You are on page 1of 17

Received: 2 March 2021 Revised: 2 April 2021 Accepted: 26 April 2021

DOI: 10.1002/jmr.2901

REVIEW

Applications of isothermal titration calorimetry in pure and


applied research from 2016 to 2020

Robert J. Falconer1 | Boelo Schuur2 | Anthony K. Mittermaier3

1
School of Chemical Engineering & Advanced
Materials, University of Adelaide, Adelaide, Abstract
South Australia, Australia The last 5 years have seen a series of advances in the application of isothermal titra-
2
Faculty of Science and Technology,
tion microcalorimetry (ITC) and interpretation of ITC data. ITC has played an invalu-
University of Twente, Enschede, Netherlands
3
Department of Chemistry, McGill University, able role in understanding multiprotein complex formation including proteolysis-
Montreal, Quebec, Canada targeting chimeras (PROTACS), and mitochondrial autophagy receptor Nix interaction
Correspondence with LC3 and GABARAP. It has also helped elucidate complex allosteric communica-
Robert J. Falconer, School of Chemical tion in protein complexes like trp RNA-binding attenuation protein (TRAP) complex.
Engineering & Advanced Materials, University
of Adelaide, Adelaide, SA 5005, Australia. Advances in kinetics analysis have enabled the calculation of kinetic rate constants
Email: robert.falconer@adelaide.edu.au from pre-existing ITC data sets. Diverse strategies have also been developed to study
enzyme kinetics and enzyme-inhibitor interactions. ITC has also been applied to
study small molecule solvent and solute interactions involved in extraction, separa-
tion, and purification applications including liquid-liquid separation and extractive dis-
tillation. Diverse applications of ITC have been developed from the analysis of
protein instability at different temperatures, determination of enzyme kinetics in sus-
pensions of living cells to the adsorption of uremic toxins from aqueous streams.

KEYWORDS
allostery, distillation, entropy, enzyme kinetics, kinITC, kinITC-ETC, liquid-liquid extraction,
supramolecular

1 | I N T RO DU CT I O N cell biology. From a societal or commercial perspective, it was the use


of ITC in drug discovery and understanding drug-target interactions
The applications for analytical technology are driven by the pressing that has arguably made the greatest contribution.
research questions of the day. The first high sensitivity isothermal Since 1990, there has been a steady rise in publications citing iso-
titration calorimetry (ITC) instruments to be commercially available thermal titration calorimetry until 2018 when the output of publica-
were released in 1990 and the early publications focused on biological tions peaked (Figure 1). Protein chemistry continues to dominate ITC
1,2
applications of this technology. In subsequent years, our biochemi- use in research (Figure 2). In the period 2016 to 2020, 30 years after
cal knowledge underwent massive growth as the human genome was ITCs release, it might be expected that microcalorimetry would be
sequenced and structural genomics determined the 3D structures of stagnating. This is not the case. In protein chemistry, there have been
an enormous library of proteins. ITC played an ever-increasing role in significant advances in data analysis of ITC study of multi-protein
characterizing proteins' interactions with other proteins, other bio- complexes, and the use of ITC to study interaction and reaction kinet-
macromolecules, and with small molecules including drugs. This was ics. In synthetic chemistry, ITC has played an ongoing role in deter-
aided by the publication of a series of papers explaining the method, mining the functionality of supramolecular structures and has future
the mathematics, the interpretation of the data, and methods for scope to determine the mechanisms for binding interactions and the
3-7
overcoming technical limitations of the technology. From a scien- role of solvents in this process. The application of ITC, previously used
tific perspective, it was the use of ITC to study multi-protein com- to study biomacromolecules to studying synthetic macromolecules is
plexes that has made the greatest contribution to our knowledge in relatively straightforward extrapolation. The application of ITC to

J Mol Recognit. 2021;e2901. wileyonlinelibrary.com/journal/jmr © 2021 John Wiley & Sons Ltd. 1 of 17
https://doi.org/10.1002/jmr.2901
2 of 17 FALCONER ET AL.

F I G U R E 1 Articles written with isothermal


titration calorimetry content since 1990 sourced
from the Web of Science, search terms
“isothermal and titration”

study interactions between small molecules is less obvious. It is worth 11. Early ITC and non-ITC references.189-223
remembering that the ITC is simply a titration method that will detect
any interaction where there is a sufficient change in enthalpy (ΔH) for The authors' hope by providing a survey of the published litera-
the instrument to measure. ITC is not limited to studying macromole- ture the readers will appreciate the diverse applications of this useful
cules or even aqueous systems. ITC can study interactions between analytical technique.
small molecules, interactions that play a key part in determining the
effectiveness of processes like liquid-liquid extraction and extractive
distillation. For the cell biologist, interactions between small molecules 2 | P R O T E I N CH E M I S T R Y
are often overlooked and phenomena like molecular crowding and
osmolyte control of osmotic pressure in halophiles are poorly under- 2.1 | Complex formation and allostery
stood. Interestingly, despite 30 years of undertaking ITC research
using the very sensitive microcalorimetry instruments, the physical ITC has played a role in understanding the formation of multi-protein
chemistry of intermolecular interactions still presents some chal- complexes, confirming binding interactions, determining the stoichi-
lenges. The apparent enthalpy/entropy compensation observed for ometry and order of assembly. The field of biomolecular self-assembly
binding interactions between a target and closely related ligands are has benefitted from an orthogonal approach wherein ITC was the gold
still subject to debate.8-11 standard for determining binding, while NMR, SAXS, and X-ray crys-
There were more than 3000 articles reporting the use of ITC tallography were used to generate a detailed understanding of mul-
between January 2016 and December 2020 so the authors have tiprotein complex structure, dynamics, and function. The last 5 years
selected approximately 180 that they feel best to represent the field have yielded a series of articles that are the culmination of many years
and acknowledge many excellent papers were omitted from the selec- of research on multi-protein complex formation. Endosomal transport
tion. These references have been classified into the following broad is reliant on a series of multi-protein complexes and has been the sub-
categories: ject of a series of excellent reviews including caveolae formation,189
retromer and retriever function in endosomal trafficking,190 and
1-11
1. Introduction, retromer-independent endosomal cargo recycling.191
12-19
2. Review and Perspective Articles, Three recent studies nicely illustrate the role ITC can play in teas-
3. Methods Papers,20-29 ing out the details of protein complex formation. Proteolysis-targeting
4. Peptides, Proteins including Enzymes,30-85 chimeras (PROTACS) are involved in selective protein degradation by
5. Lipids, Micelles, and Membranes,86-94 bringing the target protein into proximity with E3 ubiquitin ligases,
6. Nucleic Acids including Aptamers,95-116 which in turn attach a ubiquitin molecule to the target, leading to sub-
7. Supramolecular Structures and Nanoparticles,17,117-157 sequent degradation in proteasomes. ITC demonstrated that the for-
158-167
8. Small Chemicals, Polymers, and Minerals, mation of the ligase-PROTAC-target complex occurs cooperatively.41
26,43,168-174
9. Binding and Reaction Kinetics, The second example is the use of ITC alongside NMR to observe the
10. Process Development,175-188 and role of phosphorylation of the mitochondrial autophagy receptor Nix
FALCONER ET AL. 3 of 17

in promoting the formation of complexes with LC3B. This is an impor- SEDPHAT-based simultaneous analysis was recently applied to
tant step in Nix interaction with LC3/GABARAP, which leads to the better understand T cell activation in the immune response.64 One of
degradation of mitochondria in autophagosomes.70 In a third study, the early steps in transducing the signal from T cell antigen receptors
ITC was used alongside NMR, x-ray crystallography, and molecular is the activation of the enzyme phospholipase-Cγ1, which is recruited
dynamic simulation to study the tetradecameric caseinolytic protease to the plasma membrane in complex with three other proteins. This
(ClpP) complex. The proteasome inhibitor bortezomib mimics a pep- hetero-tetramer is of interest as a drug target for suppressing T cell
tide substrate by binding to the serine in the ClpP active-site inducing activation with potential applications in treating autoimmune diseases
allosteric activation of the enzymatic complex.40 and transplant rejection. Samelson and co-workers combined analyti-
The methodology for performing ITC analysis of multiprotein cal ultracentrifugation data together with three binary ITC titrations
complex formation has also advanced. Protein complexes and their (monomer into monomer), four ternary titrations (monomer into
interactions with ligands are often substantially more complicated dimer), and one quaternary titration (monomer into trimer). They
than the simple 1:1 binding reactions frequently studied by ITC. For found that the last step in the formation of the quaternary complex
example, the energetic dissection of protein complex formation usu- was accompanied by a large favorable binding enthalpy that was
ally requires multiple ITC experiments focusing on different steps in almost completely compensated by a large unfavorable binding
the assembly pathway. Interpretation of these experiments in terms entropy. This is a hallmark of coupled folding and binding in intrinsi-
of a global model of assembly is best achieved by fitting all the cally disordered regions (several of the components have large
datasets simultaneously to a single set of thermodynamic parameters unfolded regions in the monomeric state) and is thought to help pro-
that describe the overall process. There are a number of different tein/protein interactions achieve high specificity without becoming
software packages capable to some extent of globally fitting multiple overly tight, among other advantages.196,197 ITC was uniquely able to
ITC datasets, including the native software provided with MicroCal detect this assembly-driven folding event since it directly gives the
(Malvern Panalytical) instruments, and the AFFINImeter package heats of binding; the phenomenon would be virtually invisible to
(S4SD).26 Notable among these is a suite of open-source software for methods that measure affinity only, because the enthalpic and entro-
simultaneous analyses of ITC and multi-modal datasets developed by pic components cancel out leaving a net free energy change of
Schuck and co-workers at the NIH. In 2016, they published an article almost zero.
in Nature Protocols20 with step-by-step instructions for combined Similarly, there have been recent advances in using ITC to unravel
analysis of ITC data using NITPIC (which performs an analysis of ITC complex allosteric communication networks underlying the binding
peak shapes to improve the accuracy of integrated heats),192 interactions of high-order multimers. Foster et al used ITC to investi-
SEDPHAT (which performs the simultaneous analysis),193,194 and gate interactions of the trp RNA-binding Attenuation Protein (TRAP)
GUSSI (which analyses the uncertainties in the extracted thermody- with L-tryptophan.24 TRAP forms a ring-shaped complex of 11 identi-
namic parameters).195 Together, these articles have been cited more cal subunits, which binds up to 11 molecules of tryptophan and inter-
than 500 times in 2016-2020, according to Clarivate Analytics Web acts with RNA to suppress transcription of the trp operon when
of Science, pointing to considerable uptake by the community tryptophan is in abundance. Previous work had shown that TRAP
(Figure 2). binds tryptophan with moderately positive cooperativity, raising the

F I G U R E 2 Articles written on ITC for the


years 2000, 2005, 2010, 2015 and 2020 sourced
from the Web of Science, search terms
“isothermal and titration” plus the term on the
x-axis
4 of 17 FALCONER ET AL.

