic sideside chains, chains, seeing seeing as as this this will will be be the the residue residue farther farther inside inside CB8s CB8s cavity cavity and and the the larger larger size size isis needed needed in in order order to to have have 1:1 1:1 binding. binding. X2 X2 corresponds corresponds to to 18 18 possible possible amino amino acids, acids, leaving leaving offoff only only Trp Trp and and Cis Cis from from the the existing existing 20 human20 human physiological physiological amino amino acids, acids, due due to possible to possi- interferenceble interference in the in measurements the measurements [123]. [123]. This study showed that CB8 had affinity towards peptides with Tyr, Phe, Ile, and Leu at X1 and Tyr, Phe, Leu, and Lys at X2. Sequences with Met, Arg and Lys also showed considerable affinity. The affinity towards methionine at the X1 position was the one explored in this study, seeing as Met is often found on N-terminal of proteins before further post-translational modification, being of particular interest to study these PTM mechanisms [139].

Figure 21. Semiempirical structure obtained by Smith et al. (2015) of the complex CB8-(Tyr-Leu-Ala)—up view (left) with the electrostatic interactions represented by dashed lines, and side view (right), where it is observable the two N-terminal residues inserted in the cavity of the macrocycle. Reprinted with permission from [125]. Copyright (2015) American Chem- ical Society.

Molecules 2021, 26, 106 25 of 44

(a) (b)

Figure 20. Crystal structure of the complexes with CB8 (structure in white) and the peptides (structures in black)—(a) CB8- (Trp-Gly-Gly) and (b) CB8-(Phe-Gly-Gly)2. Despite the formation of 1:2 complexes for both these peptides, Heitmann et al. were only able to obtain this complex structure for Phe-Gly-Gly. Reprinted with permission from [108]. Copyright (2006) American Chemical Society.

More studies have been done with CB8, screening for any specificity among peptides with N-terminal aromatic residues [125]. Most of these were performed with tripeptides, in which the N-terminal residue was aromatic, and the two others varied, in order to as- sess their importance in the binding process. It was discovered that, even though most of the peptides bind in a 2:1 ratio to CB8, the sequence Tyr-Leu-Ala showed high affinity (in the nM range) for this macrocycle. However, it bonded in a 1:1 ratio instead of the 2:1 observed previously. This was not only true for Tyr-Leu-Ala; when tyrosine was the N- terminal amino acid, some peptides formed a 1:1 complex with CB8, because the adjacent amino acid could also be encapsulated in the CB8′s cavity along with L-Tyr, leaving the C-terminal at the entrance of the macrocycle (Figure 21). These peptides were of the Tyr- X-Ala kind (with X being either Leu, Lys, Phe or Tyr), and did not bind in this way to CB8 when X and Ala were switched. This high affinity towards a Tyr-Leu-Ala peptide is a step towards developing simple supramolecular systems that mimic the specificity displayed by antibody-based receptors. The same researchers proceeded to do further screening, namely with a library of 144 tripeptides, X1-X2-Ala. X1 consisted of amino acids with large hydrophobic or cationic side chains, seeing as this will be the residue farther inside CB8s cavity and the larger size Molecules 2021, 26, 106 is needed in order to have 1:1 binding. X2 corresponds to 18 possible amino acids,24 of 41 leaving off only Trp and Cis from the existing 20 human physiological amino acids, due to possi- ble interference in the measurements [123].

Figure 21. SemiempiricalFigure structure 21. Semiempirical obtained by structure Smith et obtainedal. (2015) byof the Smith complex et al. CB8-(Tyr-Leu-Ala)—up (2015) of the complex CB8-(Tyr-Leu- view (left) with the electrostatic interactionsAla)—up represente view (left)d withby dashed the electrostatic lines, and side interactions view (right), represented where it by is dashedobservable lines, the and two side N-terminal view residues inserted in(right), the cavity where of the it ismacrocycle. observable Reprinte the twodN with-terminal permission residues from inserted [125]. Copyright in the cavity (2015) of the American macrocycle. Chem- ical Society. Reprinted with permission from [125]. Copyright (2015) American Chemical Society.

Analyzing the binding by ITC studies, showed that the second amino acid should be large and hydrophobic or cationic, but without branching in the beta carbon, while the third

amino acid should be small, so as to avoid having stereochemical hindrances [123]. These types of tripeptides show sub micromolar affinities towards CB8 and are also a plausible target for the detection of proteins by their N-terminal, seeing as, despite methionine being a common site of excision, large amino acids in the position next to Met can block the binding of the enzymes that catalyze this process, which distinguishes these N-terminal sequences from the excised ones [139]. Additionally, this functionality of the macrocycle can be used as a protection against excision of the N-terminal, for proteins presenting these sequences [109]. More recent studies focus on different modes of peptide binding to the receptor CB8, with a focus on peptides containing the pairs of residues Tyr-Leu or Phe-Leu. Here it is shown that CB8 can bind to these residues even if farther away from the N-terminal, with a diminished affinity by one order of magnitude, and that the binding can occur not only in a 1:1 ratio, but also in a 1:2 ratio for longer peptides with repeated Tyr-Leu sequences (Tyr-Leu-Ala-Gly-Gly-Ala-Leu-Tyr) [140]. Due to the presence of aromatic amino acids in insulin’s N-terminal, this protein comes up as a possible target for this type of receptors. With this in mind, Chinai et al. evaluated the affinity of this system towards human insulin and a mutant variety without the N-terminal aromatic amino acid [124]. This latter variant showed an obvious decrease in affinity towards CB7 when compared to human insulin. The latter showed similar affinity towards the macrocycle as Phe-Gly-Gly. Furthermore, it was observable a slight change in conformation in insulin to allow for this binding, exposing the N-terminal to the complexation, but without altering the overall structure of the protein. This implies that even less exposed N-terminal amino acids could be targets for this type of sensing without any major interference with the rest of the structure, while also distinguishing them from other aromatic amino acids in a protein’s surface or in a peptide’s sequence (Figure 22). Additionally, the N-terminal is usually a more exposed and flexible region in proteins, being possible that this could be more easily transposed to studies with peptides with a corresponding sequence. As can be seen by this example, CB[n]’s can be used for the detection of proteins, due to its specificity towards aromatic and cationic residues. Control over protein assembly and function is another route that is being explored for this supramolecular system [9,141–148]. Brunsveld and co-workers have used CB8 as a tool to aid in the dimerization of proteins, making use of the target Phe-Gly-Gly motif as an N-terminal modification, in order to better control enzyme activity [149] and protein assemblies [142]. Molecules 2021, 26, 106 26 of 44

This study showed that CB8 had affinity towards peptides with Tyr, Phe, Ile, and Leu at X1 and Tyr, Phe, Leu, and Lys at X2. Sequences with Met, Arg and Lys also showed considerable affinity. The affinity towards methionine at the X1 position was the one ex- plored in this study, seeing as Met is often found on N-terminal of proteins before further post-translational modification, being of particular interest to study these PTM mecha- nisms [139]. Analyzing the binding by ITC studies, showed that the second amino acid should be large and hydrophobic or cationic, but without branching in the beta carbon, while the third amino acid should be small, so as to avoid having stereochemical hindrances [123]. These types of tripeptides show sub micromolar affinities towards CB8 and are also a plausible target for the detection of proteins by their N-terminal, seeing as, despite methi- onine being a common site of excision, large amino acids in the position next to Met can block the binding of the enzymes that catalyze this process, which distinguishes these N- terminal sequences from the excised ones [139]. Additionally, this functionality of the macrocycle can be used as a protection against excision of the N-terminal, for proteins presenting these sequences [109]. More recent studies focus on different modes of peptide binding to the receptor CB8, with a focus on peptides containing the pairs of residues Tyr-Leu or Phe-Leu. Here it is shown that CB8 can bind to these residues even if farther away from the N-terminal, with a diminished affinity by one order of magnitude, and that the binding can occur not only in a 1:1 ratio, but also in a 1:2 ratio for longer peptides with repeated Tyr-Leu sequences (Tyr-Leu-Ala-Gly-Gly-Ala-Leu-Tyr) [140]. Due to the presence of aromatic amino acids in insulin’s N-terminal, this protein comes up as a possible target for this type of receptors. With this in mind, Chinai et al. evaluated the affinity of this system towards human insulin and a mutant variety without the N-terminal aromatic amino acid [124]. This latter variant showed an obvious decrease in affinity towards CB7 when compared to human insulin. The latter showed similar af- finity towards the macrocycle as Phe-Gly-Gly. Furthermore, it was observable a slight change in conformation in insulin to allow for this binding, exposing the N-terminal to the complexation, but without altering the overall structure of the protein. This implies that even less exposed N-terminal amino acids could be targets for this type of sensing without any major interference with the rest of the structure, while also distinguishing them from other aromatic amino acids in a protein’s surface or in a peptide’s sequence Molecules 2021, 26, 106 (Figure 22). Additionally, the N-terminal is usually a more exposed and flexible region in 25 of 41 proteins, being possible that this could be more easily transposed to studies with peptides with a corresponding sequence.

Figure 22. FigureCrystal 22.structureCrystal of structure the complex of the formed complex between formed CB7 between and human CB7 andinsulin human (light insulin green) (light green) and of freeand insulin of free(black). insulin Reprinted (black). with Reprinted permission with from permission [124]. Copyright from [124 (2011)]. Copyright American (2011) American Chemical Society.Chemical Society.

The second example consists of the use of a modified peptide, with a 14-3-3 protein binding site at the C-terminal and with an Phe-Gly-Gly moiety at the N-terminal. This structure allows for the binding of this peptide to the 14-3-3 protein, which presents two binding sites, with affinities comparable to those of the physiological targets. CB8 can be added to stabilize this bivalent complex by 14 times, by complexation of the two N- terminals of the peptides bound to this protein, serving as a tool to switch the “availability” of the 14-3-3 binding site [142]. A more recent study reported the role of CB8 as a modulator of enzymatic activity using caspase 8, an enzyme that is typically only active upon dimerization. In this work Dang et al., after observing that the Casp-8 N-terminal residues of the two monomers are in close proximity in the dimeric active state, engineered this region of the Casp-8 to have an Phe-Gly-Gly motif [149]. This motif allowed for the use of CB8 to control the dimerization in the inactive mutants and resulted in an easily modulated, dimeric form of Casp-8 active towards a synthetic ligand as well as its physiological substrate, caspase-3. The CB[n]’s affinity towards basic residues has also been explored in the scope of protein recognition. Namely, Crowley and co-workers tested CB70s affinity towards a demethylated lysine in a protein, Ralstonia solanacearum lectin (RSL). Despite having other methylated lysine residues in its sequence, CB7 recognized selectively the Lys34Me2, which is located in a more accessible region of RSL, yielding an affinity in the mM range (Table2). Moreover, CB7 has also been successfully exploited in this context to create novel protein assemblies around CB7 trimers and tetramers, in the crystal structure of the CB7-RSL complexes [150].

4.1.5. Cyclodextrins As described in the previous sections, the properties of these natural occurring macro- cycles translate to higher affinities towards aromatic residues or residues with small, apolar side chains in the context of peptides and proteins [35,41,119,151,152]. The same types of interactions occur with both amino acids and peptides—the binding at the cyclodextrin cavity is generally driven by hydrophobic effects, with some hydrogen bonds formed with the outer shell of the macrocycle. In most cases, these interactions only allow for small affinities, only surpassing mM affinities in 50% methanol solutions [152], or with further modification of the original macrocycle [151].

4.1.6. Crown Ethers A different approach for detection of aromatic amino acids and small peptides con- taining these residues was presented by Weißenstein et al. [153]. In their studies they joined Molecules 2021, 26, 106 26 of 41

the properties of two different moieties—as readout mechanism they used a perylene bisimide dye (PBI), which has high fluorescence quantum yield and can be quenched by photoinduced electron transfer (PET)—this process can occur when this group forms bonds with electron rich moieties, e.g., aromatic amino acids which can bind to PBI by π–π stacking of aromatic groups; the other distinct moiety is composed of two crown ethers that serve as a receptor for ammonium cations, e.g., the N-terminal of short peptides or amine groups present in cationic amino acids. The presence of two crown ethers allows for Molecules 2021, 26, 106 the binding of two analytes, while PBI can form two π–π stacking bonds with the aromatic28 of 44

amino acids (Figure 23).