question of what kind of binding model is appropriate for this system. site. An apolar part of the drug candidate that localizes to an apolar
Combinatorial calculations show that there are 125 unique patterns portion of the target binding site can be beneficial but contributes to
of bound and unbound subunits in a circular complex with 11 binding TΔS not ΔH. In a paper from 2010, Ladbury et al200 (all eminent ITC
sites.24
According to binding polynomial theory, 197
the equilibrium is scientists) defined the ΔH of binding as “a direct measure of the net
described with as many as 11 sets of change in enthalpy (ΔH) and change in the number and/or strength of the non-covalent bonds on
change in entropy (ΔS) binding parameters. Thus, at first glance, the going from the free to bound state.” The paper then went on to illus-
system could appear recalcitrant to detailed characterization. Never- trate how statins and HIV protease inhibitors had been improved over
theless, Foster and co-workers showed that ITC data collected over a time by shifting from entropy-driven to enthalpy-driven binding. If the
range of temperatures could be well fit with a simple model compris- interpretation of ΔH was simply a measure of the number and/or
ing three distinct types of binding event: binding to a subunit flanked strength of the non-covalent bonds, then modifying the drug candi-
by either two empty subunits, one bound, and one empty subunit, or date to enable additional or stronger non-covalent bonds would result
two bound subunits. Values of ΔH, ΔS, and change in heat capacity in greater ΔG of binding and stronger association. A complicating fac-
(ΔCp) were obtained for each of the three types of reaction. Binding to tor was the observation of entropy-enthalpy compensation (see later
subunits adjacent to one or two bound subunits occurred with about section) where the improvement in ΔH was offset by a corresponding
5-fold higher affinity than when both adjacent subunits were empty. change in TΔS. ITC data are used during the later stages of the drug
Furthermore the magnitude of ΔCp was about 6-fold larger and nega- development pathway where they are combined with X-ray crystal-
tive when at least one of the adjacent subunits was already bound, lography, NMR, and SPR to aid in the choice of lead candidates from
compared to the case when both adjacent subunits were empty, the panel of hits identified during screening, and subsequently during
pointing to the existence of allosterically-modulated conformational the lead optimization process. Decision-making based on ITC data in
changes linked to tryptophan binding that could trigger interactions isolation would be a mistake.
with RNA. One factor that is often overlooked when studying interactions
like drug candidate binding to a target is the role of the solution com-
position. Intriguing drug binding data produced at different tempera-
2.2 | Drug discovery tures and in the presence of different buffers demonstrates that the
ΔG of a binding may be unchanged, while both ΔH and TΔS can be
The last 30 years have seen major changes in the way drugs are devel- affected.16 Entropy-enthalpy compensation was evident in all of these
oped. X-ray crystallography enabled the three-dimensional structures cases. It serves as a useful reminder to ensure solution conditions
of thousands of proteins to be determined with atomic resolution. should be as “natural” as practical to make rational decisions on drug
This in turn enabled drugs to be designed to interact with structurally selection based on ΔH and TΔS values. The challenge of interpreting
well-defined binding sites. Biophysical techniques were applied to the thermodynamic data is worth meeting. Protonation states and
study the resulting target/drug binding. X-ray crystallography is rou- water effects have been overlooked in the past but play a role in bind-
tinely used with crystals of the target/drug complex to derive the ing that cannot be ignored. The beauty of the ITC is that it illustrates
geometry of the binding interaction. Nuclear magnetic resonance the differences between apolar and polar interactions as it can detect
(NMR) is also used to determine the structures of the drug targets the change in water molecules and the transfer of protons, elements
with the candidates, in-situ. ITC gives the association constants and in drug binding that are invisible using X-ray crystallography, NMR,
the thermodynamic parameters for the binding interactions and sur- and SPR.
face plasmon resonance (SPR) yields the association and dissociation
constants for the binding interaction.18 Both ITC and SPR determine
the equilibrium constant of the binding interaction from which the 2.3 | Interaction and reaction kinetics
change in free energy (ΔG) of binding can be calculated [Equation (1)].
ITC provides the observed ΔH and the calculated TΔS of binding ITC is predominantly used to measure the thermodynamics of binding
[Equation (2)]. and assembly processes, however, there is growing interest in using it
to measure the kinetics of reactions as well. These applications are
Δ G ¼ RTlnK a ð1Þ based on the idea that the raw ITC output gives a nearly (but not
exactly) real time readout of the rate that heat is being generated or
ΔG¼ Δ HT ΔS ð2Þ absorbed in the sample cell. The heat flow due to a chemical reaction
is directly proportional to its rate multiplied by the change in enthalpy.
Historically, enthalpy-driven binding was preferred over entropy- Thus, ITC datasets can be readily interpreted in terms of reaction
driven binding and was seen as the key to better drug candidates. The velocities. As most reaction enthalpies are non-zero, ITC represents,
theory was that ΔH is due to hydrogen bonding or similar non- in principle, a nearly universal way to characterize the rates of chemi-
covalent bonding and quite location specific, entropy on the other cal and biological processes. The primary complication in these types
hand was due to water displacement and quite nonspecific.199,200 In of analyses is that there are several factors that contribute to the
principle, this is correct but in practice, it is dependent on the binding shape of the ITC signal in addition to the kinetics of the reactions
FALCONER ET AL. 5 of 17

themselves. These include the rates of heat transfer within the sample riboswitch/thiamine interactions.6,21 Alternatively, the simplified
cell and its contents, the electronic response of the feedback circuitry, kinITC-ETC method analyses only the lengths of time required for ITC
7,169,201
and physical mixing of the injected titrant with the analyte. As peaks to return to baseline following the injections (τend). For systems
a result, a rapid burst of heat in the sample, generated for example by giving useful kinetic data, the values of τend are shorter for the early
a rapid (sub-second) injection of ligand into a concentrated solution of and late injections and longest near the middle of the titration, for the
binding partner, is detected as a peak lasting tens of seconds. The reasons mentioned above.21 The τend vs injection number profile is
instrument response is often represented by the single time constant, governed exclusively by kon (or koff) and KD (along with concentrations,
τITC, which describes the rate of exponential decay of the ITC signal injection volumes, etc.) The value of KD is obtained from the tradi-
following a reference heater pulse or calibration injection.7,202 Note tional thermodynamic analysis. Thus fitting of the τend profile gives
that these methods can give slightly different results, likely because one of the rate constants while the other is calculated from KD=koff/
the heat transfer steps differ in the two cases: heat is generated in the kon. The advantage of this approach is that the complicated effects of
wall of the sample cell during a heater pulse and in the sample solu- mixing and instrument response primarily affect the early portions of
tion itself for the injection.203 While this description holds for longer ITC peaks, while the overall lengths of the peaks are dominated simply
time points, the shape of the ITC response at short times is more com- by the binding kinetics (provided they are sufficiently slow). As a
169
plicated. Regardless of the level of detail of the description used, rough guide, the authors recommend that in order for kinITC-ETC to
the delayed response of the ITC instrument places an upper limit on be successful, the following inequality should hold:
pffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
the rates of reactions that can be characterized by ITC and must be τITC koff kon ½A0 < 5, where [A]0 is the total amount of binding partner,
accounted for quantitatively in any rigorous analyses of ITC that is, [A] + [AB].6 Representative values of τITC vary between about
kinetic data. 3.5 seconds for the Malvern ITC200 to 15 seconds for the Malvern
One particularly interesting aspect of ITC kinetics analysis is that VP-ITC. For a Wiseman c value of 10 ([A]0 = 10koff/kon), this means
a wealth of potentially informative kinetic data has been hiding in that koff should be slower than about 0.5 to 0.1 s1. Importantly,
plain sight. Many ITC datasets for 1:1 binding interactions contain the kinITC-ETC results are generally in good agreement with those of
distinctive feature wherein peaks become broader near the equiva- SPR6,21,29,39,106 and fluorescence spectroscopy,99 providing validation
lence point of the titration (equal mole fractions of binding partners), for the approach. The main disadvantage of this approach is that is
with all other injection parameters held constant. Egawa et al202 and only applies to simple 1:1 (or 1 to multiple identical sites) binding reac-
subsequently Dumas et al6 showed that this peak broadening could tions, as much of the granular detail in the data is deliberately
be quantitatively explained in terms of the association and dissocia- discarded.
tion rate constants, kon and koff. For a simple reaction A(cell) + Recently, Dumas has partnered with the S4SD company to
B(syringe) $ AB, the A component is in excess early in the titration include kinITC-ETC analysis as a standard feature of the
and B is consumed with a pseudo-first order rate constant kon[A]. The AFFINImeter-ITC software package, extending the accessibility of the
concentration of free component A drops throughout the titration as method. The program has been used to study the binding kinetics of
it is converted to AB, leading to successive reductions in the rate of aptamers,99 carbonic anhydrase inhibitors,21,43 HIV reverse transcrip-
binding and broader peaks. Near the end of the titration, the B com- tase inhibitors,21 the nickel binding to the NikR transcription factor,39
ponent is in excess and A is consumed with the pseudo-first order aminopolycarboxylate ion binding,166 and the lectin domain of FimH,
rate constant kon[B]. As free [B] accumulates, the forward rate an E. coli virulence factor.29,49 This last case is notable in that ITC data
increases once again and peaks narrow. Thus, conventional ITC exper- had already been obtained for a panel of potential mannosidic inhibi-
iments can encode kinetic information that is untapped in typical ther- tors and used to elucidate structure-activity relationships based on
modynamics analyses in which only the peak integrals are considered. binding affinity. Ernst et al re-mined the existing data to extract bind-
The researchers provided two alternative approaches to ing kinetic information, discovering that slow on-rates correlated with
extracting kinetic information, which they refer to as kinITC6 and H-bond formation while rapid on-rates correlated with long-range
kinITC-ETC (Equilibration Time Curve).21 In the first, raw ITC data are electrostatic forces and conformational restrictions.29 There is grow-
fitted directly to chemical kinetic equations together with those ing awareness in the drug development field that efficacy is related to
accounting for reagent mixing and the instrument response. This both the strength and the kinetics of drug binding,204 thus structure-
approach has the advantage of being completely general in the type kinetics relationships, such as those uncovered here, are of great
of reactions that can be studied. For example, kinITC was used to interest. The fact that both thermodynamic and kinetic information
study thiamine binding to two different E. coli TPP riboswitches and can be obtained simultaneously in a straightforward manner has impli-
was able to resolve the kinetics of both of the binding step and a sub- cations for the future role of ITC in drug development campaigns.
sequent RNA folding event.6,106 The drawback of this method is that
the mixing and the instrument response must be well accounted for in
order to faithfully reproduce experimental ITC peak shapes, which can 2.4 | Enzyme catalysis of single reactions
present considerable challenges, as mentioned above. Possibly due to
the relative complexity of the analysis, the examples of full kinITC Another area where the use of ITC to measure chemical kinetics has
peak fitting are thus far limited to HIV protease/Nevirapine and TPP grown is in the characterization of enzyme kinetics. According to our
6 of 17 FALCONER ET AL.

search of the literature, there were about 10, 8, 28, and 37 papers after each injection is known from the concentration of substrate in
published on this topic in the 5-year periods beginning in 2001, 2006, the syringe and volumes of all injections. The reaction velocity can be
2011, and 2016. In these applications, the heat measured by the calo- read directly from the vertical position of each step, tracing out a
rimeter is generated by the chemical reaction catalyzed by the enzyme complete ν versus [S] Michaelis-Menten curve, which can be fitted to
rather than by binding and concomitant structural changes, as is the the modified Michaelis-Menten equation. In practice, we find that the
case for the majority of ITC experiments. This has several implications condition of negligible substrate consumption is met when [E]0 ≤
for experimental design. Firstly, the fact that each enzyme typically (104 s)  Km/kcat.
performs multiple catalytic turnovers during the ITC experiment effec- In a single injection ITC kinetic assay, the amount of enzyme is
tively amplifies the heat signal, and the amount of enzyme required is typically chosen to be large enough so that the substrate can be fully
far less (as low as picomolar)168 than that required for binding experi- converted to product on the timescale of minutes or tens of minutes.
ments. In addition to the advantages of requiring less material and dis- The concentration of substrate is chosen so that enzyme is initially
couraging enzyme self-association, this also means that the heats saturated following (each) injection ([S]>>Km).206 Single injection
generated by association and dissociation of substrates, products, and assays can be initiated either by injecting substrate (syringe) into the
inhibitors can be safely ignored, as they are far smaller than the heats enzyme (cell) or by injecting enzyme (syringe) into the substrate (cell).
of catalysis. Secondly, the fact that an enzyme provided with suffi- In either case, the ITC signal exhibits a large deflection immediately
cient substrate can continue to generate heat signals almost indefi- after the injection with large heat flows continuing as long as the
nitely opens the door to a wide variety of experimental designs. All enzyme remains saturated with substrate. The signal gradually returns
ITC enzyme kinetics measurements rely on the simple relationship to the pre-injection baseline as the substrate is consumed. When the
wherein the displacement of the ITC signal from the no catalysis base- substrate is present in the syringe, this procedure may be repeated
line is directly proportional to the velocity of the reaction. Thus, the several times within the same experiment. When enzyme is in the
ITC output represents a (nearly) real-time readout of enzyme activity, syringe, only a single injection can be made, as all the substrate is con-
subject to the caveats of mixing and instrument response times sumed after the first injection. Once again, the velocity of reaction (ν)
described above. Beyond this commonality, a diversity of approaches can be read directly from the displacement of the ITC signal as a func-
is possible. tion of time. The amount of substrate at each time point is known
In many cases, enzyme kinetics are well described by the follow- from partial integration of the ITC peak (the fraction of total substrate
ing modified Michaelis-Menten equation [Equation (3)]205 remaining at time t is equal to the fraction of the total area of the
peak lying to the right of time t). Together the ν and [S] values trace
½E0 kcat ½S out a complete Michaelis-Menten curve. We find that substrate is
ν¼     ð3Þ
K m 1 þ K½Ii þ ½S 1 þ K½I0i consumed sufficiently rapidly for this technique to be applied when
[E] ≥(102 s)Km/kcat. When the concentration of enzyme is too low,
the heat spike persists for such a long time (several hours or more)
where ν is the rate of the reaction (-d[S]/dt), [S], [E]0, and [I] are the that the return to baseline is difficult to distinguish. However, the
concentrations of substrate, enzyme, and inhibitor, respectively. kcat is enzyme must be at a low enough concentration so that the return to
the maximum number of reactions catalyzed by each molecule of baseline takes at least seconds to tens of seconds. More rapid reac-
enzyme per unit time, Km is the concentration of substrate required to tions start to become obscured by the instrument response, as men-
reach half-maximal velocity in the absence of inhibitors, and Ki and K'I tioned above.169 Compared to multiple-injection experiments, the
are dissociation equilibrium constants for inhibitor binding to the single injection method is more rapid and allows kcat and Km to be
enzyme and enzyme-substrate complex. Most enzyme assays aim to extracted from as little as tens of seconds of raw ITC data. As well, a
determine the values of these kinetic parameters. In their seminal great deal more product is generated in a single-injection experiment,
2001 paper, Todd and Gomez describe two general strategies, which which can be advantageous for researchers seeking to characterize
are now generally referred to as “multiple injection” and “single injec- product inhibition but is a disadvantage for those seeking to avoid this
tion” experiments.4 In a multiple injection ITC enzyme kinetic assay, complicating factor.77
the sample cell contains an enzyme solution and the syringe contains The multiple-injection (dilute enzyme) and single-injection (con-
the substrate. The enzyme concentration is chosen to be sufficiently centrated enzyme) approaches provide frameworks for developing
low so that substrate depletion during the experiment is negligible. As new types of ITC kinetic assays. In a recent example, dilute enzyme
a result, the ITC signal is ideally constant (horizontal) between sub- and substrate were both placed in the sample cell with an inhibitor in
strate injections and resembles a series of steps, one per injection. the syringe, leading to a constant reaction velocity and horizontal ITC
The injections are designed such that early steps have [S]<<Km and signal prior to the first injection. Each injection of inhibitor reduced
the final injections have nearly saturated the enzyme with [S]>>Km. the velocity and heat flow, but the change in reaction rate occurred
The displacement of each step relative to the initial baseline is directly over a period of several minutes as the inhibitor gradually bound to
proportional to the reaction velocity, such that each step is smaller the active site of the enzyme. These time traces were fit to extract
than the one preceding it as the enzyme is gradually saturated with the inhibitor association and dissociation rate constants, providing an
substrate. The concentration of substrate present in the sample cell efficient method for rapidly determining inhibitor binding kinetics.168
FALCONER ET AL. 7 of 17

In another example, in a modified single injection experiment, the sub- 2.5 | Enzyme catalysis of multiple reactions
strate was placed together with inhibitor in the syringe and injected
multiple times into concentrated enzyme in the sample cell.170 Each To understand the kinetics of hydrolytic enzymes (glycoside hydro-
injection generated a peak, each of which was fitted to obtain values lase) that digest homopolymers like starch, cellulose, and chitin, you
of kcat and Km. The inhibitor accumulated in the sample cell with each need to consider the sequence of events that occur during cataly-
injection, thereby generating datasets of kcat and Km as functions [I], sis. Non-processive glycoside hydrolases usually follow the
allowing determination of Ki and K'I (modified Michaelis-Menten sequence, (a) attach to the polymeric substrate, (b) hydrolyze the
equation) that is, completely characterizing the strength and mode of bond, (c) separate from the polymer, (d) release the cleavage prod-
inhibition. The method has since been applied to inhibitors of epoxide uct, then reattach to the polymer and start the process again. These
hydrolase, which is of interest as a drug target.30 Notably, the previ- enzymes usually follow classic Michaelis-Menten kinetics, an exam-
ous (non-ITC) assay for this enzyme involved hyphenated liquid chro- ple being the hydrolytic cleavage of maltoheptose by human saliva
matography and mass spectrometry, which involves considerably α-amylase.58 Substrates that are crystalline homopolymers, such as
more time and experimental uncertainty than the ITC-based method. chitin or cellulose are often structurally difficult to attack chemi-
ITC offers many advantages compared to traditional enzyme cally or enzymatically due to the tightly packed polymers locked
assays. Firstly, ITC offers real-time monitoring of enzymatic reactions together by hydrogen bonds. The ability of processive enzymes to
in cases where other types of continuous assays are unavailable, such attach to the polymer prevents them from diffusing away from their
as for epoxide hydrolase, above.30 It is also unique in that traditional target and makes them more efficient than non-processive enzymes
enzyme assays measure concentrations of substrates and products, at digesting these difficult substrates. Attachment of a processive
with rates determined indirectly. ITC directly detects the reaction enzyme to a substrate can be studied using ITC. Serratia marcescens
velocity, enabling new applications such as the inhibitor binding chitinases, ChiA and ChiB binding to (GlcNAc)6 was demonstrated
kinetic experiments described above.168 The ability to employ natural by ITC using catalytically inactivated enzyme.50 Processive
substrates is another large asset for ITC, as traditional assays often enzymes generally don't follow classic Michaelis-Menten kinetics as
involve modified colorigenic or fluorogenic substrate analogs that do the rate of catalysis is partly determined by the level of crystallinity
not necessarily have the same kinetic parameters as the native sub- of the substrate, which can be heterogeneous across different
strate.58 ITC also offers advantages for enzymes where the standard regions of a single sample. Hydrolytic enzymes that break down
assay involves indirect readout with a coupled-enzyme system.207,208 heteropolymers can follow Michaelis-Menten kinetics acting on
This is particularly true when adding co-solutes or inhibitors that homogenous model substrates (eg, trypsin/Nα-Benzoyl-L-arginine
affect enzymatic activity since the secondary enzymes can be affected ethyl ester BAEE). Digestion of heterogeneous substrates is more
as well as the enzyme of interest. In addition, testing spectroscopically complicated. A single injection assay using ITC to study trypsin
active inhibitors or other effector molecules can become a challenge hydrolysis of casein predictably deviates from Michaelis-Menten
when using spectrophotometric assays, that is, with chromogenic or kinetics.67 Small trypsin substrates with a single cleavage site (eg,
fluorogenic probes, or with coupled assays. In contrast, deeply colored BAEE) are the method of choice for studying phenomenon like
inhibitors are fully compatible with ITC inhibition assays.209 Further- molecular crowding as the interpretation of the results is easier
more, ITC's ability to characterize opaque samples further extends the than a complex substrate like casein.67
reach of this technique beyond spectroscopically-accessible systems. Digestion of phytate (myo-inositol hexakisphosphate) by
One particularly interesting example involves using ITC to perform Citrobacter braakii 6-phytase has five discrete cleavage steps and the
enzyme kinetics experiments on suspensions of living cells.59,63,79,85 reaction deviates radically from Michaelis-Menten kinetics.54 The
For instance, Yang et al used ITC to study the metallo-β-lactamase thermogram produced using a single injection of phytate into phytase
NDM-1 in living cultures of E. coli. NDM-1 cleaves carbapenems, pro- had two peaks consistent with two periods of high enzyme activity
viding bacterial resistance to these “last resort” β-lactam antibiotics.85 with a bottleneck at the third cleavage step.
Development of NDM-1 inhibitors has the potential to resensitize
resistant bacteria and offers an avenue for treating these kinds of seri-
ous drug-resistant infections. Single injection experiments with 2.6 | Thermostability of proteins
cefazolin as a test substrate in the syringe and either purified NDM-1
or live E. coli bacteria expressing NDM-1 in the sample cell gave very One of the more innovative uses of ITC published in the last 5 years
similar ITC heat signals. The authors went on to show that ITC could was a paper describing the analysis of the thermostability of lysozyme
diagnose the presence or absence of active NDM-1 in clinical strains using the ITC in isothermal mode without titration.71 Traditional calo-
85
of pathogenic bacteria and used the in vivo ITC experiment to dis- rimetric measurements of thermostability employ differential scanning
cover a new inhibitor scaffold.53 A similar approach was used to iden- calorimetry (DSC) where the instrument measures the heat flow
tify the presence of extended-spectrum β-lactamase activity in a panel required to increase the temperature of a protein sample at a constant
of 69 clinical Enterobacteriaceae samples, yielding results in just an rate. As the temperature rises, the protein unfolds, leading to an
hour, compared to the 32 to 48 hours typically required for traditional increased heat absorption (or heat capacity, Cp) over the transition
growth inhibition assays.59 region. The temperature corresponding to the peak (maximum Cp
8 of 17 FALCONER ET AL.

value) in the unfolding thermogram (Tm) is used as a measure of pro- interactions between low charge density ions and hydrophobic
tein stability. pockets.144 For example, the presence of low charge density ions in a
The ITC research studied the irreversible unfolding and subse- hydrophobic pocket is not driven by electrostatic interactions, since
quent aggregation of hen egg lysozyme in a buffer at pH 9.0, using a the interacting partners are charge neutral, but is due to the energeti-
TA Instruments TAM IV.72 The temperature in the ITC was set at cally favorable displacement of water from this interface. The knowl-
57 C, 58 C, and 59 C, that is, below the Tm of 71 C, and followed the edge gained by studying interactions with non-biological model
heat flow over 6 days. The experiment demonstrated an alternative structures can be transferred to more complex systems like proteins,
method for studying the thermal stability of protein formulations at nucleic acids, and macromolecular complexes.
temperatures that are more representative of physiological or ambient Supramolecular structures that are luminescent or fluorescent
conditions than those used in a DSC. The technique has merit for the with specific recognition capabilities for molecules such as gasses,
development of stable protein formulations and has also been demon- ions, metabolites, drugs, etc. have potential applications as sen-
strated for immunoglobulins36 which are the leading family of thera- sors.136,211 ITC is a useful in these efforts when the analyte is soluble
peutic proteins. and can be employed to characterize closely related molecules and to
ascertain the selectivity molecular recognition.
Drug delivery is an attractive application for supramolecular
3 | S Y N T H E T I C CH E M I S T R Y structures. Many therapeutic agents are relatively hydrophobic and
have low solubility in water or blood. Encasing the therapeutic agent
3.1 | Supramolecular structures in a molecular cage has been used to improve drug solubility in an
aqueous environment.212 For example, a new, highly insoluble
Great advances have been made in the field of supramolecular chem- antidiabetes drug was successfully solubilized by forming inclusion
istry and an incredible diversity of supramolecular structures have complexes with β-cyclodextrin and hydroxypropyl-β-cyclodextrin.146
been synthesized. The molecular cages are an important family within Similar cyclodextrin inclusion strategies have been used with a range
the supramolecular structures. These three-dimensional hollow struc- of drugs, terpenes, and steroids.134 Cyclodextrins have also been pro-
tures include the crown ethers, cryptands, calixarenes, resorcinarenes, posed to deliver drugs for release in the intestine where bile salts help
curcurbiturils, cyclodextrins, pillararenes, cryptophanes, and hemi- displace the drug from the complex, as demonstrated in vitro,137 and
cryptophanes.155 The cavities within these structures provide envi- have also been developed for siRNA delivery.143
ronments with specific dimensions and electric fields that will only Supramolecular structures have been used to create artificial
allow a limited number of molecules to enter. This ability to interact enzymes.148,150 ITC was used to study oxidase-like catalysis using the
with specific atoms or molecules provides a degree of specificity that multiple injection technique developed for studying enzyme kinetics
makes the supramolecular structures potentially useful as sensors, (see above).152 The possibility of designing synthetic enzymes with
drug delivery vehicles, catalytic sites, and as smart interlocking better stability and efficacy than biological enzymes is an obvious goal
materials. with great potential benefits.
A range of analytical techniques are used to study supramolecular Supramolecular polymer networks (SPNs) are polymers cross-
structures including X-ray crystallography to determine the molecular linked by non-covalent interactions.213 These structures differ from
structure, NMR to ascertain the positioning of binding partners within typical covalently linked polymer networks in being capable of revers-
the supramolecular structure, and ITC to study the stoichiometry and ible self-assembly and disassembly, as dictated by physical stress and
thermodynamics of the interaction. ITC is usually confined to deter- environmental changes. This leads to interesting properties such as
mining the association constant, the stoichiometry, and the change in superior toughness, durability, and self-healing. ITC is uniquely suited
enthalpy and entropy for the interaction although in principle it also to measuring the stoichiometry of polymers cross-linked within the
could be used to study the kinetics of interactions and reaction kinet- supramolecular structures.
ics if the molecule has the catalytic capability (as described above).
Early work with ITC tended to focus on macromolecules of bio-
logical origin such as proteins, nucleic acids, lipids, and complexes, 4 | P H Y S I CA L C H E M I S T R Y
which made it difficult to interpret experimental data at the molecular
level. The chemical simplicity of supramolecular structures enabled 4.1 | Enthalpy/entropy compensation
them to be used as research tools to study intermolecular interactions
and study specific interactions. Research using supramolecular struc- There are many examples where chemical changes to either the host
tures provides a unique opportunity to study the energies associated or guest in a binding interaction have resulted in an increase in ΔH
with hydrogen bonding, electrostatic interactions, π π-stacking, cation but was accompanied with a corresponding increase in TΔS that mini-
π and anion π interactions, and water displacement (hydrophobic mized the change in ΔG.9 This enthalpy-entropy compensation is an
210
effect). The cavities formed in supramolecular structures represent interesting phenomenon that has puzzled scientists since it was
excellent model systems for studying phenomena like the exclusion of observed in the 1970s.214 The introduction of ultrasensitive ITCs in
145
water from hydrophobic pockets or constricted spaces or the 1990s made studying entropy-enthalpy compensation easier and
FALCONER ET AL. 9 of 17