FigureFigure 23. 23. StructureStructure of of the the crown crown ether ether molecular molecular tw tweezer,eezer, perylene perylene bisimide bisimide dye dye (PBI) (PBI) [153]. [153].

TheThe highest highest affinity affinity of of PBI, PBI, at least oneone orderorder of of magnitude magnitude above above that that of of other other peptides pep- tidesand and amino amino acids acids tested, tested, was towardswas towards Ala-Trp. Ala- ThisTrp. peptide This peptide is believed is believed to have to the have optimal the optimalcharacteristics characteristics for binding for binding to occur to dueoccur to due the to separation the separation between between the ammonium the ammonium ion at ionthe atN the-terminal N-terminal and theand aromatic the aromatic residue. residue. It must It must be ideal be ideal for the for formation the formation of interactions of inter- actionsbetween between the N-terminal the N-terminal ammonium ammonium group andgroup the and crown the ethercrown and ether between and between the aromatic the aromaticside chain side and chain the and perylene the perylene bisimide bisimide unit, at theunit, same at the time. same The time. downside The downside of this probeof this is probethat itsis that studies its studies were not were made not in made water in (probably water (probably due to poordue solubilityto poor solubility of the receptor) of the receptor)and so, theand conclusionsso, the conclusions taken, andtaken, the and overall the overall system system are, most are, likely,most likely, not applicable not appli- in cablewater in andwater physiological and physiological conditions. conditions. SimilarSimilar systems systems have have been been explored explored before, before, in in water, water, as as is is seen seen as as early early as as 1998 1998 in in the the workwork of of Hossain Hossain and and Schneider Schneider [154]. [154]. They They used used a acrown crown ether ether and and a trimethylammonium a trimethylammonium moietiesmoieties to to bind bind to to the the peptide’s peptide’s NN andand C-terminals, C-terminals, respectively. respectively. These These moieties moieties were were connectedconnected by by a ahydrophobic hydrophobic linker, linker, with an addedadded fluorescentfluorescent dansyl dansyl unit unit for for the the detection detec- tionof theof the biomolecules biomolecules (Figure (Figure 24 ).24 The). The synthesized synthesized receptor, receptor, CENMe CENMe3, reached3, reached affinities affinities up 3 −1 upto to 10 10M3 M−1,, forfor both Gly-Phe-Gly and and Gly-Trp-Gly, Gly-Trp-Gly, at at least least 1 1 order order of of magnitude magnitude above above thethe affinities affinities towards towards other other tested tested peptides. peptides. Ev Evenen though though this this receptor receptor shows shows specificity specificity towardstowards the the middle middle positioning positioning of of an an aromatic aromatic residueresidue inin thisthis typetype of of peptides, peptides, for for biological biolog- 3 −1 icalapplications, applications, a 10 a 10M3 M−1affinity affinity constant constant is is not not the the most most desired desired value, value, since since physiological physiolog- icalconcentrations concentrations are are much much lower lower than thosethan those needed needed for considerable for considerable complex complex formation. for- mation. 4.1.7. Cavitands Cavitand receptors have seen multiple applications in recognition of small organic molecules, including peptides and proteins. Some examples comprise the integration of these macrocycles in sensing scaffolds [155,156], nanostructures [157], as well as their use in solution [158–160]. The latter includes a group of self-folding deep cavitands, DCv (Figure 25), which have been applied in the detection of select histone sequences and their

Figure 24. Structure of the crown ether and trimethylammonium sensor, CENMe3 [154].

Molecules 2021, 26, 106 28 of 44

Figure 23. Structure of the crown ether molecular tweezer, perylene bisimide dye (PBI) [153].

The highest affinity of PBI, at least one order of magnitude above that of other pep- tides and amino acids tested, was towards Ala-Trp. This peptide is believed to have the optimal characteristics for binding to occur due to the separation between the ammonium ion at the N-terminal and the aromatic residue. It must be ideal for the formation of inter- actions between the N-terminal ammonium group and the crown ether and between the aromatic side chain and the perylene bisimide unit, at the same time. The downside of this probe is that its studies were not made in water (probably due to poor solubility of the Molecules 2021, 26, 106 receptor) and so, the conclusions taken, and the overall system are, most likely, not27 appli- of 41 cable in water and physiological conditions. Similar systems have been explored before, in water, as is seen as early as 1998 in the work of Hossain and Schneider [154]. They used a crown ether and a trimethylammonium respectivemoieties to post-translational bind to the peptide’s modifications N and C-terminals, (PTM) in water.respectively. These These interact moieties with lysine were residues,connected showing by a hydrophobic different affinities linker, with depending an added on thefluorescent R5 group’s dansyl nature unit (Figure for the 25 detec-) and the degree of methylation of the guest. The complexes established are stabilized mainly tion of the biomolecules (Figure 24). The synthesized receptor, CENMe3, reached affinities π byup cation–to 103 M−interactions1, for both Gly-Phe-Gly but also with and the Gly-Trp-Gly, contribution at ofleast a variety 1 order of of other magnitude electrostatic above interactionsthe affinities [ 157towards,159,160 other]. Furthermore, tested peptides. all three Even of though the deep this cavitands receptor schematized shows specificity have beentowards applied the middle in a dye positioning displacement of an assay aromatic for the re screeningsidue in this and type monitoring of peptides, of the for PTM biolog- on aical select applications, library of peptides.a 103 M−1 affinity These included constant several is not the methylation most desired sites value, that are since recognized physiolog- by bothical concentrations histone methyltransferase are much andlower demethylase, than those allowingneeded for anconsiderable analysis of theircomplex activity for- at several of the specific sites in which they act upon [159]. mation. Molecules 2021, 26, 106 29 of 44

4.1.7. Cavitands Cavitand receptors have seen multiple applications in recognition of small organic molecules, including peptides and proteins. Some examples comprise the integration of these macrocycles in sensing scaffolds [155,156], nanostructures [157], as well as their use in solution [158–160]. The latter includes a group of self-folding deep cavitands, DCv (Fig- ure 25), which have been applied in the detection of select histone sequences and their respective post-translational modifications (PTM) in water. These interact with lysine res- idues, showing different affinities depending on the R5 group’s nature (Figure 25) and the degree of methylation of the guest. The complexes established are stabilized mainly by cation–π interactions but also with the contribution of a variety of other electrostatic in- teractions [157,159,160]. Furthermore, all three of the deep cavitands schematized have been applied in a dye displacement assay for the screening and monitoring of the PTM on a select library of peptides. These included several methylation sites that are recognized Figure 24. Structure of the crown ether and trimethylammonium sensor, CENMe3 [154]. by Figureboth histone 24. Structure methyltransferase of the crown and ether demethyla and trimethylammoniumse, allowing for an sensor, analysis CENMe of their3 [ 154ac-]. tivity at several of the specific sites in which they act upon [159].

FigureFigure 25. Structure 25. Structure of the of deep the cavitands, deep cavitands, DCv1, DCv2 DCv1, and DCv2 DCv3 and [158,159]. DCv3 [158,159].

4.2. Open-Chain Receptors Besides the obvious interest in peptide and protein sensing, many receptors for pep- tides have been explored focusing on the role of these biomolecules as targets for the ac- tion of several pharmacological agents. The and its derivatives are amongst the most recognized examples. Vancomycin binds a specific sequence in the of gram-positive bacteria, D- Ala-D-Ala-OH, disabling the action of a transpeptidase at this site, which has a very im- portant role in the crosslinking of several peptide strands in the peptidoglycan layer and stabilizing it. Without this crosslinking, the cell envelope is severely destabilized, and lysis

Molecules 2021, 26, 106 28 of 41

4.2. Open-Chain Receptors Besides the obvious interest in peptide and protein sensing, many receptors for pep- tides have been explored focusing on the role of these biomolecules as targets for the action of several pharmacological agents. The antibiotic vancomycin and its derivatives are amongst the most recognized examples. Vancomycin binds a specific sequence in the cell wall of gram-positive bacteria, D-Ala- Molecules 2021, 26, 106 30 of 44 D-Ala-OH, disabling the action of a transpeptidase at this site, which has a very important role in the crosslinking of several peptide strands in the peptidoglycan layer and stabilizing it. Without this crosslinking, the cell envelope is severely destabilized, and lysis of the cell ofwill the cell occur will if thereoccur areif there variations are variations in the local in the osmotic local osmotic pressure. pressure. In drug In resistant drug resistant strands strandsof the sameof the bacteria, same bacteria, this target this sequence target sequence is mutated is mutated to D-Ala- toD -DLac-OH.-Ala-D-Lac-OH. This change This in changeone amino in one acid amino breaks acid a bondbreaks between a bond vancomycinbetween vancomycin and the peptide and the and peptide adds electrostaticand adds electrostaticrepulsion between repulsion the between two oxygens the two indicated oxygens below indicated (Figure below 26). This (Figure diminishes 26). This the dimin- affinity ishesconstant the affinity of vancomycin constant byof vancomycin two orders of by magnitude, two orders but of doesmagnitude, not affect but the does binding not affect of the thebacterial binding transpeptidase of the bacterial [ 6transpeptidase]. [6].

(a)

(b) (c)

Figure 26. (a) Vancomycin, (b) its target sequence in the cell wall of gram positive bacteria, D-Ala-D- Figure 26. (a) Vancomycin, (b) its target sequence in the cell wall of gram positive bacteria, D-Ala- Ala-OH and (c) the mutated sequence, D-Ala-D-Lac-OH. The sites in vancomycin highlighted in pink, D-Ala-OH and (c) the mutated sequence, D-Ala-D-Lac-OH. The sites in vancomycin highlighted in blue and green interact with the same colored groups in the sequences. In the mutated sequence, pink, blue and green interact with the same colored groups in the sequences. In the mutated se- one of the hydrogen bonds is broken and there is instead electrostatic repulsion between the groups quence, one of the hydrogen bonds is broken and there is instead electrostatic repulsion between thein groups blue [6 ].in blue [6]. In order to circumvent bacterial mechanisms of resistance, Ellman and co-workers [161] In order to circumvent bacterial mechanisms of resistance, Ellman and co-workers synthesized a library of water soluble receptors, based on vancomycin’s structure, but [161] synthesized a library of water soluble receptors, based on vancomycin’s structure, with a peptidyl sequence in the region where the repulsive forces were introduced when but with a peptidyl sequence in the region where the repulsive forces were introduced a mutation occurred. The peptidyl group has freer rotation than the rigid group of van- when a mutation occurred. The peptidyl group has freer rotation than the rigid group of comycin, and this will allow for the increase of the distance between receptor and peptide, vancomycin, and this will allow for the increase of the distance between receptor and pep- diminishing the effect of the repulsion when binding occurs. The receptor shows better com- tide, diminishing the effect of the repulsion when binding occurs. The receptor shows bet- plementarity to the sequences and, so, the highest affinity was observed for VD1 (104 M−1), ter complementarity to the sequences and, so, the highest affinity was observed for VD1 represented in Figure 27. (104 M−1), represented in Figure 27.

Figure 27. Structure of the peptidyl vancomycin derivative receptor, VD1 [161].

Molecules 2021, 26, 106 30 of 44

of the cell will occur if there are variations in the local osmotic pressure. In drug resistant strands of the same bacteria, this target sequence is mutated to D-Ala-D-Lac-OH. This change in one amino acid breaks a bond between vancomycin and the peptide and adds electrostatic repulsion between the two oxygens indicated below (Figure 26). This dimin- ishes the affinity constant of vancomycin by two orders of magnitude, but does not affect the binding of the bacterial transpeptidase [6].

(a)

(b) (c)

Figure 26. (a) Vancomycin, (b) its target sequence in the cell wall of gram positive bacteria, D-Ala- D-Ala-OH and (c) the mutated sequence, D-Ala-D-Lac-OH. The sites in vancomycin highlighted in pink, blue and green interact with the same colored groups in the sequences. In the mutated se- quence, one of the hydrogen bonds is broken and there is instead electrostatic repulsion between the groups in blue [6].

In order to circumvent bacterial mechanisms of resistance, Ellman and co-workers [161] synthesized a library of water soluble receptors, based on vancomycin’s structure, but with a peptidyl sequence in the region where the repulsive forces were introduced when a mutation occurred. The peptidyl group has freer rotation than the rigid group of vancomycin, and this will allow for the increase of the distance between receptor and pep- Molecules 2021, 26, 106 tide, diminishing the effect of the repulsion when binding occurs. The receptor shows29 of bet- 41 ter complementarity to the sequences and, so, the highest affinity was observed for VD1 (104 M−1), represented in Figure 27.