raised its prominence within the scientific community.8,9 The consen- based separations are common unit operations applied in many appli-
sus is that the solvent plays an important role in entropy-enthalpy cation areas, including oil refineries, bulk chemical production plants,
10,11
compensation but definitive evidence for this hypothesis has yet the mining industries, and for purification of fine chemicals and phar-
to be found. maceuticals. For any solvent-based separation, the separation perfor-
Schönbeck and Holm published research in 2019, where they mance depends largely on intermolecular interactions between the
used a relatively simple synthetic model system to experimentally solvent and the various solutes in the mixture to separate.
140
study entropy-enthalpy compensation. They used adamantane Historically, information on intermolecular interactions has been
derivatives binding to the cavity in cyclodextrins with hydroxypropyl mostly derived indirectly from biphasic equilibrium measurements and
groups attached to the rim of the cyclodextrin to increase hydropho- assumed interaction mechanisms, sometimes supported by spectro-
bic contacts made during the binding interaction. The interaction was scopic evidence. For example, by measuring compositions in vapor-
enthalpy-driven partly due to the semipolar nature of the cyclodextrin liquid equilibria (VLE) or liquid-liquid equilibria (LLE), information on
and possibly due to the release of water constrained within the tight the activity coefficients of the solutes in the solvent phase can be
cavity. The experiment determined the ΔH, TΔS, ΔG, and ΔCp of the obtained. VLE measurements are labor intensive, and don't provide
different binding interactions between the adamantane derivatives enough information to describe the interactions within the system.
and the modified cyclodextrins. While the experiment did not provide For example, when strong complexes are formed between the solvent
a definitive confirmation that dehydration of apolar moieties is the and solute, additional insights are useful to aid solvent selection. Spec-
cause of observed entropy-enthalpy compensation, it does illustrate a troscopic approaches (such as NMR or FT-IR) have been used to aid
rational approach to this research. When combined with molecular understanding of the interactions within these mixtures but it is here
dynamic simulation this approach could well solve the puzzle of that ITC can play an invaluable role.
entropy-enthalpy compensation.

5.2 | Liquid-liquid extraction and extractive


5 | N E W A R EA S O F I T C R E S E A R C H distillation of organic chemicals

5.1 | ITC analysis of small molecule solvent and ITC is a technique that has been used to study fluid separations, as
solute interactions direct measurement of heat effects arising from titrating a solute into
a solvent provides valuable information on the affinity between the
For historic reasons, there is a tendency to think of ITC as being useful solvent and the solute. Two types of solvents may be considered in
for studying interactions involving macromolecules usually of biologi- fluid separations, single molecule solvents, and composite solvents
cal origin. This is partially due to the release of the first commercially comprised of multiple types of molecules. Typically, single molecule
available microcalorimeters at the time when structural genomics was solvents are physical solvents, and although hydrogen bonding and
emerging as an important area of research. The importance of interac- polarity interactions may occur, strong complexations are hardly
tions between small molecules is obvious to process engineers where found. In composite solvents on the contrary, often an extractant is
these interactions affect processes like distillation and liquid-liquid dissolved in a diluent, and the extractant is added with the purpose to
extraction. form complexes with a solute that is aimed to be extracted or
We have dedicated a section of this review on the research into entrained (the typical term in extractive distillation). In the past few
small molecule interactions. For the cell biologist or protein chemist years, attempts have been made with ITC to characterize both com-
small molecules are important and often overlooked. An obvious posite solvents and single molecule solvents.183,184
example is the action of osmolytes in regulating in vivo osmotic pres- The use of ITC to characterize liquid-liquid separations and link
sure in halophiles, without disrupting cell function. In mammalian cells, experimentally determined interaction energies directly to extraction
small molecules play a role in regulating in vivo ionic strength and vis- performance was pioneered by Cuypers et al in 2008.216 They applied
cosity that helps maintain cell integrity, and they play a role in molecu- ITC to investigate on the thermodynamics of hydrogen bonding com-
lar crowding. Protein chemists are aware that small molecules alter plexations of various phenolics with tributyl phosphate (TBP) and
protein solubility and stability, and have created theories like prefer- trioctyl phosphine oxide (TOPO) extractants. Importantly, they inves-
ential hydration to explain their observations without studying small tigated complexation of TBP and TOPO in neat diluents, but also in
molecule-water interactions to verify these theories' validity. It is water-saturated diluents. The impact of water was obvious, and inter-
interesting that chemical engineers have taken the lead in using ITC to pretation of the role of water in the complexation thermodynamics, as
investigate small molecule interactions where interactions like hydro- well as the impact that this has on the liquid extraction performance
gen bonding, electrostatic interaction or proton exchange can influ- was studied. Especially, considering that the ITC was used to under-
ence the success of distillation and liquid-liquid extraction processes. stand liquid-liquid extraction to remove phenols from aqueous solu-
In chemical engineering, the field of separations is an important tions, it is key to understand the impact of water that is being co-
field, responsible for approximately half of the energy demand of the extracted, even when it is only present in the organic solution in small
entire chemical industry.215 Next to traditional distillations, solvent- concentrations. They found that water interfered in hydrogen bonding
10 of 17 FALCONER ET AL.

between the extractant and the solute, the fitted complexation con- also information on the Gibbs energy was lacking, and as a result, it
stant was significantly lower than in the neat solvent. The constant was not possible to determine a quantitative relation to the VLE with
obtained with ITC corresponded well with the observed complexation ITC results only.
constant based on fitting models to data from liquid-liquid extraction In conclusion, for the standard fluid separation operations in the
experiments. chemical industry, using ITC, possibly in combination with other tech-
In liquid-liquid separations, simple 1:1 interactions often seen in niques such as molecular modeling or spectroscopic techniques, can
ITC biological applications are atypical. ITC data is ideal for determin- yield very valuable insights in intermolecular interactions, whether
ing the stoichiometry of interactions.183 The work of Sprakel and interactions are exothermic or endothermic, and the magnitude of the
Schuur183 investigated the solute concentration range in the molar overall heat effect. Quantification of models describing phase equilib-
range, which is appropriate for bulk chemical separations. This con- ria is not always directly possible, and depends on the presence of the
216
centration range differentiates this work from Cuypers et al and characteristic S-shaped ITC curve. For the specific situation that affin-
ITC experiments undertaken by protein chemists or synthetic chem- ity separations are addressed, with clear attractive interactions involv-
ists. In combination with allowing for complex stoichiometry, this ing the solutes to separate (and typically exothermic heat effects), the
research applied a sequential reaction according to A + B ⇆ AB and typical S-shaped curve allows one to fit one or more complexation
A + AB ⇆ A2B and found that obtaining equilibrium constants for the constants. From these, the Gibbs energies are easily calculated all-
sequential reaction model is possible in the molar concentration owing quantitative comparisons with phase behavior. If the S-curve is
range. not clear enough, quantification of the Gibbs energy is difficult, and
Having established and validated the application of ITC to the thus far, only qualitative insights have been obtained.
183
area of fluid separations, Sprakel and Schuur have used the tech-
nique to investigate on the impact of diluents on the interactions
between extractants and solutes.186 For decades, extraction studies 5.3 | Liquid-liquid extraction for mineral
have appeared claiming both hydrogen bonding complexes and proton processing
transfer from the acid to the base216 and often it was unclear which
of the mechanisms should be applied. Using a combination of ITC and To recover metal ions from leachates, liquid-liquid extraction is com-
molecular modeling,186 for each of the diluents applied it could be monly applied. Mechanistically, liquid-liquid metal extractions differ
very clearly seen when ion exchange did occur and when it did not. from liquid-liquid extraction of most organics, because in contrary to
Active diluents capable of supporting hydrogen bonds such as organics, the physical solubility of metal ions organic solvents is typi-
1-octanol clearly showed 1:1 stoichiometry and proton transfer, cally negligible. Because of the negligible solubility in the organic dilu-
resulting in the larger values of ΔH1,1 observed by ITC, was confirmed ent, doing homogeneous organic phase ITC is thus not an option.
by molecular modeling. Such understanding of extractant – solute Perhaps this limitation to biphasic systems and the corresponding
interactions can also be extended to study the impact of variations in additional degree of complexity is the reason that only a very limited
the extractant molecular structure. By investigating on the impact of number of studies have appeared to date. As early as 1973, Marcus
the molecular structure on the binding of the solute,185 insights were and Kolarik219 published an article on the use of an ITC for characteri-
obtained on the temperature dependency of liquid-liquid extraction, zation of lanthanide extractions, the machine was based on an
which aids process design. Another example where ITC helped under- 800 mL measurement cell. In subsequent years, their equipment
standing phenomena observed in liquid-liquid extraction systems reduced in size so that approximately 100 mL was sufficient.220
180
involved a phosphonium phosphinate ionic liquid. In order to Although this early work showed that determination of thermody-
understand why it was so hard to recover acetic acid from the ionic namic information including the enthalpy of the complexation reaction
1 31
liquid, a multitude of techniques including H-NMR, P-NMR, FT-IR, in the extraction, the Gibbs energy, and corresponding entropy is pos-
and ITC were applied. ITC confirmed earlier suggested multi-acid to sible, most other researchers continued to use other approaches,
phosphinate interactions,218 as well as a clear trend in interaction including several spectroscopic techniques. Around the same time
energies, showing strong binding of the first acid. that Cuypers et al216 reported the coupling of ITC with liquid-liquid
Aiming at understanding also the impact of entrainers (solvents) extraction of phenolics, the topic was picked up again, now with a
in extractive distillation, Sprakel et al184 performed ITC studies on a micro-ITC described in a paper by Zalupski and Nash, who applied a
wide range of entrainers for a variety of binary distillation systems. biphasic ITC analysis.221 One key aspect for carrying out ITC in liquid-
Although good insights were obtained on the energies required to liquid biphasic systems is to pre-saturate the solvent with the appro-
yield interactions that were sufficiently strong to modify the relative priate aqueous solution (eg, aqueous nitrate solution if the metal ion
volatility in the extractive distillation, but sufficiently weak to allow is leached using nitric acid) to avoid additional heat effects interfering
regeneration of the complexes, quantification of complexation con- with the measurement.
stants was mostly not possible, because the typical S-shaped curves in An important mechanism in metal extractions is chelation,
the ITC data were not obtained. Not being able to quantify complexes because to overcome the strong endothermic dehydration of the
is primarily due to the absence of strong complexes when a single metal ions in their aqueous solution,221 a strong interaction in the
molecular solvent is applied. As no complex constants could be fitted, organic phase is necessary. A recent study showed that also in the
FALCONER ET AL. 11 of 17

aqueous phase, chelation of metal ions can take place, and the ther- Studying weaker interactions continue to present a challenge to
modynamics including enthalpy, entropy, and equilibrium constant for some research groups. The authors refer the reader seeking assistance
rare earth ion complexations with linear poly(ethylenimine in this field to Joel Tellinghuisen's 2008 paper on low c binding.223
13
methylenephosophonate) were determined. The work by Archer The take home message from this paper is ITC can be used to
and Schulz13 very nicely displayed that the aqueous phase complexa- understand interactions between a range of molecules from
tions are entropically driven, and depending on the degree of func- nanoparticles down to relatively small solutes and solvents. ITC is not
tionalization with phosphonate groups, a stoichiometry of between only able to detect interactions but also measure their kinetics. ITC's
two and five phosphonate groups per rare earth element ion was capacity to answer research questions is more versatile than most
observed, the maximum value being reported for Eu(III) and the lowest researchers currently utilize. It will be interesting to see if the next
for Dy(III). Although in this work no extraction was studied, it clearly 5 years will see yet more innovative uses for ITC.
demonstrated that ITC can be of great value in characterization of the
nature and strength of complexes formed with rare earth elements DATA AVAILABILITY STAT EMEN T
and other metals, and the work may be of use for extending these Data derived from public domain resources.
findings to aqueous two-phase systems. Among other applications,
aqueous two-phase systems comprising a polymer that binds metals OR CID
may be useful for extraction of rare earth elements. This is certainly of Robert J. Falconer https://orcid.org/0000-0002-9912-9036

interest for the process engineering community related to metals Boelo Schuur https://orcid.org/0000-0001-5169-4311
extraction and recycling, and is likely to become a subject of studies in Anthony K. Mittermaier https://orcid.org/0000-0002-3767-1225
the coming years.
RE FE RE NCE S
(1) Introduction (2) Reviews and Perspective Articles (3)-
5.4 | Adsorption processing for toxin removal Methods Papers (4) Peptides and Proteins including Enzymes (5)-
Lipids, Micelles and Membranes (6) Nucleic Acid including
A totally different application field is adsorbing toxins from aqueous Aptamers (7) Supramolecular Structures and Nanoparticles (8)-
streams, an important field in process engineering. Kato et al177 reported Other Small Chemicals, Polymers and Minerals (9) Binding and
an ITC study on the adsorption of uremic toxins (potassium cresyl sulfate Reaction Kinetics (10) Process Development (11) Early ITC
and potassium indoxyl sulfate) on Zr-based metal organic frameworks and non-ITC References
(Zr-MOFs). It was found by ITC that the complexation of the Zr-MOFs 1. Freire E, Mayorga OL, Straume M. Isothermal titration calorimetry.
Anal. Chem. 1990;62:950A-959A.
with the uremic toxins was enthalpic in nature while a positive entropic
2. Morin PE, Freire E. Direct calorimetric analysis of the enzymatic activ-
term TΔS was observed. The ITC study was supported by density func- ity of yeast cytochrome c oxidase. Biochemistry. 1991;30:8494-8500.
tional theory (DFT) calculations, identifying π-π interactions between the 3. Jelesarov I, Bosshard HR. Isothermal titration calorimetry and differen-
pyridine functionality in the Zr-MOF and the indoxyl groups of the cresyl tial scanning calorimetry as complementary tools to investigate the
energetics of biomolecular recognition. J. Mol. Recognit. 1999;12:
moiety. This study clearly shows how powerful ITC can be also for char-
3-18.
acterization of adsorbent – adsorbate interactions. 4. Todd MJ, Gomez J. Enzyme kinetics determined using calorimetry: a
general assay for enzyme activity? Anal. Biochem. 2001;296:179-187.
5. Olsen SN. Applications of isothermal titration calorimetry to measure
enzyme kinetics and activity in complex solutions. Thermochim. Acta.
6 | C LO S I N G R E M A R K
2006;448:12-18.
6. Burnouf D, Ennifar E, Guedich S, et al. kinITC: a new method for
6.1 | Strengths and weaknesses in published obtaining joint thermodynamic and kinetic data by isothermal titration
research calorimetry. J. Am. Chem. Soc. 2012;134:559-565.
7. Freiburger LA, Auclair K, Mittermaier AK. Elucidating protein binding
mechanisms by variable-c ITC. Chembiochem. 2009;10:2871-2873.
Publication of the thermograms in the manuscript or in the supple-
8. Cooper A, Johnson CM, Lakey JH, Nollmann M. Heat does not come
mentary data has increased over the last 5 years enabling appraisal of in different colours: entropy/enthalpy compensation, free energy win-
the quality of the data and building confidence in the accuracy of the dows, quantum confinement, pressure perturbation calorimetry, solva-
analysis. Quality of much of the published ITC research is very good tion and the multiple causes of heat capacity effects in biomolecular
interactions. Biophys. Chem. 2001;93:215-230.
and the authors have sensibly limited the interpretation of the data.
9. Chodera JD, Mobley DL. Entropy-enthalpy compensation: role and
Probably the most common published error in ITC analysis is failure to ramifications in biomolecular ligand recognition and design. Ann. Rev.
let the signal equilibrate before the next injection. This mistake builds Biophys. 2013;42:121-142.
a systematic error into the calculated thermodynamic constants. The 10. Dragan AI, Read CM, Crane-Robinson C. Enthalpyentropy compen-
sation: the role of solvation. Eur. Biophys. J. 2017;2017(46):301-308.
criticism by Brian Pethica of published ITC research is a cautionary
11. Fox JM, Zhao M, Fink MJ, Kang K, Whitesides GM. The molecular ori-
warning to ITC users.222 It also can serve as guidance to help improve gin of enthalpy/entropy compensation in biomolecular recognition.
the quality of ITC research.15 Annu. Rev. Biophys. 2018;47:223-250.
12 of 17 FALCONER ET AL.

12. Aldeghi M, Heifetz A, Bodkin MJ, Knapp S, Biggin PC. Predictions of 33. Chen C-N, Paul LN, Mermoud JC, Steussy CN, Stauffacher CV. Visu-
ligand selectivity from absolute binding free energy calculations. J. Am. alizing the enzyme mechanism of mevalonate diphosphate decarboxyl-
Chem. Soc. 2017;139:946-957. ase. Nat. Commun. 2020;11:39-69.
13. Archer WR, Schulz MD. Isothermal titration calorimetry: practical 34. Chen P, Guo Z, Chen C, et al. Identification of dual histone
approaches and current applications in soft matter. Soft Matter. 2020; modification-binding protein interaction by combining mass spectrome-
16:8760. try and isothermal titration calorimetric analysis. J. Adv. Res. 2020;22:
14. Claveria-Gimeno R, Vega S, Abian O, Velazquez-Campoy A. A look at 35-42.
ligand binding thermodynamics in drug discovery. Expert Opin. Drug 35. Clairfeuille T, Mas C, Chan ASM, et al. A molecular code for
Discov. 2017;12:363-377. endosomal recycling of phosphorylated cargos by the SNX27-retromer
15. Falconer RJ. Applications of isothermal titration calorimetry – the complex. Nat. Struct. Mol. Biol. 2016;23:921-932.
research and technical developments from 2011 to 2015. J. Mol. 36. Clarkson BR, Chaudri R, Schoen A, Cooper JW, Kuelo L, Freire E.
Recognit. 2016;29:504-515. Long term stability of a HIV-1 neutralizing monoclonal antibody using
16. Klebe G. Broad-scale analysis of thermodynamic signatures in medicinal isothermal calorimetry. Anal. Biochem. 2018;554:61-69.
chemistry: are enthalpy-favored binders the better development option? 37. Darby SJ, Platts L, Daniel MS, Cowieson AJ, Falconer RJ. An isother-
Drug Discovery Today. 2019;24:943-948. mal titration calorimetry study of phytate binding to lysozyme.
17. Prozeller D, Morsbach S, Landfester K. Isothermal titration calorime- J. Therm. Anal. Calorim. 2017;127:1201-1208.
try as a complementary method for investigating nanoparticle-protein 38. Esposito D, Gunster RA, Martino L, et al. Structural basis for the
interactions. Nanoscale. 2019;11:19265-19273. glycosyltransferase activity of the Salmonella effector SseK3. J. Biol.
18. Renaud J-P, Chung C-W, Danielson UH, et al. Biophysics in drug dis- Chem. 2018;293:5064-5078.
covery: impact, challenges and opportunities. Nat. Rev. Drug Discov. 39. Fabini E, Zambelli B, Mazzei L, Ciurli S, Bertucci C. Surface plasmon
2016;15:679-698. resonance and isothermal titration calorimetry to monitor the Ni(II)-
19. Sandner A, Huefner-Wulsdorf T, Heine A, Steinmetzer T, Klebe G. dependent binding of Helicobacter pylori NikR to DNA. Anal. Bioanal.
Strategies for late-stage optimization: profiling thermodynamics by pre- Chem. 2016;408:7971-7980.
organization and salt bridge shielding. J. Med. Chem. 2019;62:9753- 40. Felix J, Weinhaupl K, Chipot C, et al. Mechanism of the allosteric acti-
9771. vation of the ClpP protease machinery by substrates and active-site
20. Brautigam CA, Zhao H, Vargas C, Keller S, Schuck P. Integration and inhibitors. Sci. Adv. 2019;5:eaaw3818.
global analysis of isothermal titration calorimetry data for studying 41. Gadd MS, Testa A, Lucas X, et al. Structural basis of PROTAC coopera-
macromolecular interactions. Nat. Protoc. 2016;11:882-894. tive recognition for selective protein degradation. Nat. Chem. 2017;13:
21. Dumas P, Ennifar E, Da Veiga C, et al. Extending ITC to kinetics with 514-521.
kinITC. Methods Enzymol. 2016;567:157-180. 42. Giri M, Maulik A, Singh M. Signatures of specific DNA binding by the
22. Ge XJ, Li DX, Chen L, Liu RX, Ge GL. A practical method to evaluate AT-rich interaction domain of BAF250a. Biochemistry. 2020;29:
major statistical errors in isothermal titration calorimetry. Thermochim 100-113.
Acta. 2020;689:178626. 43. Glöckner S, Ngo K, Sager CP, Hüfner-Wulsdorf T, Heine A, Klebe G.
23. Hevener KE, Pesavento R, Ren JH, Lee H, Ratia K, Johnson ME. Hit- Conformational changes in alkyl chains determine the thermodynamic
to-lead: hit validation and assessment. Methods Enzymol. 2018;610: and kinetic binding profiles of carbonic anhydrase inhibitors. ACS
265-309. Chem. Biol. 2020;15:675-685.
24. Ihms EC, Kleckner IR, Gollnick P, Foster MP. Mechanistic models fit 44. Hansen LD, Transtrum MK, Quinn C, Demarse N. Enzyme-catalyzed
to variable temperature calorimetric data provide insights into coopera- and binding reaction kinetics determined by titration calorimetry. Bio-
tivity. Biophys. J. 2017;112:1328-1338. chim. Biophys. Acta. 2016;1860:957-966.
25. Paketuryte V, Linkuviene V, Krainer G, Chen W-Y, Matulis D. 45. Hayama R, Sparks S, Hecht LM, et al. Thermodynamic characteriza-
Repeatability, precision, and accuracy of the enthalpies and Gibbs ener- tion of the multivalent interactions underlying rapid and selective trans-
gies of a protein-ligand binding reaction measured by isothermal titra- location through the nuclear pore complex. J. Biol. Chem. 2018;293:
tion calorimetry. Eur. Biophys. J. 2019;48:139-152. 4555-4563.
26. Piñeiro A, Mun oz E, Sabin J, et al. AFFINImeter: a software to analyze 46. Hijazi M, Krumm C, Cinar S, et al. Entropically driven polymeric
molecular recognition processes from experimental data. Anal. Bio- enzyme inhibitors by end-group directed conjugation. Chem.-Eur. J.
chem. 2019;577:117-134. 2018;24:4523-4527.
27. Scheuermann TH, Padrick SB, Gardner KH, Brautigam CA. On the 47. Hines RM, Maric HM, Hines DJ, et al. Developmental seizures and
acquisition and analysis of microscale thermophoresis data. Anal. Bio- mortality result from reducing GABAA receptor α2-subunit interaction
chem. 2016;496:79-93. with collybistin. Nat. Commun. 2018;9:3130.
28. Tellinghuisen J. Analysis of multitemperature isothermal titration calo- 48. Hirao K, Andrews S, Kuroki K, et al. Structure of HIV-2 Nef reveals
rimetry data at very low c: global beats van't Hoff. Anal. Biochem. features distinct from HIV-1 involved in immune regulation. iScience.
2016;513:43-46. 2020;23:100758.
29. Zihlmann P, Silbermann M, Sharpe T, et al. KinITC-one method sup- 49. Igde S, Röblitz S, Müller A, et al. Linear precision glycomacromolecules
ports both thermodynamic and kinetic SARs as exemplified on FimH with varying interligand spacing and linker functionalities binding to
antagonists. Chemistry. 2018;24:13049-13057. Concanavalin A and the bacterial lectin FimH. Macromol. Biosci.
30. Abis G, Pacheco-Gomez R, Bui TTT, Conte MR. Isothermal titration 2017;17:1700198.
calorimetry enables rapid characterisation of enzyme kinetics and inhi- 50. Jana S, Hamre AG, Wildberger P, et al. Aromatic-mediated carbohy-
bition for the human soluble epoxide hydrolase. Anal. Chem. 2019;91: drate recognition in processive Serratia marcescens chitinases. J. Phys.
14865-14872. Chem. B. 2016;120:1236-1249.
31. Bonin JP, Sapienza PJ, Wilkerson E, et al. Positive cooperativity in 51. Jankovic B, Gulzar A, Zanobini C, et al. Photocontrolling protein-
substrate binding by human thymidylate synthase. Biophys. J. 2019; peptide interactions: from minimal perturbation to complete unbinding.
117:1074-1084. J. Am. Chem. Soc. 2019;141:10702-10710.
32. Chandra M, Chin YK-Y, Mas C, et al. Classification of the human phox 52. Kaira GS, Kapoor M. How substrate subsites in GH26 endo-mannanase
homology (PX) domains based on their phosphoinositide binding speci- contribute towards mannan binding. Biochem. Biophys. Res. Commun.
ficities. Nat. Commun. 2019;10:1528. 2019;510:358-363.
FALCONER ET AL. 13 of 17