Molecules 2021, 26, 106 31 of 44

FigureFigure 27. 27.Structure Structure of of the the peptidyl peptidyl vancomycin vancomycin derivative derivative receptor, receptor, VD1 VD1 [161 [161].]. In more recent studies, with other receptors, smaller libraries are being utilized, tak- ing advantageIn more of information recent studies, obtained with other from receptors, previous smaller studies libraries to focus are the being design utilized, of new taking receptorsadvantage towards of the information properties obtainedthat are needed from previous [162]. studies to focus the design of new

Inreceptors a study towards by Schmuck the properties and Heil, that with are the needed tetrapeptide [162]. N-Ac-D-Glu-L-Lys-D-Ala-D- Ala-OH—EKAA—inIn a study bymind Schmuck as a target and Heil, (a bigger with thefragment tetrapeptide of the Npeptide-Ac-D-Glu- recognizedL-Lys-D by-Ala- D- vancomycin),Ala-OH—EKAA—in a small library mind of water-soluble as a target (a receptors bigger fragment was created, of the with peptide 512 members. recognized by They vancomycin),used a similar astrategy small library as the ofone water-soluble used for VD1, receptors synthesizing was created,these receptors with 512 with members. a peptidylThey chain, used with a similar three strategyvariable asresidues, the one adding used for the VD1, guanidiniocarbonyl synthesizing these pyrrole receptors bind- with ing motifa peptidyl (GCP), chain, which with was threepreviously variable shown residues, to bind adding to the theC-terminal guanidiniocarbonyl of peptides (as pyrrole mentionedbinding in motifSection (GCP), 2.3, relative which wasto the previously binding shownof single to amino bind to acids) the C-terminal [117]. The of GCP peptides binding(as motif mentioned shows in similarities Section 2.3 with, relative vancomyc to thein binding in the oftypes single of interactions amino acids) established [117]. The GCP with bindingthe carbonyl motif group shows of similarities this sequence with vancomycinwith several in H-bonds the types stabilizing of interactions both establishedcom- plexes.with In thethe carbonylGCP-peptide group complex of this sequence there is the with additional several H-bonds ionic bond stabilizing previously both complexes.men- tioned.In the GCP-peptide complex there is the additional ionic bond previously mentioned. The residuesThe residues of the of peptidyl the peptidyl group group varied varied between between Lys, Tyr, Lys, Ser, Tyr, Glu, Ser, Phe, Glu, Val, Phe, Leu, Val, Leu, and Trp,and being Trp, beingexpected expected that some that of some these of can these form can more form ionic more interactions ionic interactions with the withan- the alyte analyteand further and stabilize further the stabilize resulting the complex. resulting Due complex. to the pyrrole Due to moiety the pyrrole it was moiety possible it was to monitorpossible the to binding monitor of the the binding target peptides of the target with peptides the receptors with the from receptors the variations from the in variations the absorptionin the and absorption fluorescence and fluorescence spectra of the spectra receptors. of the It receptors. was found It wasthat foundthe receptor that the with receptor the sequencewith the Lys-Lys-Phe, sequence Lys-Lys-Phe, GCP-KKF GCP-KKF (Figure 28 (Figure), was the28), one was which the one presented which presented higher af- higher finityaffinity towards towards both EKAA both EKAAand the and opposite the opposite sequence, sequence, AAKE, AAKE, but with but markedly with markedly higher higher 4 −1 affinityaffinity towards towards the peptide the peptide with with Ala Alaat the at C-terminal the C-terminal (Ka (K= 1.71a = 1.71 × 10×4 M10−1, M4 times, 4 timeshigher higher than thanthat for that AAKE), for AAKE), indicating indicating the importance the importance of the of C-terminal the C-terminal in the in binding the binding of the of the peptide.peptide. Furthermore, Furthermore, the lysine the lysine residue residue also alsotakes takes part part in the in thebinding binding with with the the C-termi- C-terminal, nal, stabilizingstabilizing the the binding binding to to the the tetrapeptide tetrapeptide further further [163]. [163].

FigureFigure 28. Structure 28. Structure of the ofpeptidyl the peptidyl Guanidinioca Guanidiniocarbonylrbonyl pyrrole pyrrole receptor, receptor, GCP-KKF GCP-KKF [163]. [163].

The receptorThe receptor which which showed showed highest highest affinity affinity towards towards EKAA EKAA was was also also tested tested for its affin- affinityity towards towards other other tetrapeptides, with variablevariable residues,residues, originatingoriginating a a library library of of ~300 ~300 target target tetrapeptides, including those with D-Ala and D-Lac in the C-terminal (the first sim- ilar to the native sequence of Gram positive bacteria peptidoglycan peptidyl chain and respective mutated sequence, related to vancomycin resistance). The GCP-KKF receptor showed an even higher affinity towards Ac-D-Glu-D-Glu-D-Glu-D-Glu-OH (K = 2.65 × 104 M−1) and a very accentuated stereoselectivity for D-Ala in D-Glu-D-Glu-D/L-Ala-D-Glu-OH peptide (almost 4× higher than for L-Ala, Ka (L stereoisomer) = 1.40 × 103 M−1 and Ka (D stereoisomer) = 4.50 × 103 M−1) [164]. This shows that focusing on libraries and taking inspiration on known and well-stud- ied host-guest complexes helps to understand the bonds created and how to further sta- bilize other peptide-receptor complexes [6,163,164]. Molecular Tweezers

Molecules 2021, 26, 106 30 of 41

tetrapeptides, including those with D-Ala and D-Lac in the C-terminal (the first similar to the native sequence of Gram positive bacteria peptidoglycan peptidyl chain and respective mutated sequence, related to vancomycin resistance). The GCP-KKF receptor showed − an even higher affinity towards Ac-D-Glu-D-Glu-D-Glu-D-Glu-OH (K = 2.65 × 104 M 1) and a very accentuated stereoselectivity for D-Ala in D-Glu-D-Glu-D/L-Ala-D-Glu-OH 3 −1 peptide (almost 4× higher than for L-Ala, Ka (L stereoisomer) = 1.40 × 10 M and 3 −1 Ka (D stereoisomer) = 4.50 × 10 M )[164]. This shows that focusing on libraries and taking inspiration on known and well- studied host-guest complexes helps to understand the bonds created and how to further stabilize other peptide-receptor complexes [6,163,164].

Molecular Tweezers Molecular tweezer and clips constitute another example of small supramolecular receptors that have been widely explored for the recognition of peptides. These com- pounds offer an (often electron-rich aromatic) pocket-like binding site, which is usually decorated with functional groups that may provide additional sites of interaction and water-solubility [165–167]. The most popular molecular tweezers with applications in the recognition of amino acids and peptides are probably those developed by the Klärner group, which bind pref- erentially basic amino acid lysine and arginine, as mentioned in Section 3.2.[167]. Some of these compounds show a tendency to form self-assembled dimers, a process that may compete with the complexation of selected targets and lead to concentration dependent apparent binding constants [168]. However, anionic tweezers such as the ones shown is Figure 15 (see Section 3.2) are fluorescent, allowing the monitorization of the complexation process and determination of the binding constants at low concentrations [168]. Besides the amino acids tested, a small library of bioactive peptides with basic residues was also investigated, with importance and applications in several fields [167]. The results showed that the stability of the complexes depends on the anionic groups decorating the receptor and increases according to the following trend: O-methylenecarboxylate < phosphonate < sulfate < phosphate. Due to their electron-rich cavities and anionic functional groups this type of receptor fails to bind peptides lacking basic amino acids, showing, in the other hand, high affinity (µM range) for peptides bearing arginine and/or lysine residues, being the last generally more stable probably due to charge delocalization in the guanidinium groups. The highest affinities were reported for the peptides comprising several adjacent lysine residues at the N-terminal which provide more sites to form ionic interactions while minimizing the repulsion of the C-terminal [25]. In general, the complexes seem to be slightly more stable in water than in the methanol, demonstrating the important role of the hydrophobic effect that, nevertheless, is coun- terbalanced by the increasing strength of the ionic interactions in the organic solvent. Noteworthy, although the binding constant present modest variation upon changing from aqueous to organic media, NMR studies clearly showed that the type of complexes can be dramatically different depending on the solvent: in water the formation of inclusion complexes predominate while methanol favors external complexes [168]. However, it should be stressed that this behavior cannot be generalized and depends on the nature of the anionic groups. The phosphonate, phosphate and sulfate containing molecular tweezers all complexed peptides with at least one L-Lys residue, while the carboxylate containing tweezer had rather low affinities or did not bind at all to the peptides studied, similarly to their behavior in the binding of the amino acids. The phosphate molecular tweezer, MPT, has been of great interest, due to its high affinity and specificity towards the positive residues of peptides. Early on, they have been tested as a possible pharmacological agent [25,169–171] and showed positive therapeutic effects in tests with animal models of several diseases, without significant cytotoxicity— they bind to amyloidogenic lysine and arginine-rich proteins (e.g., α-synuclein), inhibiting Molecules 2021, 26, 106 31 of 41

their aggregation [25]; MPT also has shown the ability to disrupt and distort viral envelopes (e.g., Zika, Ebola [170], HIV [169,170]), due to their high levels of sphingomyelin and cholesterol. The carboxylate molecular tweezer, MCT, also shows to inhibit viral activity and disrupt the viral envelope, despite not binding strongly with cationic residues—it was hypothesized, in a 2020 study [171], that the MCT and MPT tweezers are able to interact at virus-like lipid rafts, forming inclusion complexes with sphingomyelin, and disrupt them at the interface with the rest of the membrane, by increasing the already high surface tension of these envelopes. Other studies explored the importance of the linker in these tweezer receptors, show- ing again that it is necessary to have some rigidity in the linker between the “arms of the Molecules 2021, 26, 106 33 of 44 tweezer” and a certain angle and distance between the arms to be able to encase the peptide between them [172–174].

4.3.4.3. Self-Assembled Self-Assembled Coordination Coordination Cages Cages MetalMetal organic organic capsules capsules (or (or self-assembled self-assembled coordination coordination cages) cages) constructed constructed from from or- or- ganicganic ligands ligands and and metal metal ions ions oftenoften displaydisplay wellwell defineddefined binding pockets, pockets, suitable suitable to to ac- accommodatecommodate complementary guest guest molecules molecules in in their their interior. interior. These These have have been been widely widely ex- exploredplored as receptorsreceptors forfor aa varietyvariety of of molecular molecular targets targets [175 [175–177].–177]. The The positively positively charged charged Pd L coordination cage (Figure 29) reported by Fujita was shown to bind complementary Pd6 64L4 coordination cage (Figure 29) reported by Fujita was shown to bind complementary oligopeptides with high affinity and impressive sequence selectivity [178,179]. The Pd L oligopeptides with high affinity and impressive sequence selectivity [178,179]. The 6Pd46L4 bindingbinding pocket pocket was was found found to to be be large large enough enough to to accommodate accommodate up up to to three three amino amino acid acid residues in its interior. By analyzing tripeptides with different sequences strong binding residues in its interior. By analyzing tripeptides with different6 sequences−1 strong binding was observed for the Ac-Trp-Trp-Ala-NH2 sequence (K > 10 M ) while very similar was observed for the Ac-Trp-Trp-Ala-NH2 sequence (K > 106 M−1) while very similar se- sequences, such as Ac- Trp-Ala-Trp-NH2, Ac-Ala-Trp-Trp-NH2, Ac-Trp-Trp-Gly-NH2, Ac- quences, such as Ac- Trp-Ala-Trp-NH2, Ac-Ala-Trp-Trp-NH2, Ac-Trp-Trp-Gly-NH2, Ac- Trp-Tyr-Ala-NH2 showed much lower affinity (affinities of the first two sequences can be Trp-Tyr-Ala-NH2 showed much lower affinity (affinities of the first two sequences can be seen in Table2)[ 179]. The results suggest that the cooperative interactions between the two seen in Table 2) [179]. The results suggest that the cooperative interactions between the indole rings and the Ala methyl group with the cage and as well intramolecular interactions two indole rings and the Ala methyl group with the cage and as well intramolecular in- between the residues (such as π–π and CH-π) may impart a decisive contribution to the teractions between the residues (such as π–π and CH-π) may impart a decisive contribu- thermodynamic driving force. This seems to put in evidence the potential advantages of tion to the thermodynamic driving force. This seems to put in evidence the potential ad- using receptors with cavities large enough to accommodate more than one residues in its vantages of using receptors with cavities large enough to accommodate more than one interior as potential strategies to reach exquisite selectivity [179]. residues in its interior as potential strategies to reach exquisite selectivity [179].