53. Kang PW, Su JP, Sun LY, Gao H, Yang KW. 3-Bromopyruvate as a 72. Schön A, Clarkson BR, Jaime M, Freire E. Temperature stability of pro-
potent covalently reversible inhibitor of New Delhi metallo- teins: analysis of irreversible denaturation using isothermal calorimetry.
β-lactamase-1 (NDM-1). Eur. J. Pharm. Sci. 2020;142. 105161. Proteins. 2017;85:2009-2016.
https://doi.org/10.1016/j.ejps.2019.105161. 73. Soini L, Leysen S, Davis J, Westwood M, Ottmann C. The
54. Kempapidis T, Bradshaw NJ, Hodges HE, Cowieson AJ, 14-3-3/SLP76 protein-protein interaction in T-cell receptor signalling: a
Cameron DD, Falconer RJ. Phytase catalysis of dephosphorylation structural and biophysical characterization. FEBS Lett. 2020;595.
studied using isothermal titration calorimetry and electrospray ioniza- https://doi.org/10.1002/1873-3468.13993.
tion time-of-flight mass spectroscopy. Anal. Biochem. 2020;606: 74. Takahashi T, Nagatoishi S, Kuroda D, Tsumoto K. Thermodynamic
113859. and computational analyses reveal the functional roles of the galloyl
55. Kilmister RL, Faulkner P, Downey MO, Darby SJ, Falconer RJ. The group of tea catechins in molecular recognition. PLoS ONE. 2018;13:
complexity of condensed tannin binding to bovine serum albumin - An e0204856.
isothermal titration calorimetry study. Food Chem. 2016;190: 75. Tiwari N, Srivastava A, Kundu B, Munde M. Biophysical insight into
173-178. the heparin-peptide interaction and its modulation by a small molecule.
56. Krimmer SG, Cramer J, Betz M, et al. Rational design of thermody- J. Mol. Recognit. 2018;31:e2674.
namic and kinetic binding profiles by optimizing surface water networks 76. Verteramo ML, Stenström O, Ignjatovic MM, et al. Interplay between
coating protein-bound ligands. J. Med. Chem. 2016;59:10530-10548. conformational entropy and solvation entropy in protein-ligand binding.
57. Krimmer SG, Cramer J, Schiebel J, Heine A, Klebe G. How nothing J. Am. Chem. Soc. 2019;141:2012-2026.
boosts affinity: hydrophobic ligand binding to the virtually vacated S1' 77. Wang W-J, Wang Q, Zhang Y, et al. Characterization of β-lactamase
pocket of thermolysin. J. Am. Chem. Soc. 2017;139:10419-10431. activity using isothermal titration calorimetry. Biochim Biophys Acta
58. Lehoczki G, Szabo  K, Tak acs I, Kandra L, Gyémant G. Simple ITC Gen Subj. 2017;1861:2031-2038.
method for activity and inhibition studies on human salivary α-amylase. 78. Wang Y, Guan J, Di Trani JM, Auclair K, Mittermaier AK. Inhibition
J. Enzyme Inhib. Med. Chem. 2016;31:1648-1653. and activation pf kinase by reaction products: a reporter-free assay.
59. Lei JE, Wang Q, Lin Y, et al. Rapid detection of extended-spectrum Anal. Chem. 2019;91:11803-11811.
β-lactamases producers in Enterobacteriaceae using a calorimetry 79. Wang Y, Wang G, Moitessier N, Mittermaier AK. Enzyme kinetics by
approach. J. Appl. Microbiol. 2020;130. https://doi.org/10.1111/ isothermal titration calorimetry: allostery, inhibition, and dynamics.
jam.14841. Front Mol. Biosci. 2020;7:583826.
60. Li L-L, Yuan H, Liao F, et al. Rational design of artificial dye- 80. Wienen-Schmidt B, Jonker HRA, Wulsdorf T, et al. Paradoxically,
decolorizing peroxidases using myoglobin by engineering Tyr/Trp in the most flexible ligand binds most entropy-favored: intriguing impact of
heme center. Dalton Trans. 2017;46:11230-11238. ligand flexibility and solvation on drug-kinase binding. J. Med. Chem.
61. Loerch S, Leach JR, Horner SW, et al. The pre-mRNA splicing and 2018;61:5922-5933.
transcription factor Tat-SF1 is a functional partner of the spliceosome 81. Wilson LA, Garcia D, Pedroso MM, Schulz BL, Guddat LW,
SF3b1 subunit via a U2AF homology motif interface. J. Biol. Chem. Schenk G. Adaptation of a continuous, calorimetric kinetic assay to
2019;294:2892-2902. study the agmatinase-catalyzed hydrolytic reaction. Anal. Biochem.
62. Luo Q, Chen D, Boom RM, Janssen AEM. Revisiting the enzymatic 2020;595:113618.
kinetics of pepsin using isothermal titration calorimetry. Food Chem. 82. Yachnin BJ, Lau PCK, Berghuis AM. The role of conformational flexibil-
2018;268:94-100. ity in Baeyer-Villiger monooxygenase catalysis and structure. Biochim.
63. Lv M, Zhang Y-J, Zhou F, et al. Real-time monitoring of D-Ala-D-Ala Biophys. Acta. 2016;1864:1641-1648.
dipeptidase activity of VanX in living bacteria by isothermal titration 83. Yuan Z, Praxenthaler H, Tabaja N, et al. Structure and function of the
calorimetry. Anal. Biochem. 2019;578:29-35. Su(H)-hairless repressor complex, the major antagonist of notch signal-
64. Manna A, Zjao HY, Wada J, et al. Cooperative assembly of a four- ing in Drosophila melanogaster. PLoS Biol. 2016;14:e1002509.
molecule signaling complex formed upon T cell antigen receptor activa- 84. Zhang X, Ding X, Zhu Y, et al. Structural and functional characteriza-
tion. Proc. Natl. Acad. Sci. USA. 2018;115:E11914-E11923. tion of HIV-1 cell fusion inhibitor T20. AIDS. 2019;33:1-11.
65. Marquette A, Bechinger B. Biophysical investigations elucidating the 85. Zhang Y-J, Wang W-M, Oelschlaeger P, et al. Real-time monitoring of
mechanisms of Action of antimicrobial peptides and their synergism. NDM-1 activity in live bacterial cells by isothermal titration calorimetry:
Biomolecules. 2018;8:18. a new approach to measure inhibition of antibiotic-resistant bacteria.
66. Matsarskaia O, Braun MK, Roosen-Runge F, et al. Cation-induced ACS Infect. Dis. 2018;4:1671-1678.
hydration effects cause lower critical solution temperature behavior in 86. Bai L, Xiang W, Huan S, Rojas OJ. Formulation and stabilization of
protein solutions. J. Phys. Chem. B. 2016;120:7731-7736. concentrated edible oil-in-water emulsions based on electrostatic com-
67. Maximova K, Wojtczak J, Trylska J. Enzyme kinetics in crowded solu- plexes of a food-grade cationic surfactant (ethyl lauroyl arginate) and
tions from isothermal titration calorimetry. Anal. Biochem. 2019;567: cellulose nanocrystals. Biomacromolecules. 2018;19:1674-1685.
96-105. 87. Cobanov I, Sarac B, Medos Z, et al. Effect of cationic structure of sur-
68. Porter NJ, Wagner FF, Christianson DW. Entropy as a driver of selec- face active ionic liquids on their micellization: a thermodynamic study.
tivity for inhibitor binding to histone deacetylase 6. Biochemistry. J. Mol. Liq. 2018;271:437-442.
2018;57:3916-3924. 88. Danino D, Abezgauz L, Portnaya I, Dan N. From discs to ribbons net-
69. Profeta GS, dos Reis CV, Santiago AS, et al. Binding and structural works: the second critical micelle concentration in nonionic sterol solu-
analyses of potent inhibitors of the human Ca2+/calmodulin dependent tions. J. Phys. Chem. Lett. 2016;17:1434-1439.
protein kinase kinase 2 (CAMKK2) identified from a collection of 89. Fernandez-Alvarez R, Dordovic V, Uchman M, Matejicek P. Amphi-
commercially-available kinase inhibitors. Sci. Rep. 2019;9:16452. philes without head-and-tail design: nanostructures based on the self-
70. Rogov VV, Suzuki H, Marinkovic M, et al. Phosphorylation of the assembly of anionic boron cluster compounds. Langmuir. 2018;34:
mitochondrial autophagy receptor Nix enhances its interaction with 3541-3554.
LC3 proteins. Sci. Rep. 2017;7:1131. 90. Henderson JM, Iyengar NS, Lam KLH, et al. Beyond electrostatics:
71. Sager CP, Fiege B, Zihlmann P, et al. The price of flexibility - A case antimicrobial peptide selectivity and the influence of cholesterol-
study on septanoses as pyranose mimetics. Chem. Sci. 2018;9: mediated fluidity and lipid chain length on protegrin-1 activity. Biochim
646-654. Biophys Acta Biomembr. 2019;1861:182977.
14 of 17 FALCONER ET AL.

91. Loh W, Brinatti C, Tam KC. Use of isothermal titration calorimetry to 110. Neves MAD, Slavkovic S, Churcher Z, Johnson PE. Salt-mediated
study surfactant aggregation in colloidal systems. Biochim Biophys two-site ligand binding by the cocaine-binding aptamer. Nucleic Acid
Acta. 2016;1860:999-1016. Res. 2017;45:1041-1048.
92. Medos Z, Plechkova NV, Friesen S, Buchner R, Bester-Rogac M. 111. Neves MAD, Slavkovic S, Reinstein O, Shoara AA, Johnson PE. A
Insight into the hydration of cationic surfactants: a thermodynamic and proof of concept application of aptachain: ligand-induced self-assembly
dielectric study of functionalized quaternary ammonium chlorides. of a DNA aptamer. RSC Adv. 2019;9:1690-1695.
Langmuir. 2019;35:3759-3772. 112. Oguro A, Yanagida A, Fujieda Y, et al. Two stems with different char-
93. Singh G, Singh G, Kang TS. Micellization behavior of surface active acteristics and an internal loop in an RNA aptamer contribute to
ionic liquids having aromatic counterions in aqueous media. J. Phys. spermine-binding. J. Biochem. 2017;161:197-206.
Chem. B. 2016;120:1092-1105. 113. Schnarr L, Jana J, Preckwinkel P, Weisz K. Impact of a snap-back loop
94. Tso S-C, Mahler F, Hoering J, Keller S, Brautigam CA. Fast and robust on stability and ligand binding to a parallel G-quadruplex. J. Phys.
quantification of detergent micellization thermodynamics from isother- Chem. B. 2020;124:2778-2787.
mal titration calorimetry. Anal. Chem. 2020;92:1154-1161. 114. Shoara AA, Reinstein O, Borhani OA, et al. Development of a
95. Abraham KM, Roueinfar M, Ponce AT, Lussier ME, Benson DB, thermal-stable structure-switching cocaine-binding aptamer. Bio-
Hong KL. In vitro selection and characterization of a single-stranded chimie. 2018;145:137-144.
DNA aptamer against the herbicide atrazine. ACS Omega. 2018;3: 115. Zhang Z, Oni O, Liu J. New insights into a classic aptamer: binding
13576-13583. sites, cooperativity and more sensitive adenosine detection. Nucleic
96. Amano R, Takada K, Tanaka Y, et al. Kinetic and thermodynamic ana- Acid Res. 2017;45:7593-7601.
lyses of interaction between a high affinity RNA aptamer and its target 116. Zong C, Liu J. The arsenic-binding aptamer cannot bind arsenic: critical
protein. Biochemistry. 2016;55:6221-6229. evaluation of aptamer selection and binding. Anal. Chem. 2019;91:
97. Bayrac C, Oktem HA. Evaluation of Staphylococcus aureus DNA 10887-10893.
aptamer by enzyme-linked aptamer assay and isothermal titration calo- 117. Andersen NN, Eriksen K, Lisbjerg M, et al. Entropy/enthalpy compen-
rimetry. J. Mol. Recognit. 2017;30:e2583. sation in anion binding: biotin[6]uril and biotin-L-sulfoxide[6]uril reveal
98. Bell DR, Weber JK, Yin W, Huynh T, Duan W, Zhou R. In silico design strong solvent dependency. J. Org. Chem. 2019;84:2577-2584.
and validation of high-affinity RNA aptamers targeting epithelial cellular 118. Bai D, Gao Z, Tao Z, et al. A study of the interaction between inverted
adhesion molecule dimers. Proc. Natl. Acad. Sci. USA. 2020;117: cucurbit[6]uril and symmetric viologens. New J. Chem. 2018;42:
8486-8493. 11085-11092.
99. Challier L, Miranda-Castro R, Barbe B, et al. Multianalytical study of 119. Bartus E, Olajos G, Schuster I, et al. Structural optimization of
the binding between a small chiral molecule and a DNA aptamer: evi- foldamer-dendrimer conjugates as multivalent agents against the toxic
dence for asymmetric steric effect upon 3'-versus 5'-end sequence mod- effects of amyloid beta oligomers. Molecules. 2018;23:2523.
ification. Anal. Chem. 2016;88:11963-11971. 120. Borissov A, Marques I, Lim JYC, Felix V, Smith MD, Beer PD. Anion
100. DuPont JI, Henderson KL, Metz A, Le VH, Emerson JP, Lewis EA. recognition in water by charge-neutral halogen and chalcogen bonding
Calorimetric and spectroscopic investigations of the binding of meta- foldamer receptors. J. Am. Chem. Soc. 2019;141:4119-4129.
llated porphyrins to G-quadruplex DNA. Biochim. Biophys. Acta. 121. Chen D, Yang M, Zheng N, et al. A novel aptasensor for electrochemical
2016;1860:902-909. detection of ractopamine, clenbuterol, salbutamol, phenylethanolamine
101. Dutta D, Wedekind JE. Nucleobase mutants of a bacterial preQ1-II and procaterol. Biosens. Bioelectron. 2016;80:525-531.
riboswitch that uncouple metabolite sensing from gene regulation. 122. Das NK, Chakraborty S, Mukherjee M, Mukherjee S. Enhanced lumi-
J. Biol. Chem. 2020;295:2555-2567. nescent properties of photo-stable copper nanoclusters through forma-
102. Fong L-K, Wang Z, Schatz GC, Luijten E, Mirkin CA. The role of struc- tion of “protein-corona”-like assemblies. Chem. Phys. Chem. 2018;19:
tural enthalpy in spherical nucleic acid hybridization. J. Am. Chem. Soc. 2218-2223.
2018;140:6226-6230. 123. Dey P, Rajdev P, Pramanik P, Ghosh S. Specific supramolecular inter-
103. Garci A, Castor KJ, Fakhoury J, et al. Efficient and rapid mechano- action regulated entropically favorable assembly of amphiphilic macro-
chemical assembly of platinum(II) squares for guanine quadruplex molecules. Macromolecules. 2018;51:5182-5190.
targeting. J. Am. Chem. Soc. 2017;139:16913-16922. 124. Dreger A, Engelage E, Mallick B, Beer PD, Huber SM. The role of
104. Ghosh A, Ekka MK, Tawani A, Kumar A, Chakraborty D, Maiti S. Res- charge in 1,2,3-triazol(ium)-based halogen bonding activators. Chem.
toration of miRNA-149 expression by TmPyP4 induced unfolding of Commun. 2018;54:4013-4016.
quadruplex within its precursor. Biochemistry. 2019;58:514-525. 125. Eggers DK, Fu S, Ngo DV, Vuong EH, Brotin T. Thermodynamic con-
105. Grau FC, Jaeger J, Groher F, Suess B, Muller YA. The complex formed tributions of water in crytophane host-guest binding reaction. J. Phys.
between a synthetic RNA aptamer and the transcription repressor TetR Chem. B. 2020;124:6585-6591.
is a structural and functional twin of the operator DNA-TetR regulator 126. Feng H, Kan J, Redshaw C, Bian B, Tao Z, Xiao X. Supramolecular
complex. Nucleic Acids Res. 2020;48:3366-3378. drug inclusion complex constructed from cucurbit[7]uril and the hepati-
106. Guedich S, Puffer-Enders B, Baltzinger M, et al. Quantitative and pre- tis B drug Adefovir. Supramol. Chem. 2019;31:260-267.
dictive model of kinetic regulation by E. coli TPP riboswitches. RNA 127. Gao Z-Z, Kan J-L, Chen L-X, et al. Binding and selectivity of essential
Biol. 2016;13:373-390. amino acid guests to the inverted cucurbit[7]uril host. ACS Omega.
107. Gulbakan B, Barylyuk K, Schneider P, Pillong M, Schneider G, 2017;2:5633-5640.
Zenobi R. Native electrospray ionization mass spectrometry reveals 128. George KL, Horne WS. Heterogeneous-backbone foldamer mimics of
multiple facets of aptamer-ligand interactions: from mechanism to bind- zinc finger tertiary structure. J. Am. Chem. Soc. 2017;139:7931-
ing constants. J. Am. Chem. Soc. 2018;140:7486-7497. 7938.
108. Liu K, Xu C, Lei M, et al. Structural basis for the ability of MBD 129. Hirani Z, Taylor HF, Babcock EF, et al. Molecular recognition of
domains to bind methyl-CG and TG sites in DNA. J. Biol. Chem. 2018; methionine-terminated peptides by cucurbit[8]uril. J. Am. Chem. Soc.
293:7344-7354. 2018;140:12263-12269.
109. Mondal S, Jana J, Sengupta P, Jana S, Chatterjee S. Myricetin arrests 130. Huang R, Lau BLT. Biomolecule–nanoparticle interactions: elucidation
human telomeric G-quadruplex structure: a new mechanistic approach of the thermodynamics by isothermal titration calorimetry. Biochim.
as an anticancer agent. Mol. BioSyst. 2016;12:2506-2518. Biophys. Acta. 2016;1860:945-956.
FALCONER ET AL. 15 of 17

131. Huang Q, Li S, Ding Y-F, Yin H, Wang L-H, Wang R. Macrocycle- 152. Xin Q, Jia X, Nawaz A, Xie W, Li L, Gong JR. Mimicking peroxidase
wrapped polyethylenimine for gene delivery with reduced cytotoxicity. active site microenvironment by functionalized graphene quantum dots.
Biomater. Sci. 2018;6:1031-1039. Nano Res. 2020;13:1427-1433.
132. Kaabel S, Adamson J, Topic F, et al. Chiral hemicucurbit[8]uril as an 153. Yao QF, Lu BZ, Ji CD, Yin MZ. Supramolecular host-guest systems as
anion receptor: selectivity to size, shape and charge distribution. Chem. ratiometric Fe3+ ion sensor based water-soluble pillar[5]arene. ACS
Sci. 2017;8:2184-2190. Appl. Mater. Interfaces. 2017;9:36320-36326.
133. Kohler J, Schonbeck C, Westh P, Holm R. Hydration differences 154. Yin M-M, Chen W-Q, Lu Y-Q, Han J-Y, Liu Y, Jiang F-L. A model
explain the large variations in the complexation thermodynamics of beyond protein corona: thermodynamics and binding stoichiometries of
modified gamma-cyclodextrins with bile salts. J. Phys. Chem. B. 2016; the interactions between ultrasmall gold nanoclusters and proteins.
120:396-405. Nanoscale. 2020;12:4573-4585.
134. Lazar AI, Biedermann F, Mustafina KR, Assaf KI, Hennig A, 155. Zhang D, Martinez A, Dutasta J-P. Emergence of hemicryptophases:
Nau WM. Nanomolar binding of steroids to cucurbit[n]urils: selectivity from synthesis to applications for recognition, molecular machines, and
and applications. J. Am. Chem. Soc. 2016;138:13022-13029. supramolecular catalysis. Chem. Rev. 2017;117:4900-4942.
135. Liang H, Lin F, Zhang Z, et al. Multicopper laccase mimicking 156. Zhang Z, Li Y, Zhang X, Liu J. Molecularly imprinted nanozymes with
nanozymes with nucleotides as ligands. ACS Appl. Mater. Interfaces. faster catalytic activity and better specificity. Nanoscale. 2019;11:
2017;9:1352-1360. 4854-4863.
136. Molina P, Zapata F, Caballero A. Anion recognition strategies based on 157. Zhang Z, Zhang X, Liu B, Liu J. Molecular imprinting on inorganic
combined noncovalent interactions. Chem. Rev. 2017;117:9907- nanozymes for hundred-fold enzyme specificity. J. Am. Chem. Soc.
9972. 2017;139:5412-5419.
137. Olsen NE, Westh P, Holm R. Displacement of drugs from cyclodextrin 158. Chen H, Koopal LK, Xiong J, Avena M, Tan W. Mechanisms of soil
complexes by bile salts: a suggestion of an intestinal drug-solubilizing humic acid adsorption onto montmorillonite and kaolinite. J. Colloid
capacity from an in vitro model. J. Pharm. Sci. 2016;105:2640-2647. Interface Sci. 2017;504:457-467.
138. Paul BK, Ghosh N, Mukherjee S. Interaction of bile salts with beta- 159. Enke M, Jehle F, Bode S, et al. Histidine-zinc interactions investigated
cyclodextrins reveals nonclassical hydrophobic effect and enthalpy- by isothermal titration calorimetry (ITC) and their application in self-
entropy compensation. J. Phys. Chem. B. 2016;120:3963-3968. healing polymers. Macromolecules. 2017;218:1600458.
139. Pessego M, Mendoza J, da Silva JP, Basilio N, Garcia-Rio L. Unveiling 160. Huynh UTD, Lerbret A, Neiers F, Chambin O, Assifaoui A. Binding of
the formation 1:2 supramolecular complexes between cucurbit[7]uril divalent cations to polygalacturonate: a mechanism driven by the
and a cationic calix[4]arene derivative. Chem. Comm. 2019;55: hydration water. J. Phys. Chem. B. 2016;120:1021-1032.
13828-13831. 161. Kwasny MT, Zhu L, Hickner MA, Tew GN. Thermodynamics of coun-
140. Schönbeck C, Holm R. Exploring the origins of enthalpy-entropy com- terion release is critical for anion exchange membrane conductivity.
pensation by calorimetric studies of cyclodextrin complexes. J. Phys. J. Am. Chem. Soc. 2018;140:7961-7969.
Chem. 2019;123:6686-6693. 162. Lounis FM, Chamieh J, Leclercq L, et al. Interactions between oppo-
141. Schöttler S, Becker G, Winzen S, et al. Protein adsorption is required sitely charged polyelectrolytes by isothermal titration calorimetry: effect
for stealth effect of poly(ethylene glycol)- and poly(phosphoester)- of ionic strength and charge density. J. Phys. Chem B. 2017;121:
coated nanocarriers. Nat. Nanotechnol. 2016;11:372-377. 2684-2694.
142. Shimoyama D, Haino T. Entropy-driven cooperativity in the guest bind- 163. Luo J, Chen K, Yin P, et al. Effect of cation-π interaction on macroionic
ing of an octaphosphonate bis-cavitand. Chem.-Eur. J. 2020;26:3074- self-assembly. Angew. Chem. Int. Ed. 2018;57:4067-4072.
3079. 164. Renard CMGC, Watrelot AA, Le Bourvellec C. Interactions between
143. Singh RP, Hidalgo T, Cazade P-A, et al. Self-assembled cationic polyphenols and polysaccharides: mechanisms and consequences in
β-cyclodextrin nanostructures for siRNA delivery. Mol. Pharm. 2019; food processing and digestion. Trends Food Sci. Technol. 2017;60:
16:1358-1366. 43-51.
144. Sokkalingam P, Shraberg J, Rick SW, Gibb BC. Binding hydrated 165. Taguta J, O'Connor CT, McFadzean B. The effect of the alkyl chain
anions with hydrophobic pockets. J. Am. Chem. Soc. 2016;138:48-51. length and ligand type of thiol collectors on the heat of adsorption and
145. Suating P, Nguyen TT, Ernst NE, et al. Proximal charge effects on floatability of sulphide minerals. Miner. Eng. 2017;110:145-152.
guest binding to a nonpolar pocket. Chem. Sci. 2020;11:3656-3663. 166. Tesmar A, Wyrzykowski D, Munoz E, et al. Simultaneous determina-
146. Uria-Canseco E, Perez-Casas S, Navarrete-Vazquez G. Thermody- tion of thermodynamic and kinetic parameters of aminopolycarbonate
namic characterization of the inclusion complexes formation between complexes of cobalt(II) and nickel(II) based on isothermal titration calo-
antidiabetic new drugs and cyclodextrins. J. Chem. Thermodyn. 2019; rimetry data. J. Mol. Recognit. 2017;30:e2589.
129:416-422. 167. Watrelot AA, Day MP, Schulkin A, et al. Oxygen exposure during red
147. von Krbek LKS, Schalley CA, Thordarson P. Assessing cooperativity in wine fermentation modifies tannin reactivity with poly-l-proline. Food
supramolecular systems. Chem. Soc. Rev. 2017;46:2622-2637. Chem. 2019;297:124923.
148. Wang XY, Hu YH, Wei H. Nanozymes in bionanotechnology: from 168. Di Trani JM, De Cesco S, O'Leary R, et al. Rapid measurement of
sensing to therapeutics and beyond. Inorg. Chem. Front. 2016;3: inhibitor binding kinetics by isothermal titration calorimetry. Nat.
41-60. Commun. 2018;9:893.
149. Wu G, Olesinska M, Wu Y, Matak-Vinkovic D, Scherman OA. Mining 169. Di Trani JM, Moitessier N, Mittermaier AK. Measuring rapid time-
2:2 complexes from 1:1 stoichiometry: formation of cucurbit[8]uril- scale reaction kinetics using isothermal titration calorimetry. Anal.
diarylviologen quaternary complexes favored by electron-donating sub- Chem. 2017;89:7022-7030.
stituents. J. Am. Chem. Soc. 2017;139:3202-3208. 170. Di Trani JM, Moitessier N, Mittermaier AK. Complete kinetic charac-
150. Wu JJX, Wang XY, Wang Q, et al. Nanomaterials with enzyme-like terization of enzyme inhibition in a single isothermal titration calorimet-
characteristics (nanozymes): next-generation artificial enzymes (II). ric experiment. Anal. Chem. 2018;90:8430-8435.
Chem. Soc. Rev. 2019;48:1004-1076. 171. Erlekam F, Igde S, Roblitz S, Hartmann L, Weber M. Modeling of mul-
151. Wu Y, Xu L, Shen Y, et al. The smallest cucurbituril analogue with high tivalent ligand-receptor binding measured by kinITC. Computation.
affinity for Ag+. Chem. Commun. 2017;53:4070-4072. 2019;7:46.
16 of 17 FALCONER ET AL.