Figure 29. Chemical structure of the Fujita’s Pd6L4 coordination cage. Reprinted with permission Figure 29. Chemical structure of the Fujita’s Pd6L4 coordination cage. Reprinted with permission from [179]. Copyright (2005) American Chemical Society. from [179]. Copyright (2005) American Chemical Society.

More recently, Nitschke and co-workers reported a large Fe8L6 cubic coordination More recently, Nitschke and co-workers reported a large Fe8L6 cubic coordination cagecage assembled assembled from from Zn-porphyrin Zn-porphyrin based based ligands ligands and and iron(II) iron(II) metal metal ions, ions, FeZnP, FeZnP, able able to to encapsulateencapsulate guest guest molecule molecule containing containing imidazole imidazol functionale functional groups groups that that were were anticipated anticipated to interact with the Zn-porphyrin capsule walls [180]. After demonstrating the affinity of the capsule for this type of guests, the authors investigated the ability of the nanocontainer to bind peptides with histidine residues in a 1:1 acetonitrile:water solvent system. Cla- vanin A, a peptide antibiotic, containing four histidines among its 23 amino acids was found to bind strongly to the capsule with a dissociation constant of 80 nM in a 1:2 binding manner (Table 2). A less hydrophobic peptide containing only three histidines was found to bind 100-fold more weakly, while peptides lacking histidine residues did not interact at all.

Molecules 2021, 26, 106 32 of 41

to interact with the Zn-porphyrin capsule walls [180]. After demonstrating the affinity of the capsule for this type of guests, the authors investigated the ability of the nanocontainer to bind peptides with histidine residues in a 1:1 acetonitrile:water solvent system. Clavanin A, a peptide antibiotic, containing four histidines among its 23 amino acids was found to bind strongly to the capsule with a dissociation constant of 80 nM in a 1:2 binding manner (Table2). A less hydrophobic peptide containing only three histidines was found to bind 100-fold more weakly, while peptides lacking histidine residues did not interact at all.

Table 2. Binding constants (Ka) for the formation of host-guest complexes between small peptides and synthetic supramolecular receptors.

−1 Receptor Guest Ka (M ) Conditions Method Arg-Arg 7.00 × 103 Arg-Lys 3.90 × 103 Arg-Arg-Arg 3.30 × 104 90% water/10% Deuterium oxide ◦ 1H-NMR 3 Phosphate Buffer 10 mM pH 8, 20 C Lys-Lys 3.40 × 10 [22] Lys-Arg 3.70 × 103 4 SC4 Lys-Lys-Lys 3.30 × 10 Trp-Lys-Arg-Thr-Leu- 1.20 × 106 HEPES buffer 10 mM pH 7, 20 ◦C Fluorescence Arg-Arg-Leu Spectroscopy Protamine 1.20 × 109 Phosphate buffer 10 mM pH, 25 ◦C [127]

Cyt C binding site 1 1.20 × 103 20 mM Potassium dihydrogenophosphate, 50 mM NaCl, 1 [131] 2 mM sodium ascorbate, 10% D O, Cyt C binding site 2 6.30 × 10 2 pH 6, 30 ◦C 7 Fluorescence (Arg)7 7.00 × 10 Spectroscopy (Arg) 2.90 × 107 ◦ 7 HEPES buffer 10 mM pH 7, 25 C [121] SC4-C5 Trp-Lys-Arg-Thr-Leu- 2.80 × 106 Arg-Arg-Leu C4Pyr Glu-Glu 1.7 × 103 Water pH 6 [52] Ala-Arg-Ala 4.20 × 103 ◦ 1 CP5 Deuterium oxide pD 7.2, 25 C H-NMR [26] Ala-Lys-Ala 7.50 × 102 Phe-Gly 3.00 × 107 Gly-Phe 1.30 × 103 Tyr-Gly 3.60 × 106 [122] Water, 25 ◦C Gly-Tyr 2.00 × 102 ITC Trp-Gly 5.60 × 105 CB7 Gly-Trp 2.80 × 102 Phe-Gly-Gly 2.80 × 106 Sodium Phosphate 10 mM pH 7, 27 ◦C [124] Insulin 1.50 × 106 20 mM Potassium Phosphate, 50 mM 3 Sodium Chloride,1.2 mM 1 15 RSL-Lys34Me2 1.00 × 10 H N HSQC [150] α-methyl-L-fucoside, 90%H2O/10%D2O, pH 6, 30 ◦C Trp-Gly-Gly 1.30 × 105 Gly-Trp-Gly × 4 2.10 10 Sodium Phosphate 10 mM pH 7, [37] CB8 ITC Gly-Gly-Trp 3.1 × 103 co-binding with MV, 27 ◦C Gly-Gly-Trp-Gly-Gly 2.50 × 104 Molecules 2021, 26, 106 33 of 41

Table 2. Cont.

−1 Receptor Guest Ka (M ) Conditions Method Phe-Gly-Gly a 1.50 × 1011 Sodium Phosphate 10 mM pH 7, [108] CB8-peptide complex, 27 ◦C Trp-Gly-Gly a 3.60 × 109 2 Tyr-Leu-Ala 1.39 × 108 Tyr-Lys-Ala 5.00 × 106 [37] Tyr-Phe-Ala 3.44 × 106 Tyr-Tyr-Ala 1.43 × 106 Met-Phe-Ala 7.14 × 106 ◦ Met-Tyr-Ala 4.00 × 106 Sodium Phosphate 10 mM pH 7, 27 C Met-Leu-Ala 1.39 × 106 Met-Lys-Ala 3.85 × 105 [123] Met-Tyr-Gly-Gly-Tyr 6.25 × 106 Met-Leu-Gly-Gly-Tyr 3.33 × 106 Leu-Met-Gly-Gly-Tyr 6.25 × 106 Met-Lys-Gly-Gly-Tyr 2.38 × 106 4 Gly2-Trp-Gly2 2.20 × 10 Sodium Phosphate 10 mM pH 7, (Gly -Trp-Gly ) 5.00 × 105 2 2 2 co-binding with peptide-MV [181] ◦ Asp -(Gly -Trp- conjugates, 27 C 2 2 4.70 × 106 Gly2)3-Asp2 Deuterium oxide, dihydrogenophosphate 1.20 × 103 Lys-Ala-Ala 25 mM pH 4.4 3 1.10 × 10 Deuterium oxide, Phosphate buffer 200 Lys-Lys-Leu-Val-Phe- 1.41 × 104 mM pH 7.6 Phe 3.80 × 104 MPnT 1H-NMR [24] Lys-Thr-Thr-Lys 5.50 × 103 Deuterium oxide, Sodium Lys-Thr-Thr-Lys-Ser 4.20 × 103 dihydrogenophosphate 25 mM pH 4.4 Gly-Arg-Gly-Gly 9.00 × 102 1.20 × 103 Arg-Gly-Asp 1.00 × 103 Deuterium oxide unbuffered Lys-Ala-Ala 3.33 × 104 Lys-Leu-Val-Phe-Phe 5.00 × 104 Lys-Lys-Leu- 2.50 × 105 MPT Val-Phe-Phe Phosphate buffer 200 mM pH 7.6 Lys-Lys-Leu-Val-Phe 1.43 × 105 -Phe-Ala-Lys Lys-Lys-Lys-Lys 1 × 105 Fluorescence [25] Spectroscopy Arg-Gly-Asp 1.16 × 104 Lys-Ala-Ala 3.30 × 103 MST Phosphate buffer 10 mM pH 7.6 Lys-Leu-Val-Phe-Phe 2.63 × 104 MCT Lys-Ala-Ala 30 Phosphate buffer 10 mM pH 7.2 Acetonitrile:Methanol (9:1) 2:1 host guest PBI Ala-Trp a 3.72 × 109 [153] complexes, 23 ◦C Molecules 2021, 26, 106 34 of 41

Table 2. Cont.

−1 Receptor Guest Ka (M ) Conditions Method Gly-Gly-Gly 2.10 × 102 3 CENMe3 Gly-Trp-Gly 2.15 × 10 Water, 25 ◦C [154] Gly.Phe-Gly 1.7 × 103 Gly-Gly-Phe 2.15 × 102 Gly-Leu 2.63 × 102 α-CDx Formic Acid 50%(v/v), 25 ◦C ITC [35] Gly-Val 2.82 × 102 Tyr-Ile-Gly-Ser-Arg 2.24 × 102 Phosphate Buffer 0.01 M pH 7.0, 20 ◦C Fluorescence [119] Tyr-Gly-Gly-Phe-Leu 1.23 × 102 Spectroscopy β-CDx cyclic peptide ◦ -Asp-Phe-D-Pro-Asp- 2.20 × 102 Bicarbonate buffer 0.2 M pH 9, 25 C [151] Phe-D-Pro- H3 (1–21) b 2.11 × 105 ◦ c 5 Tris Buffer 20 mM, pH 7.4, 25 C [159] DCv1 PKP 2.68 × 10 ITC MBP d 3.00 × 104

N-Ac -L-Lys-D- 2 1.02 × 105 Ala-D-Ala VD1 Water [161] N-Ac -L-Lys-D- 2 3.19 × 104 Ala-D-Lac 4 GP3KKF (D-Glu)4 2.37 × 10 Bis Tris Buffer 1.5 mM pH 6.10 [164] 6 Ac-Trp-Trp-Ala-NH2 >1.00 × 10 5 ◦ Pd6L4 Ac-Trp-Ala-Trp-NH2 2.50 × 10 Water, 20 C UV-Visible [179] 4 Spectroscopy Ac-Ala-Trp-Trp-NH2 2.10 × 10 FeII L Clavanin analogue e 1.11 × 105 8 6 [180] cage Acetonitrile:water (1:1) Clavanin A a,f 1.25 × 107 a 1:2 host-guest stoichiometry. Overall binding constant in M−2. The overall binding constant corresponds to the product of the stepwise b binding constants for the 1:1 and 1:2 complexes, i.e., K11.K12. Ala-Arg-Thr-Lys-Gln-Thr-Ala-Arg-Lys-Ser-Thr-Gly-Gly-Lys-Ala-Pro- Arg-Lys-Gln-Leu-Ala. c Gly-Arg-Thr-Gly-Arg-Arg-Asn-Ser-Ile. d Ala-Pro-Arg-Thr-Pro-Gly-Gly-Arg-Arg. e Sequence of the analogue: Ser-Ser-Trp-Gly-His-Val-Gly-Lys-Tyr-Val-His-Gly-Trp-Ser-His-Val-Ser. f Val-Phe-Gln-Phe-Leu-Gly-Lys-Ile-Ile-His-His-Val-Gly-Asn-Phe- Val-His-Gly-Phe-Ser-His-Val-Phe.

5. Conclusions and Outlook Despite the clear difficulty in the binding and recognition of the biological targets here presented, supramolecular systems begin to approach affinities and selectivities that are observable in nature, as is the case of protein–protein interactions, lock-and-key complementarity and the highly specific –antibody recognition. Furthermore, many of the receptors explored here already possess the ability to directly modify or detect their target analytes, despite the majority of them presenting simple, modular structures. This is the case of p-sulfonatocalix[4]arene and cucurbit[n]urils, which can modulate the properties of proteins, such as cytochrome c and Casp-8, respectively. Additionally, some of these receptors present either an intrinsic colorimetric or fluorometric response in the presence of the respective ligands, e.g., the tweezers MPT and MCT, making them especially promising in sensing applications. Moreover, these characteristics are not the only ones that can be explored, seeing as the receptors can also be integrated in more complex structures, as synthetic binding pockets, with the addition of other groups for the creation of new functionalities. Molecules 2021, 26, 106 35 of 41

The logical steps going forward are the continuous creation of new receptors, the eventual conjugation of several of these molecules for the multivalent binding of one complex analyte with several binding sights and smaller modifications and optimizations of the preexisting receptors, to enhance their selectivity and affinity towards one particular target ligand.

Funding: This research was funded by the Associate Laboratory for Green Chemistry—LAQV (UIDB/50006/2020), financed by FCT/MCTES. Data Availability Statement: No new data were created or analyzed in this study. Data sharing is not applicable to this article. Acknowledgments: N.B. acknowledges the FCT/MCTES for a research contract CEECIND/ 00466/2017. Conflicts of Interest: The authors declare no conflict of interest.

References 1. Camilli, A.; Bassler, B.L. Bacterial small-molecule signaling pathways. Science 2006, 311, 1113–1116. [CrossRef] 2. Sewald, N.; Jakubke, H. Peptides: Chemistry and , 2nd ed.; Wiley-VCH: Weinheim, Germany, 2009; ISBN 9783527318674. 3. Maity, D.; Schmuck, C. Synthetic Receptors for Biomolecules; Smith, B.D., Ed.; Royal Society of Chemistry: London, UK, 2015; ISBN 978-1-84973-971-9. 4. Broza, Y.Y.; Zhou, X.; Yuan, M.; Qu, D.; Zheng, Y.; Vishinkin, R.; Khatib, M.; Wu, W.; Haick, H. Disease Detection with Molecular Biomarkers: From Chemistry of Body Fluids to Nature-Inspired Chemical Sensors. Chem. Rev. 2019, 119, 11761–11817. [CrossRef] 5. Kruse, U.; Bantscheff, M.; Drewes, G.; Hopf, C. Chemical and Pathway Proteomics. Mol. Cell. Proteom. 2008, 7, 1887– 1901. [CrossRef] 6. Urban, G.; Piletsky, S.A.; Whitcombe, M.J. (Eds.) Designing Receptors for the Next Generation of Biosensors; Springer: Berlin/Heidelberg, Germany, 2013; Volume 11, ISBN 978-3-642-28093-1. 7. Kubota, R.; Hamachi, I. Protein recognition using synthetic small-molecular binders toward optical protein sensing in vitro and in live cells. Chem. Soc. Rev. 2015, 44, 4454–4471. [CrossRef] 8. Peczuh, M.W.; Hamilton, A.D. Peptide and Protein Recognition by Designed Molecules. Chem. Rev. 2000, 100, 2479–2494. [CrossRef] 9. Van Dun, S.; Ottmann, C.; Milroy, L.G.; Brunsveld, L. Supramolecular Chemistry Targeting Proteins. J. Am. Chem. Soc. 2017, 139, 13960–13968. [CrossRef] 10. Park, H.S.; Lin, Q.; Hamilton, A.D. Protein Surface Recognition by Synthetic Receptors: A Route to Novel Submicromolar Inhibitors for α-Chymotrypsin. J. Am. Chem. Soc. 1999, 121, 8–13. [CrossRef] 11. Hatai, J.; Schmuck, C. Diverse Properties of Guanidiniocarbonyl Pyrrole-Based Molecules: Artificial Analogues of Arginine. Acc. Chem. Res. 2019, 52, 1709–1720. [CrossRef] 12. Tabet, M.; Labroo, V.; Sheppard, P.; Sasaki, T. Spermine-Induced Conformational Changes of a Synthetic Peptide. J. Am. Chem. Soc. 1993, 115, 3866–3868. [CrossRef] 13. Tanaka, A.; Fujiyoshi, S.; Motomura, K.; Hayashida, O.; Hisaeda, Y.; Murakami, Y. Preparation and host-guest interactions of novel cage-type cyclophanes bearing chiral binding sites provided by dipeptide residues. Tetrahedron 1998, 54, 5187–5206. [CrossRef] 14. Yoon, S.S.; Still, W.C. An Exceptional Synthetic Receptor for Peptides. J. Am. Chem. Soc. 1993, 115, 823–824. [CrossRef] 15. Rick, W. Methods of Enzymatic Analysis, 2nd ed.; Bergmeyer, H.U., Ed.; Academic Press: Cambridge, MA, USA, 1974; Volume 2. 16. Whetstine, J.R. Histone Methylation: Chemically Inert but Chromatin Dynamic, 2nd ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2010; Volume 3, ISBN 9780123741455. 17. Erce, M.A.; Abeygunawardena, D.; Low, J.K.K.; Hart-Smith, G.; Wilkins, M.R. Interactions affected by arginine methylation in the yeast protein-protein interaction network. Mol. Cell. Proteom. 2013, 12, 3184–3198. [CrossRef][PubMed] 18. Graveley, B.R.; Maniatis, T. Arginine/Serine-Rich Domains of SR Proteins Can Function as Activators of Pre-mRNA Splicing splicing of all metazoan introns and therefore play a central role in the basic splicing reaction. One function of SR proteins is to promote the binding of U1 snRN. Mol. Cell 1998, 1, 765–771. [CrossRef] 19. Klumpp, L.M.; Mackey, A.T.; Farrell, C.M.; Rosenberg, J.M.; Gilbert, S.P. A kinesin switch I arginine to lysine mutation rescues microtubule function. J. Biol. Chem. 2003, 278, 39059–39067. [CrossRef] 20. Vazdar, M.; Heyda, J.; Mason, P.E.; Tesei, G.; Allolio, C.; Lund, M.; Jungwirth, P. Arginine “magic”: Guanidinium Like-Charge Ion Pairing from Aqueous Salts to Cell Penetrating Peptides. Acc. Chem. Res. 2018, 51, 1455–1464. [CrossRef] 21. Douteau-Guével, N.; Coleman, A.W.; Morel, J.P.; Morel-Desrosiers, N. Complexation of the basic amino acids lysine and arginine by three sulfonatocalix [n] arenes (n = 4, 6 and 8) in water: Microcalorimetric determination of the Gibbs energies, enthalpies and entropies of complexation. J. Chem. Soc. Perkin Trans. 2 1999, 629–633. [CrossRef] 22. Douteau-Guével, N.; Perret, F.; Coleman, A.W.; Morela, J.P.; Morel-Desrosiers, N. Binding of dipeptides and tripeptides containing lysine or arginine by p-sulfonatocalixarenes in water: NMR and microcalorimetric studies. J. Chem. Soc. Perkin Trans. 2 2002, 524–532. [CrossRef] Molecules 2021, 26, 106 36 of 41

23. Lazar, A.; Da Silva, E.; Navaza, A.; Barbey, C.; Coleman, A.W. A new packing motif for para-sulfonatocalix[4]arene: The solid state structure of the para-sulfonatocalix[4]arene D-arginine complex. Chem. Commun. 2004, 10, 2162–2163. [CrossRef] 24. Fokkens, M.; Schrader, T.; Klärner, F.G. A molecular tweezer for lysine and arginine. J. Am. Chem. Soc. 2005, 127, 14415– 14421. [CrossRef] 25. Schrader, T.; Bitan, G.; Klärner, F.G. Molecular tweezers for lysine and arginine-powerful inhibitors of pathologic protein aggregation. Chem. Commun. 2016, 52, 11318–11334. [CrossRef] 26. Li, C.; Ma, J.; Zhao, L.; Zhang, Y.; Yu, Y.; Shu, X.; Li, J.; Jia, X. Molecular selective binding of basic amino acids by a water-soluble pillar[5]arene. Chem. Commun. 2013, 49, 1924–1926. [CrossRef][PubMed] 27. Liao, S.-M.; Du, Q.-S.; Meng, J.-Z.; Pang, Z.-W.; Huang, R.-B. The multiple roles of histidine in protein interactions. Chem. Cent. J. 2013, 7, 1–12. [CrossRef][PubMed] 28. Da Silva, E.; Coleman, A.W. Synthesis and complexation properties towards amino acids of mono-substituted p-sulphonato-calix- [n]-arenes. Tetrahedron 2003, 59, 7357–7364. [CrossRef] 29. Bogan, A.A.; Thorn, K.S. Anatomy of hot spots in protein interfaces. J. Mol. Biol. 1998, 280, 1–9. [CrossRef] 30. Cordes, M.; Köttgen, A.; Jasper, C.; Jacques, O.; Boudebous, H.; Giese, B. Influence of amino acid side chains on long-distance electron transfer in peptides: Electron hopping via “stepping stones”. Angew. Chem. Int. Ed. 2008, 47, 3461–3463. [CrossRef] 31. Giese, B.; Eckhardt, S.; Lauz, M. Electron Transfer in Peptides and Proteins. Encycl. Radic. Chem. Biol. Mater. 2012.[CrossRef] 32. Li, Q.; Guo, Y.; Shao, S. A BODIPY based fluorescent chemosensor for Cu(II) ions and homocysteine/cysteine. Sens. Actuators B Chem. 2012, 171–172, 872–877. [CrossRef] 33. Chan, D.I.; Prenner, E.J.; Vogel, H.J. Tryptophan- and arginine-rich antimicrobial peptides: Structures and mechanisms of action. Biochim. Biophys. Acta Biomembr. 2006, 1758, 1184–1202. [CrossRef] 34. Lakowicz, J.R. Principles of Fluorescence Spectroscopy; Springer: New York, NY, USA, 2006. 35. Buschmann, H.J.; Schollmeyer, E.; Mutihac, L. The formation of amino acid and dipeptide complexes with α-cyclodextrin and cucurbit[6]uril in aqueous solutions studied by titration calorimetry. Thermochim. Acta 2003, 399, 203–208. [CrossRef] 36. Rekharsky, M.V.; Mori, T.; Yang, C.; Young, H.K.; Selvapalam, N.; Kim, H.; Sobransingh, D.; Kaifer, A.E.; Liu, S.; Isaacs, L.; et al. A synthetic host-guest system achieves avidin-biotin affinity by overcoming enthalpy—Entropy compensation. Proc. Natl. Acad. Sci. USA 2007, 104, 20737–20742. [CrossRef] 37. Bush, M.E.; Bouley, N.D.; Urbach, A.R. Charge-Mediated Recognition of N-Terminal Tryptophan in Aqueous Solution by a Synthetic Host. J. Am. Chem. Soc. 2005, 127, 14511–14517. [CrossRef] 38. Ling, Y.; Wang, W.; Kaifer, A.E. A new cucurbit[8]uril-based fluorescent receptor for indole derivatives. Chem. Commun. 2007, 610–612. [CrossRef] 39. Biedermann, F.; Rauwald, U.; Cziferszky, M.; Williams, K.A.; Gann, L.D.; Guo, B.Y.; Urbach, A.R.; Bielawski, C.W.; Scherman, O.A. Benzobis(imidazolium)-cucurbit[8]uril complexes for binding and sensing aromatic compounds in aqueous solution. Chem. A Eur. J. 2010, 16, 13716–13722. [CrossRef] 40. Lagona, J.; Wagner, B.D.; Isaacs, L. Molecular-recognition properties of a water-soluble cucurbit[6]uril analogue. J. Org. Chem. 2006, 71, 1181–1190. [CrossRef] 41. Stepniak, P.; Lainer, B.; Chmurski, K.; Jurczak, J. The effect of urea moiety in amino acid binding by β-cyclodextrin derivatives: A 1000-fold increase in efficacy comparing to native β-cyclodextrin. Carbohydr. Polym. 2017, 164, 233–241. [CrossRef] 42. Cheng, L.; Zhang, H.; Dong, Y.; Zhao, Y.; Yu, Y.; Cao, L. Tetraphenylethene-based tetracationic cyclophanes and their selective recognition for amino acids and adenosine derivatives in water. Chem. Commun. 2019, 55, 2372–2375. [CrossRef] 43. Kyne, G.M.; Light, M.E.; Hursthouse, M.B.; De Mendoza, J.; Kilburn, J.D. Enantioselective amino acid recognition using acyclic thiourea receptors. J. Chem. Soc. Perkin Trans. 1 2001, 1258–1263. [CrossRef] 44. Urbach, A.R.; Ramalingam, V. Molecular recognition of amino acids, peptides, and proteins by cucurbit [n] uril receptors. Isr. J. Chem. 2011, 51, 664–678. [CrossRef] 45. Bailey, D.M.; Hennig, A.; Uzunova, V.D.; Nau, W.M. Supramolecular tandem enzyme assays for multiparameter sensor arrays and enantiomeric excess determination of amino acids. Chem. A Eur. J. 2008, 14, 6069–6077. [CrossRef] 46. Petroff, O.A.C. GABA and glutamate in the human brain. Neuroscientist 2002, 8, 562–573. [CrossRef] 47. Grabowska, A.; Nowicki, M.; Kwinta, J. Glutamate dehydrogenase of the germinating triticale seeds: expression, activity distribution and kinetic characteristics. Acta Physiol. Plant 2011, 33, 1981–1990. [CrossRef] 48. Barmore, W.; Stone, W.L. Physiology, Urea Cycle. 2019. Available online: https://europepmc.org/article/NBK/NBK513323 (accessed on 20 July 2020). 49. Digerness, S.B.; Reddy, W.J. The malate-aspartate shuttle in heart mitochondria. J. Mol. Cell. Cardiol. 1976, 8, 779–785. [CrossRef] 50. Cox, M.; Nelson, D.L. Lehninger Principles of Biochemistry; W.H. Freeman & Company: New York, NY, USA, 2008; ISBN 0716743396. 51. Duan, Q.; Zhao, W.; Lu, K. Synthesis of a water-soluble pillar[6]arene dodecaamine and its selective binding of acidic amino acids in water. Tetrahedron Lett. 2017, 58, 4403–4406. [CrossRef] 52. Wang, K.; Cui, J.H.; Xing, S.Y.; Ren, X.W. Selective Recognition of Acidic Amino Acids in Water by Calixpyridinium. Asian J. Org. Chem. 2017, 6, 1385–1389. [CrossRef] 53. Alfonso, I.; Rebolledo, F.; Gotor, V. Optically active dioxatetraazamacrocycles: Chemoenzymatic syntheses and applications in chiral anion recognition. Chem. A Eur. J. 2000, 6, 3331–3338. [CrossRef] 54. Alfonso, I.; Solà, J. Molecular Recognition of Zwitterions with Artificial Receptors. Chem. Asian J. 2020, 15, 986–994. [CrossRef] Molecules 2021, 26, 106 37 of 41