172. Li D, Chen L, Wang R, Liu R, Ge G. Synergetic determination of ther- 191. McNally KE, Faulner R, Steinberg F, et al. Retriever is a multiprotein
modynamic and kinetic signatures using isothermal titration calorime- complex for retromer-independent endosomal cargo recycling. Nat. Cell
try: a full-curve-fitting approach. Anal. Chem. 2017;89:7130-7138. Biol. 2017;19:1214-1225.
173. Sung HL, Nesbitt DJ. Novel heat-promoted folding dynamics of the 192. Scheuermann TH, Brautigam CA. High-precision, automated integra-
yybP-ykoY manganese riboswitch: kinetic and thermodynamic studies tion of multiple isothermal titration calorimetric thermograms: new fea-
at the single-molecule level. J. Phys. Chem. B. 2019;123:5412-5422. tures of NITPIC. Methods. 2015;76:87-98.
174. Vander Meulen KA, Horowitz S, Trievel RC, Butcher SE. Measuring 193. Houtman JC, Brown PH, Bowden B, et al. Studying multisite binary
kinetics of molecular association by isothermal titration calorimetry. and ternary protein interactions by global analysis of isothermal titra-
Calorimetry. 2016;567:181-213. tion calorimetry data in SEDPHAT: application to adaptor protein com-
175. Archer WR, Fiorito A, Heinz-Kunert SL, MacNicol PL, Winn SA, plexes in cell signaling. Protein Sci. 2007;16:30-42.
Schulz MD. Synthesis and rare-earth-element chelation properties of 194. Zhao H, Piszczek G, Schuck P. SEDPHAT - A platform for global ITC
linear poly(ethylenimine methylenephosphonate). Macromolecules. analysis and global multi-method analysis of molecular interactions.
2020;53:2061-2068. Methods. 2015;76:137-148.
176. Hu Y, Guo X, Wang J. Biosorption of Sr2+ and Cs+ onto Undaria 195. Brautigam CA. Calculations and publication-quality illustrations for
pinnatifida: isothermal titration calorimetry and molecular dynamics analytical ultracentrifugation data. Methods Enzymol. 2015;562:
simulation. J. Mol. Liq. 2020;319:114146. 109-133.
177. Kato S, Drout RJ, Farha OK. Isothermal titration calorimetry to investi- 196. Berlow RB, Dyson HJ, Wright PE. Functional advantages of dynamic
gate uremic toxins adsorbing onto metal-organic frameworks. Cell Rep. protein disorder. FEBS Lett. 2015;589:2433-2440.
Phys. Sci. 2020;1:100006. 197. Dyson HJ, Wright PE. Intrinsically unstructured proteins and their
178. Oliva E, Mathiron D, Bertaut E, et al. Physico-chemical studies of res- functions. Nat. Rev. Mol. Cell Biol. 2005;6:197-208.
veratrol, methyl-jasmonate and cyclodextrin interactions: an approach 198. Freire E, Schon A, Velazquez-Campoy A. Isothermal titration calorim-
to resveratrol bioproduction optimization. RCS Adv. 2018;8:1528- etry: general formalism using binding polynomials. Methods Enzymol.
1538. 2009;455:127-155.
179. Reese HR, Shanahan CC, Lembo J, Tsonev L, Hirsh A, Menegatti S. 199. Freire E. Do enthalpy and entropy distinguish first in class from best in
Chromatographic assay to probe the binding energy and mechanisms of class? Drug Discov Today. 2008;13:869-874.
homologous proteins to surface-bound ligands. J. Chromatogr. B. 200. Ladbury JE, Klebe G, Freire E. Adding calorimetric data to decision
2020;1136:121927. making in lead discovery: a hot tip. Nat. Rev. Drug Discov. 2010;9:
180. Reyhanitash E, Fufachev E, Van Munster KD, et al. Recovery and con- 23-27.
version of acetic acid from a phosphonium phosphinate ionic liquid to 201. García-Fuentes L, Baro  n C, Mayorga OL. Influence of dynamic power
enable valorization of fermented wastewater. Green Chem. 2019;21: compensation in an isothermal titration microcalorimeter. Anal. Chem.
2023-2034. 1998;70:4615-4623.
181. Rodler A, Ueberbacher R, Beyer B, Jungbauer A. Calorimetry for 202. Egawa T, Tsuneshige A, Suematsu M, Yonetani T. Method for deter-
studying the adsorption of proteins in hydrophobic interaction chroma- mination of association and dissociation rate constants of reversible
tography. Prep. Biochem. Biotechnol. 2019;49:1-20. bimolecular reactions by isothermal titration calorimeters. Anal. Chem.
182. Simoes-Cardoso JC, Kojo H, Yoshimoto N, Yamamoto S. Microcalori- 2007;79:2972-2978.
metric analysis of the adsorption of lysozyme and cytochrome c onto 203. Velazquez-Campoy A, Lo  pez-Mayorga O, Cabrerizo-Vilchez MA.
cation-exchange chromatography resins: influence of temperature on Determination of the rigorous transfer function of an isothermal titra-
retention. Langmuir. 2020;36:3336-3345. tion microcalorimeter with Peltier compensation. J. Therm. Anal.
183. Sprakel LMJ, Schuur B. Thermal activity in affinity separation tech- Calorim. 1999;57:343-359.
niques such as liquid-liquid extraction analyzed by isothermal titration 204. Vilums M, Zweemer AJ, Yu Z, et al. Structure-kinetic relationships–an
calorimetry (ITC) and accuracy analysis of the technique in the molar overlooked parameter in hit-to-lead optimization: a case of
concentration domain. Ind. Eng. Chem. Res. 2018;57:12574-12582. cyclopentylamines as chemokine receptor 2 antagonists. J. Med. Chem.
184. Sprakel LMJ, Keijsper DJ, Nikolova AL, Schuur B. Predicting solvent 2013;56:7706-7714.
effects on relative volatility behavior in extractive distillation using iso- 205. Cornish-Bowden A. Fundamentals of Enzyme Kinetics. London, UK:
thermal titration calorimetry (ITC) and molecular modeling (MM). Butterworth & Co.; 1979.
Chem. Eng. Sci. 2019;210:115203. 206. Transtrum MK, Hansen LD, Quinn C. Enzyme kinetics determined by
185. Sprakel LMJ, Schuur B. Molecular design and engineering for affinity single-injection isothermal titration calorimetry. Methods. 2015;76:
separation processes using isothermal titration calorimetry (ITC) and 194-200.
molecular modelling (MM). J. Mol. Liq. 2019;283:312-324. 207. Lonhienne T, Baise E, Feller G, Bouriotis V, Gerday C. Enzyme activ-
186. Sprakel LMJ, Schuur B. Improving understanding of solvent effects on ity determination on macromolecular substrates by isothermal titration
intermolecular interactions in reactive liquid-liquid extraction with iso- calorimetry: application to mesophilic and psychrophilic chitinases. Bio-
thermal titration calorimetry and molecular modelling. J. Ind. Eng. chim Biophys Acta. 2001;1545:349-356.
Chem. 2019;72:364-373. 208. Lonhienne TGA, Reilly PEB, Winzor DJ. Further evidence for the reli-
187. Sutton JE, Roy S, Chowdhury AU, et al. Molecular recognition at min- ance of catalysis by rabbit muscle pyruvate kinase upon isomerization
eral interfaces: implications for the beneficiation of rare earth ores. ACS of the ternary complex between enzyme and products. Biophys. Chem.
Appl. Mater. Interfaces. 2020;12:16327-16341. 2003;104:189-198.
188. Wanhala AK, Doughty B, Bryantsev VS, et al. Adsorption mechanism 209. Zebisch M, Krauss M, Schäfer P, Sträter N. Crystallographic evidence
of alkyl hydroxamic acid onto bastnasite: fundamental steps toward for a domain motion in rat nucleoside triphosphate diphosphohydrolase
rational collector design for rare earth elements. J. Colloid Interface (NTPDase) 1. J. Mol. Biol. 2012;415:288-306.
Sci. 2019;553:210-219. 210. Biedermann F, Scheider H-J. Experimental binding energies in supra-
189. Parton RG, Tillu VA, Collins BM. Caveolae. Curr. Biol. 2018;28:R367- molecular complexes. Chem. Rev. 2016;116:5216-5300.
R420. 211. Mako TL, Racicot JM, Levine M. Supramolecular luminescent sensors.
190. Chen K-E, Healy MD, Collins BM. Towards understanding of Chem. Rev. 2018;199:322-477.
endosomal trafficking by Retromer and Retriever. Traffic. 2019;20: 212. Das D, Assaf KI, Nau WM. Applications of curcurbiturils in medicinal
465-478. chemistry and chemical biology. Front. Chem. 2019;7:619.
FALCONER ET AL. 17 of 17

213. Xia D, Wang P, Ji X, Khashab NM, Sessler JL, Huang F. Functional supra- 220. Marcus Y, Kolarik Z. Thermodynamics of liquid-liquid distribution reac-
molecular polymeric networks: the marriage of covalent polymers and tions. III. The U(VI), Nd(III) or Ho(III) nitrate-water-di(2-ethylhexyl) phos-
macrocycle-based host-guest networks. Chem. Rev. 2020;120:6070-6123. phoric acid-hydrocarbon diluent extraction system. J. Inorg. Nucl.
214. Lumry R, Rajendra S. Enthalpy–entropy compensation phenomena in Chem. 1976;38:1069-1073.
water solutions of proteins and small molecules: a ubiquitous properly 221. Zalupski PR, Nash KL. Two-phase calorimetry. I. Studies on the ther-
of water. Biopolymers. 1970;9:1125-1227. modynamics of lanthanide extraction by bis(2-ethylHexyl) phosphoric
215. Schuur B, Brouwer T, Smink D, Sprakel LMJ. Green solvents for sus- acid. Solvent Extr. Ion Exch. 2008;26:514-533.
tainable separation processes. Curr. Opin Green Sustainable Chem. 222. Pethica BA. Misuse of thermodynamics in the interpretation of isother-
2019;18:57-65. mal titration calorimetry data for ligand binding to proteins. Anal. Bio-
216. Cuypers R, Burghoff B, Marcelis ATM, Sudhölter EJR, De Haan AB, chem. 2015;472:21-29.
Zuilhof H. Complexation of phenols and thiophenol by phosphine 223. Tellinghuisen J. Isothermal titration calorimetry at very low c. Anal.
oxides and phosphates. extraction, isothermal titration calorimetry, and Biochem. 2008;373:395-397.
ab initio calculations. J. Phys. Chem. A. 2008;112:11714-11723.
217. Sprakel LMJ, Schuur B. Solvent developments for liquid-liquid extraction of
carboxylic acids in perspective. Sep. Purif. Technol. 2019;211:935-957.
218. Martak J, Schlosser S. New mechanism and model of butyric acid How to cite this article: R. J. Falconer, B. Schuur, and A.
extraction by phosphonium ionic liquid. J. Chem. Eng. Data. 2016;61:
K. Mittermaier, Applications of isothermal titration calorimetry in
2979-2996.
219. Marcus Y, Kolarik Z. Thermodynamics of liquid-liquid distribution reac- pure and applied research from 2016 to 2020, J Mol Recognit
tions. I. Dioxouranium(VI) nitrate-water-tributyl phosphate-n-dodecane (2021), e2901. https://doi.org/10.1002/jmr.2901
system. J. Chem. Eng. Data. 1973;18:155-163.

You might also like