55. Brockhausen, I.; Stanley, P. O-GalNAc Glycans. In Essentials of Glycobiology; Varki, A., Cummings, R.D., Esko, J.D., Stanley, P., Hart, G.W., Aebi, M., Darvill, A.G., Kinoshita, T., Packer, N.H., Prestegard, J.H., et al., Eds.; Cold Spring Harbor: New York, NY, USA, 2017. 56. Stanley, P.; Taniguchi, N.; Aebi, M. N-Glycans. In Essentials of Glycobiology; Varki, A., Cummings, R.D., Esko, J.D., Stanley, P., Hart, G.W., Aebi, M., Darvill, A.G., Kinoshita, T., Packer, N.H., Prestegard, J.H., et al., Eds.; Cold Spring Harbor: New York, NY, USA, 2017. 57. Guyton, A.C.; Hall, J.E. Textbook of Medical Physiology, 11th ed.; Guyton, A.C., Hall, J.E., Eds.; Elsevier Saunders: Philadelphia, PA, USA, 2006. 58. Mailloux, R.J.; Jin, X.; Willmore, W.G. Redox regulation of mitochondrial function with emphasis on cysteine oxidation reactions. Redox Biol. 2014, 2, 123–139. [CrossRef] 59. Schmuck, C.; Geiger, L. Efficient complexation of N-acetyl amino acid carboxylates in water by an artificial receptor: Unexpected cooperativity in the binding of glutamate but not aspartate. J. Am. Chem. Soc. 2005, 127, 10486–10487. [CrossRef] 60. Liu, Y.; Lv, X.; Hou, M.; Shi, Y.; Guo, W. Selective Fluorescence Detection of Cysteine over Homocysteine and Glutathione Based on a Cysteine-Triggered Dual Michael Addition/Retro-aza-aldol Cascade Reaction. Anal. Chem. 2015, 87, 11475–11483. [CrossRef] 61. Peck, E.M.; Bradley, D. Smith Applications of Synthetic Receptors for Biomolecules. In Synthetic Receptors for Biomolecules; Smith, B.D., Ed.; Royal Society of Chemistry: Cambridge, UK, 2015; pp. 1–34. 62. Pinalli, R.; Pedrini, A.; Dalcanale, E. Biochemical sensing with macrocyclic receptors. Chem. Soc. Rev. 2018, 47, 7006–7026. [CrossRef] 63. Shinkai, S.; Araki, K.; Matsuda, T.; Nishiyama, N.; Ikeda, H.; Takasu, I.; Iwamoto, M. NMR and crystallographic studies of a p-sulfonatocalix[4]arene-guest complex. J. Am. Chem. Soc. 1990, 112, 9053–9058. [CrossRef] 64. Shinkai, S.; Araki, K.; Manabe, O. NMR determination of association constants for calixarene complexes. Evidence for the formation of a 1:2 complex with calix[8]arene. J. Am. Chem. Soc. 1988, 110, 7214–7215. [CrossRef] 65. Shinkai, S.; Mori, S.; Koreishi, H.; Tsubaki, T.; Manabe, O. Hexasulfonated calix[6]arene derivatives: A new class of catalysts, surfactants, and host molecules. J. Am. Chem. Soc. 1986, 108, 2409–2416. [CrossRef][PubMed] 66. Shinkai, S. Calixarenes as new functionalized host molecules. Pure Appl. Chem. 1986, 58, 1523–1528. [CrossRef] 67. Dalgarno, S.J.; Atwood, J.L.; Raston, C.L. Sulfonatocalixarenes: Molecular capsule and ‘Russian doll’ arrays to structures mimicking viral geometry. Chem. Commun. 2006, 4567–4574. [CrossRef] 68. Shinkai, S.; Mori, S.; Tsubaki, T.; Sone, T.; Manabe, O. New water-soluble host molecules derived from calix[6]arene. Tetrahedron Lett. 1984, 25, 5315–5318. [CrossRef] 69. Coleman, A.W.; Jebors, S.; Cecillon, S.; Perret, P.; Garin, D.; Marti-Battle, D.; Moulin, M. Toxicity and biodistribution of para-sulfonato-calix[4]arene in mice. New J. Chem. 2008, 32, 780. [CrossRef] 70. Perret, F.; Coleman, A.W. Biochemistry of anionic calix [n] arenes. Chem. Commun. 2011, 47, 7303. [CrossRef] 71. Perret, F.; Lazar, A.N.; Coleman, A.W. Biochemistry of the para-sulfonato-calix [n] arenes. Chem. Commun. 2006, 2425– 2438. [CrossRef] 72. Hof, F. Host-guest chemistry that directly targets lysine methylation: Synthetic host molecules as alternatives to bio-reagents. Chem. Commun. 2016, 52, 10093–10108. [CrossRef] 73. Guo, D.-S.; Liu, Y. Supramolecular Chemistry of p -Sulfonatocalix [n] arenes and Its Biological Applications. Acc. Chem. Res. 2014, 47, 1925–1934. [CrossRef] 74. Ghale, G.; Nau, W.M. Dynamically Analyte-Responsive Macrocyclic Host–Fluorophore Systems. Acc. Chem. Res. 2014, 47, 2150–2159. [CrossRef] 75. Douteau-Guével, N.; Coleman, A.W.; Morel, J.; Morel-Desrosiers, N. Complexation of basic amino acids by water-soluble calixarene sulphonates as a study of the possiblemechanisms of recognition of calixarene sulphonates by proteins. J. Phys. Org. Chem. 1998, 11, 693–696. [CrossRef] 76. Selkti, M.; Coleman, A.W.; Nicolis, I.; Douteau-Guével, N.; Villain, F.; Tomas, A.; Rango, C. The first example of a substrate spanning the calix[4]arene bilayer: The solid state complex of p-sulfonatocalix[4]arene with L-lysine. Chem. Commun. 1999, 161–162. [CrossRef] 77. Bakirci, H.; Koner, A.L.; Nau, W.M. Binding of inorganic cations by p-sulfonatocalix[4]arene monitored through competitive fluorophore displacement in aqueous solution. Chem. Commun. 2005, 5411–5413. [CrossRef] 78. Bakirci, H.; Koner, A.L.; Schwarzlose, T.; Nau, W.M. Analysis of Host-Assisted Guest Protonation Exemplified forp- Sulfonatocalix[4]arene—Towards Enzyme-Mimetic pKa Shifts. Chem. Eur. J. 2006, 12, 4799–4807. [CrossRef] 79. Francisco, V.; Piñeiro, A.; Nau, W.M.; García-Río, L. The “True” Affinities of Metal Cations to p-Sulfonatocalix[4]arene: A Thermodynamic Study at Neutral pH Reveals a Pitfall Due to Salt Effects in Microcalorimetry. Chem. A Eur. J. 2013, 19, 17809–17820. [CrossRef] 80. Garcia-Rio, L.; Basílio, N.; Francisco, V. Counterion effect on sulfonatocalix [n] arene recognition. Pure Appl. Chem. 2019.[CrossRef] 81. Lavande, N.; Acuña, A.; Basílio, N.; Francisco, V.; Malkhede, D.D.; Garcia-Rio, L. A journey from calix[4]arene to calix[6]and calix[8]arene reveals more than a matter of size. Receptor concentration affects the stability and stoichiometric nature of the complexes. Phys. Chem. Chem. Phys. 2017, 19, 13640–13649. [CrossRef] 82. Pessêgo, M.; Basílio, N.; Muñiz, M.C.; García-Río, L. Competitive counterion complexation allows the true host: Guest binding constants from a single titration by ionic receptors. Org. Biomol. Chem. 2016, 14, 6442–6448. [CrossRef] Molecules 2021, 26, 106 38 of 41

83. Francisco, V.; Basílio, N.; García-Río, L. Ionic Exchange in p-Sulfonatocalix[4]arene-Mediated Formation of Metal–Ligand Complexes. J. Phys. Chem. B 2014, 118, 4710–4716. [CrossRef] 84. Basilio, N.; García-Río, L.; Martín-Pastor, M. Counterion Binding in Solutions of p-Sulfonatocalix[4]arene. J. Phys. Chem. B 2010, 114, 7201–7206. [CrossRef] 85. Francisco, V.; Basilio, N.; Garciía-Riío, L. Counterion exchange as a decisive factor in the formation of host: Guest complexes by p-sulfonatocalix[4]arene. J. Phys. Chem. B 2012, 116, 5308–5315. [CrossRef] 86. Romero, M.A.; Mateus, P.; Matos, B.; Acuña, Á.; García-Río, L.; Arteaga, J.F.; Pischel, U.; Basílio, N. Binding of Flavylium Ions to Sulfonatocalix[4]arene and Implication in the Photorelease of Biologically Relevant Guests in Water. J. Org. Chem. 2019, 84, 10852–10859. [CrossRef] 87. Guo, D.S.; Uzunova, V.D.; Su, X.; Liu, Y.; Nau, W.M. Operational calixarene-based fluorescent sensing systems for choline and acetylcholine and their application to enzymatic reactions. Chem. Sci. 2011, 2, 1722–1734. [CrossRef] 88. Nau, W.M.; Ghale, G.; Hennig, A.; Bakirci, H.; Bailey, D.M. Substrate-selective supramolecular tandem assays: Monitoring enzyme inhibition of arginase and diamine oxidase by fluorescent dye displacement from calixarene and cucurbituril macrocycles. J. Am. Chem. Soc. 2009, 131, 11558–11570. [CrossRef] 89. Beshara, C.S.; Jones, C.E.; Daze, K.D.; Lilgert, B.J.; Hof, F. A simple calixarene recognizes post-translationally methylated lysine. ChemBioChem 2010, 11, 63–66. [CrossRef] 90. Daze, K.D.; Ma, M.C.F.; Pineux, F.; Hof, F. Synthesis of New Trisulfonated Calix[4]arenes Functionalized at the Upper Rim, and Their Complexation with the Trimethyllysine Epigenetic Mark. Org. Lett. 2012, 14, 1512–1515. [CrossRef] 91. Ogoshi, T.; Hashizume, M.; Yamagishi, T.A.; Nakamoto, Y. Synthesis, conformational and host-guest properties of water-soluble pillar[5]arene. Chem. Commun. 2010, 46, 3708–3710. [CrossRef] 92. Murray, J.; Kim, K.; Ogoshi, T.; Yao, W.; Gibb, B.C. The aqueous supramolecular chemistry of cucurbit [n] urils, pillar [n] arenes and deep-cavity cavitands. Chem. Soc. Rev. 2017, 46, 2479–2496. [CrossRef] 93. Ogoshi, T.; Yamagishi, T.; Nakamoto, Y. Pillar-Shaped Macrocyclic Hosts Pillar [n] arenes: New Key Players for Supramolecular Chemistry. Chem. Rev. 2016, 116, 7937–8002. [CrossRef][PubMed] 94. Bojtár, M.; Paudics, A.; Hessz, D.; Kubinyi, M.; Bitter, I. Amino acid recognition by fine tuning the association constants: Tailored naphthalimides in pillar[5]arene-based indicator displacement assays. RSC Adv. 2016, 6, 86269–86275. [CrossRef] 95. Bastug, E.; Kursunlu, A.N.; Guler, E. A fluorescent clever macrocycle: Deca-bodipy bearing a pillar[5]arene and its selective binding of asparagine in half-aqueous medium. J. Lumin. 2020, 225, 117343. [CrossRef] 96. Assaf, K.I.; Nau, W.M. Cucurbiturils: From synthesis to high-affinity binding and catalysis. Chem. Soc. Rev. 2015, 44, 394–418. [CrossRef][PubMed] 97. Barrow, S.J.; Kasera, S.; Rowland, M.J.; del Barrio, J.; Scherman, O.A. Cucurbituril-Based Molecular Recognition. Chem. Rev. 2015, 115, 12320–12406. [CrossRef][PubMed] 98. Masson, E.; Ling, X.; Joseph, R.; Kyeremeh-Mensah, L.; Lu, X. Cucurbituril chemistry: A tale of supramolecular success. RSC Adv. 2012, 2, 1213–1247. [CrossRef] 99. Deng, C.-L.; Murkli, S.L.; Isaacs, L.D. Supramolecular hosts as in vivo sequestration agents for pharmaceuticals and toxins. Chem. Soc. Rev. 2020, 49, 7516–7532. [CrossRef] 100. Shetty, D.; Khedkar, J.K.; Park, K.M.; Kim, K. Can we beat the biotin–avidin pair? Cucurbit[7]uril-based ultrahigh affinity host–guest complexes and their applications. Chem. Soc. Rev. 2015, 44, 8747–8761. [CrossRef] 101. Gamal-Eldin, M.A.; MacArtney, D.H. Selective molecular recognition of methylated lysines and arginines by cucurbit[6]uril and cucurbit[7]uril in aqueous solution. Org. Biomol. Chem. 2013, 11, 488–495. [CrossRef] 102. Lee, J.W.; Lee, H.H.L.; Ko, Y.H.; Kim, K.; Kim, H.I. Deciphering the Specific High-Affinity Binding of Cucurbit[7]uril to Amino Acids in Water. J. Phys. Chem. B 2015, 119, 4628–4636. [CrossRef] 103. Liu, S.; Ruspic, C.; Mukhopadhyay, P.; Chakrabarti, S.; Zavalij, P.Y.; Isaacs, L. The Cucurbit [n] uril Family: Prime Components for Self-Sorting Systems. J. Am. Chem. Soc. 2005, 127, 15959–15967. [CrossRef][PubMed] 104. Romero, M.A.; Basílio, N.; Moro, A.J.; Domingues, M.; González-Delgado, J.A.; Arteaga, J.F.; Pischel, U. Photocaged Competitor Guests: A General Approach Toward Light-Activated Cargo Release From Cucurbiturils. Chem. A Eur. J. 2017, 23, 13105– 13111. [CrossRef] 105. Biedermann, F.; Uzunova, V.D.; Scherman, O.A.; Nau, W.M.; De Simone, A. Release of High-Energy Water as an Essential Driving Force for the High-Affinity Binding of Cucurbit [n] urils. J. Am. Chem. Soc. 2012, 134, 15318–15323. [CrossRef] 106. Biedermann, F.; Vendruscolo, M.; Scherman, O.A.; De Simone, A.; Nau, W.M. Cucurbit[8]uril and blue-box: High-energy water release overwhelms electrostatic interactions. J. Am. Chem. Soc. 2013, 135, 14879–14888. [CrossRef] 107. Biedermann, F.; Nau, W.M.; Schneider, H.-J. The Hydrophobic Effect Revisited-Studies with Supramolecular Complexes Imply High-Energy Water as a Noncovalent Driving Force. Angew. Chem. Int. Ed. 2014, 53, 11158–11171. [CrossRef] 108. Heitmann, L.M.; Taylor, A.B.; Hart, P.J.; Urbach, A.R. Sequence-specific recognition and cooperative dimerization of N-terminal aromatic peptides in aqueous solution by a synthetic host. J. Am. Chem. Soc. 2006, 128, 12574–12581. [CrossRef] 109. Liu, Y.-H.; Zhang, Y.-M.; Yu, H.-J.; Liu, Y. Cucurbituril-Based Biomacromolecular Assemblies. Angew. Chem. Int. Ed. 2020, 1–11. [CrossRef] 110. Rajgariah, P.; Urbach, A.R. Scope of amino acid recognition by cucurbit[8]uril. J. Incl. Phenom. Macrocycl. Chem. 2008, 62, 251–254. [CrossRef] Molecules 2021, 26, 106 39 of 41

111. Saenger, W.; Steiner, T. Cyclodextrin Inclusion Complexes: Host-Guest Interactions and Hydrogen-Bonding Networks. Acta Crystallogr. Sect. A Found. Crystallogr. 1998, 54, 798–805. [CrossRef] 112. Moran, J.R.; Karbach, S.; Cram, D.J. Cavitands: Synthetic Molecular Vessels. J. Am. Chem. Soc. 1982, 104, 5826–5828. [CrossRef] 113. Biavardi, E.; Tudisco, C.; Maffei, F.; Motta, A.; Massera, C.; Condorelli, G.G.; Dalcanale, E. Exclusive recognition of sarcosine in water and urine by a cavitand-functionalized silicon surface. Proc. Natl. Acad. Sci. USA 2012, 109, 2263–2268. [CrossRef] 114. Brancatelli, G.; Dalcanale, E.; Pinalli, R.; Geremia, S. Probing the structural determinants of amino acid recognition: X-ray studies of crystalline ditopic host-guest complexes of the positively charged amino acids, ARg, Lys, and His with a cavitand molecule. Molecules 2018, 23, 3368. [CrossRef] 115. Pinalli, R.; Brancatelli, G.; Pedrini, A.; Menozzi, D.; Hernández, D.; Ballester, P.; Geremia, S.; Dalcanale, E. The Origin of Selectivity in the Complexation of N -Methyl Amino Acids by Tetraphosphonate Cavitands. J. Am. Chem. Soc. 2016, 138, 8569–8580. [CrossRef] 116. Goodnow, T.T.; Reddington, M.V.; Stoddart, J.F.; Kaifer, A.E. Cyclobis(paraquat-p-phenylene): A Novel Synthetic Receptor for Amino Acids with Electron-Rich Aromatic Moieties. J. Am. Chem. Soc. 1991, 113, 4335–4337. [CrossRef] 117. Schmuck, C. Carboxylate Binding by 2-(Guanidiniocarbonyl) pyrrole Receptors in Aqueous Solvents: Improving the Binding Properties of Guanidinium Cations through Additional Hydrogen Bonds. Chem. A Eur. J. 2000, 6, 709–718. [CrossRef] 118. Mandl, C.P.; König, B. Luminescent crown ether amino acids: Selective binding to N-terminal lysine in peptides. J. Org. Chem. 2005, 70, 670–674. [CrossRef] 119. Bekos, E.J.; Gardella, J.A.; Bright, F.V. The binding of free oligopeptides to cyclodextrins: The role of the tyrosine group. J. Incl. Phenom. Mol. Recognit. Chem. 1996, 26, 185–195. [CrossRef] 120. Stepniak, P.; Lainer, B.; Chmurski, K.; Jurczak, J. pH-Controlled recognition of amino acids by urea derivatives of β-cyclodextrin. RSC Adv. 2017, 7, 15742–15746. [CrossRef] 121. Barba-Bon, A.; Pan, Y.; Biedermann, F.; Guo, D.; Nau, W.M.; Hennig, A. Fluorescence Monitoring of Peptide Transport Pathways into Large and Giant Vesicles by Supramolecular Host–Dye Reporter Pairs. J. Am. Chem. Soc. 2019, 141, 20137–20145. [CrossRef] 122. Rekharsky, M.V.; Yamamura, H.; Ko, Y.H.; Selvapalam, N.; Kim, K.; Inoue, Y. Sequence recognition and self-sorting of a dipeptide by cucurbit[6]uril and cucurbit[7]uril. Chem. Commun. 2008, 2236–2238. [CrossRef] 123. Hirani, Z.; Taylor, H.F.; Babcock, E.F.; Bockus, A.T.; Varnado, C.D.; Bielawski, C.W.; Urbach, A.R. Molecular Recognition of Methionine-Terminated Peptides by Cucurbit[8]uril. J. Am. Chem. Soc. 2018, 140, 12263–12269. [CrossRef] 124. Chinai, J.M.; Taylor, A.B.; Ryno, L.M.; Hargreaves, N.D.; Morris, C.A.; Hart, P.J.; Urbach, A.R. Molecular recognition of insulin by a synthetic receptor. J. Am. Chem. Soc. 2011, 133, 8810–8813. [CrossRef] 125. Smith, L.C.; Leach, D.G.; Blaylock, B.E.; Ali, O.A.; Urbach, A.R. Sequence-Specific, Nanomolar Peptide Binding via Cucurbit[8]uril- Induced Folding and Inclusion of Neighboring Side Chains. J. Am. Chem. Soc. 2015, 137, 3663–3669. [CrossRef] 126. Schneider, H.J. Binding mechanisms in supramolecular complexes. Angew. Chem. Int. Ed. 2009, 48, 3924–3977. [CrossRef] 127. Ghale, G.; Lanctôt, A.G.; Kreissl, H.T.; Jacob, M.H.; Weingart, H.; Winterhalter, M.; Nau, W.M. Chemosensing ensembles for monitoring biomembrane transport in real time. Angew. Chem. Int. Ed. 2014, 53, 2762–2765. [CrossRef] 128. Balhorn, R. The protamine family of sperm nuclear proteins. Genome Biol. 2007, 8.[CrossRef] 129. Voigt, A.; Jelinek, H.F. Humanin: A mitochondrial signaling peptide as a biomarker for impaired fasting glucose-related oxidative stress. Physiol. Rep. 2016, 4, 1–5. [CrossRef] 130. Memmi, L.; Lazar, A.; Brioude, A.; Ball, V.; Coleman, A.W. Protein-calixarene interactions: Complexation of Bovine Serum Albumin by sulfonatocalix [n] arenes. Chem. Commun. 2001, 2, 2474–2475. [CrossRef] 131. McGovern, R.E.; Fernandes, H.; Khan, A.R.; Power, N.P.; Crowley, P.B. Protein camouflage in cytochrome c-calixarene complexes. Nat. Chem. 2012, 4, 527–533. [CrossRef] 132. McGovern, R.E.; McCarthy, A.A.; Crowley, P.B. Protein assembly mediated by sulfonatocalix[4]arene. Chem. Commun. 2014, 50, 10412–10415. [CrossRef] 133. Rennie, M.L.; Fox, G.C.; Pérez, J.; Crowley, P.B. Auto-regulated Protein Assembly on a Supramolecular Scaffold. Angew. Chem. Int. Ed. 2018, 57, 13764–13769. [CrossRef] 134. Rennie, M.L.; Crowley, P.B. A Thermodynamic Model of Auto-regulated Protein Assembly by a Supramolecular Scaffold. ChemPhysChem 2019, 20, 1011–1017. [CrossRef] 135. Bhasikuttan, A.C.; Mohanty, J. Detection, inhibition and disintegration of amyloid fibrils: The role of optical probes and macrocyclic receptors. Chem. Commun. 2017, 53, 2789–2809. [CrossRef] 136. Nguyen, B.T.; Anslyn, E.V. Indicator—Displacement assays. Coord. Chem. Rev. 2006, 250, 3118–3127. [CrossRef] 137. Cavatorta, E.; Jonkheijm, P.; Huskens, J. Assessment of Cooperativity in Ternary Peptide-Cucurbit[8]uril Complexes. Chem. A Eur. J. 2017, 23, 4046–4050. [CrossRef] 138. Romero, M.A.; Fernandes, R.J.; Moro, A.J.; Basílio, N.; Pischel, U. Light-induced cargo release from a cucurbit[8]uril host by means of a sequential logic operation. Chem. Commun. 2018, 54, 13335–13338. [CrossRef] 139. Giglione, C.; Boularot, A.; Meinnel, T. Protein N-terminal methionine excision. Cell. Mol. Life Sci. 2004, 61, 1455–1474. [CrossRef] 140. Wu, G.; Clarke, D.E.; Wu, C.; Scherman, O.A. Oligopeptide-CB[8]complexation with switchable binding pathways. Org. Biomol. Chem. 2019.[CrossRef] 141. Lee, J.W.; Park, M.H.; Ju, J.T.; Choi, Y.S.; Hwang, S.M.; Jung, D.J.; Kim, H.I. Stabilization of compact protein structures by macrocyclic hosts cucurbit [n] urils in the gas phase. Mass Spectrom. Lett. 2016, 7, 16–20. [CrossRef] Molecules 2021, 26, 106 40 of 41

142. De Vink, P.J.; Briels, J.M.; Schrader, T.; Milroy, L.-G.; Brunsveld, L.; Ottmann, C. A Binary Bivalent Platform Based on Cucurbit[8]uril and Dimeric Adapter Protein 14-3-3. Angew. Chem. Int. Ed. 2017, 56, 8998–9002. [CrossRef] 143. Lee, H.H.; Choi, T.S.; Lee, S.J.C.; Lee, J.W.; Park, J.; Ko, Y.H.; Kim, W.J.; Kim, K.; Kim, H.I. Supramolecular inhibition of amyloid fibrillation by cucurbit[7]uril. Angew. Chem. Int. Ed. 2014, 53, 7461–7465. [CrossRef] 144. Webber, M.J.; Appel, E.A.; Vinciguerra, B.; Cortinas, A.B.; Thapa, L.S.; Jhunjhunwala, S.; Isaacs, L.; Langer, R.; Anderson, D.G. Supramolecular PEGylation of biopharmaceuticals. Proc. Natl. Acad. Sci. USA 2016, 113, 14189–14194. [CrossRef] 145. Maikawa, C.L.; Smith, A.A.A.; Zou, L.; Roth, G.A.; Gale, E.C.; Stapleton, L.M.; Baker, S.W.; Mann, J.L.; Yu, A.C.; Correa, S.; et al. A co-formulation of supramolecularly stabilized insulin and pramlintide enhances mealtime glucagon suppression in diabetic pigs. Nat. Biomed. Eng. 2020, 4, 507–517. [CrossRef] 146. Wang, R.; Qiao, S.; Zhao, L.; Hou, C.; Li, X.; Liu, Y.; Luo, Q.; Xu, J.; Li, H.; Liu, J. Dynamic protein self-assembly driven by host–guest chemistry and the folding–unfolding feature of a mutually exclusive protein. Chem. Commun. 2017, 53, 10532– 10535. [CrossRef] 147. Hou, C.; Huang, Z.; Fang, Y.; Liu, J. Construction of protein assemblies by host–guest interactions with cucurbiturils. Org. Biomol. Chem. 2017, 15, 4272–4281. [CrossRef] 148. Hou, C.; Zeng, X.; Gao, Y.; Qiao, S.; Zhang, X.; Xu, J.; Liu, J. Cucurbituril As A Versatile Tool to Tune the Functions of Proteins. Isr. J. Chem. 2017, 215163, 1–11. [CrossRef] 149. Dang, D.T.; Van Onzen, A.H.A.M.; Dorland, Y.L.; Brunsveld, L. Reactivation of an Inactivated Caspase-8 Mutant Reveals Differentiated Enzymatic Substrate Processing. ChemBioChem 2020, 19.[CrossRef] 150. Guagnini, F.; Antonik, P.M.; Rennie, M.L.; O’Byrne, P.; Khan, A.R.; Pinalli, R.; Dalcanale, E.; Crowley, P.B. Cucurbit[7]uril- Dimethyllysine Recognition in a Model Protein. Angew. Chem. Int. Ed. 2018, 57, 7126–7130. [CrossRef] 151. Breslow, R.; Yang, Z.; Ching, R.; Trojandt, G.; Odobel, F. Sequence Selective Binding of Peptides by Artificial Receptors in Aqueous Solution. J. Am. Chem. Soc. 1998, 120, 3536–3537. [CrossRef] 152. Fukuhara, G.; Inoue, Y. Peptide chirality sensing by a cyclodextrin-polythiophene conjugate. Chem. A Eur. J. 2012, 18, 11459– 11464. [CrossRef] 153. Weißenstein, A.; Saha-Möller, C.R.; Würthner, F. Optical Sensing of Aromatic Amino Acids and Dipeptides by a Crown-Ether- Functionalized Perylene Bisimide Fluorophore. Chem. A Eur. J. 2018, 24, 8009–8016. [CrossRef][PubMed] 154. Hossain, M.A.; Schneider, H.J. Sequence-selective evaluation of peptide side-chain interaction. New artificial receptors for selective recognition in water. J. Am. Chem. Soc. 1998, 120, 11208–11209. [CrossRef] 155. Liu, Y.; Liao, P.; Cheng, Q.; Hooley, R.J. Protein and small molecule recognition properties of deep cavitands in a supported lipid membrane determined by calcination-enhanced SPR spectroscopy. J. Am. Chem. Soc. 2010, 132, 10383–10390. [CrossRef][PubMed] 156. Ghang, Y.J.; Lloyd, J.J.; Moehlig, M.P.; Arguelles, J.K.; Mettry, M.; Zhang, X.; Julian, R.R.; Cheng, Q.; Hooley, R.J. Labeled protein recognition at a membrane bilayer interface by embedded synthetic receptors. Langmuir 2014, 30, 10161–10166. [CrossRef] 157. Bontempi, N.; Biavardi, E.; Bordiga, D.; Candiani, G.; Alessandri, I.; Bergese, P.; Dalcanale, E. Probing lysine mono-methylation in histone H3 tail peptides with an abiotic receptor coupled to a non-plasmonic resonator. Nanoscale 2017, 9, 8639–8646. [CrossRef] 158. Liu, Y.; Perez, L.; Mettry, M.; Easley, C.J.; Hooley, R.J.; Zhong, W. Self-Aggregating Deep Cavitand Acts as a Fluorescence Displacement Sensor for Lysine Methylation. J. Am. Chem. Soc. 2016, 138, 10746–10749. [CrossRef] 159. Liu, Y.; Perez, L.; Gill, A.D.; Mettry, M.; Li, L.; Wang, Y.; Hooley, R.J.; Zhong, W. Site-Selective Sensing of Histone Methylation Enzyme Activity via an Arrayed Supramolecular Tandem Assay. J. Am. Chem. Soc. 2017, 139, 10964–10967. [CrossRef] 160. Biros, S.M.; Ullrich, E.C.; Hof, F.; Trembleau, L.; Rebek, J. Kinetically Stable Complexes in Water: The Role of Hydration and Hydrophobicity. J. Am. Chem. Soc. 2004, 126, 2870–2876. [CrossRef] 161. Xu, R.; Greiveldinger, G.; Marenus, L.E.; Cooper, A.; Ellman, J.A. Combinatorial library approach for the identification of synthetic receptors targeting vancomycin-resistant bacteria[4]. J. Am. Chem. Soc. 1999, 121, 4898–4899. [CrossRef] 162. Chen, G.; Zheng, S.; Luo, X.; Shen, J.; Zhu, W.; Liu, H.; Gui, C.; Zhang, J.; Zheng, M.; Chum, M.P.; et al. Focused combinatorial library design based on structural diversity, druglikeness and binding affinity score. J. Comb. Chem. 2005, 7, 398–406. [CrossRef] 163. Schmuck, C.; Heil, M. One-armed artificial receptors for the binding of polar tetrapeptides in water: Probing the substrate selectivity of a combinatorial receptor library. Chem. A Eur. J. 2006, 12, 1339–1348. [CrossRef][PubMed] 164. Schmuck, C.; Wich, P. Sequence-dependent stereoselectivity in the binding of tetrapeptides in water by a flexible artificial receptor. Angew. Chem. Int. Ed. 2006, 45, 4277–4281. [CrossRef][PubMed] 165. Whitlock, H.W., Jr.; Chen, C.-W. Molecular Tweezers: A Simple Model of Bifunctional Intercalation. J. Am. Chem. Soc. 1978, 100, 4921–4922. [CrossRef] 166. Leblond, J.; Petitjean, A. Molecular Tweezers: Concepts and Applications. ChemPhysChem 2011, 12, 1043–1051. [Cross- Ref][PubMed] 167. Klärner, F.-G.; Schrader, T. Aromatic Interactions by Molecular Tweezers and Clips in Chemical and Biological Systems. Acc. Chem. Res. 2013, 46, 967–978. [CrossRef][PubMed] 168. Dutt, S.; Wilch, C.; Gersthagen, T.; Talbiersky, P.; Bravo-Rodriguez, K.; Hanni, M.; Sánchez-García, E.; Ochsenfeld, C.; Klärner, F.-G.; Schrader, T. Molecular Tweezers with Varying Anions: A Comparative Study. J. Org. Chem. 2013, 78, 6721–6734. [CrossRef] 169. Lump, E.; Castellano, L.M.; Meier, C.; Seeliger, J.; Erwin, N.; Sperlich, B.; Stürzel, C.M.; Usmani, S.; Hammond, R.M.; Von Einem, J.; et al. A molecular tweezer antagonizes seminal amyloids and HIV infection. Elife 2015, 4, 1–33. [CrossRef] Molecules 2021, 26, 106 41 of 41

170. Röcker, A.E.; Müller, J.A.; Dietzel, E.; Harms, M.; Krüger, F.; Heid, C.; Sowislok, A.; Riber, C.F.; Kupke, A.; Lippold, S.; et al. The molecular tweezer CLR01 inhibits Ebola and Zika virus infection. Antivir. Res. 2018, 152, 26–35. [CrossRef] 171. Weil, T.; Groß, R.; Röcker, A.; Bravo-Rodriguez, K.; Heid, C.; Sowislok, A.; Le, M.-H.; Erwin, N.; Dwivedi, M.; Bart, S.; et al. Supramolecular Mechanism of Viral Envelope Disruption by Molecular Tweezers. J. Am. Chem. Soc. 2020.[CrossRef] 172. Schmuck, C.; Kuchelmeister, H. An efficient synthesis of an orthogonally protected aromatic diamine as scaffold for tweezer receptors withh two different arms. Eur. J. Org. Chem. 2009, 4480–4485. 173. Löwik, D.; Weingarten, D.; Broekema, M.; Brouwer, A.; Still, W.; Liskamp, R. Tweezers with different bite: Increasing the affinity of synthetic receptors by varying the hinge part. Angew. Chem. Int. Ed. 1998, 37, 1846–1850. [CrossRef] 174. Monnee, M.C.F.; Brouwer, A.J.; Liskamp, R.M.J. Synthesis, screening and evaluation of a combined library of tweezer- and tripodal synthetic receptors. QSAR Comb. Sci. 2004, 23, 546–559. [CrossRef] 175. Zarra, S.; Wood, D.M.; Roberts, D.A.; Nitschke, J.R. Molecular containers in complex chemical systems. Chem. Soc. Rev. 2015, 44, 419–432. [CrossRef] 176. Brown, C.J.; Toste, F.D.; Bergman, R.G.; Raymond, K.N. Supramolecular Catalysis in Metal–Ligand Cluster Hosts. Chem. Rev. 2015, 115, 3012–3035. [CrossRef] 177. Chakrabarty, R.; Mukherjee, P.S.; Stang, P.J. Supramolecular Coordination: Self-Assembly of Finite Two- and Three-Dimensional Ensembles. Chem. Rev. 2011, 111, 6810–6918. [CrossRef] 178. Fujita, M.; Tominaga, M.; Hori, A.; Therrien, B. Coordination Assemblies from a Pd(II)-Cornered Square Complex. Acc. Chem. Res. 2005, 38, 369–378. [CrossRef] 179. Tashiro, S.; Tominaga, M.; Kawano, M.; Therrien, B.; Ozeki, T.; Fujita, M. Sequence-Selective Recognition of Peptides within the Single Binding Pocket of a Self-Assembled Coordination Cage. J. Am. Chem. Soc. 2005, 127, 4546–4547. [CrossRef] 180. Mosquera, J.; Szyszko, B.; Ho, S.K.Y.; Nitschke, J.R. Sequence-selective encapsulation and protection of long peptides by a self-assembled Fe II 8 L 6 cubic cage. Nat. Commun. 2017, 8, 6–11. [CrossRef] 181. Reczek, J.J.; Kennedy, A.A.; Halbert, B.T.; Urbach, A.R. Multivalent recognition of peptides by modular self-assembled receptors. J. Am. Chem. Soc. 2009, 131, 2408–2415. [CrossRef]