You are on page 1of 12

pubs.acs.

org/bc Review

Supramolecular “Click Chemistry” for Targeting in the Body


Christopher J. Addonizio,‡ Brant D. Gates,‡ and Matthew J. Webber*

Cite This: Bioconjugate Chem. 2021, 32, 1935−1946 Read Online

ACCESS Metrics & More Article Recommendations

ABSTRACT: The fields of precision imaging and drug delivery


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

have revealed a number of tools to improve target specificity and


Downloaded via INDIAN INST OF TECH MADRAS on February 10, 2023 at 06:32:17 (UTC).

increase efficacy in diagnosing and treating disease. Biological


molecules, such as antibodies, continue to be the primary means of
assuring active targeting of various payloads. However, molecular-
scale recognition motifs have emerged in recent decades to achieve
specificity through the design of interacting chemical motifs. In this
regard, an assortment of bioorthogonal covalent conjugations offer
possibilities for in situ complexation under physiological conditions.
Herein, a related concept is discussed that leverages interactions
from noncovalent or supramolecular motifs to facilitate in situ
recognition and complex formation in the body. Classic supra-
molecular motifs based on host−guest complexation offer one such
means of facilitating recognition. In addition, synthetic bioinspired motifs based on oligonucleotide hybridization and coiled-coil
peptide bundles afford other routes to form complexes in situ. The architectures to include recognition of these various motifs for
targeting enable both monovalent and multivalent presentation, seeking high affinity or engineered avidity to facilitate conjugation
even under dilute conditions of the body. Accordingly, supramolecular “click chemistry” offers a complementary tool in the growing
arsenal targeting improved healthcare efficacy.

1. INTRODUCTION molecules such as antibodies to localize therapeutics to a site


A primary goal in drug delivery is the pursuit of technologies to of need, targeting on the basis of a disease-relevant
increase the fraction of drug delivered to a site of need.1 One biomarker.8−10 Several antibody−drug conjugates have been
key characteristic of an active pharmaceutical agent is its recently FDA-approved,11 consisting of a therapeutic agent
therapeutic index, a ratio of its toxic dose (TD50) to its attached via a labile linker to a monoclonal antibody with
effective dose (ED50). Accordingly, drug delivery technologies affinity for specific biomarkers.12 Antibodies or aptamers can
seek to increase the therapeutic index through two parallel likewise be used for active targeting of nanoscale drug
mechanisms: (i) attenuating the systemic activity of a drug carriers.13−15 These and other methods of active targeting
through encapsulation and/or prodrug methodologies to are often limited by the availability of targeting antibodies
enable higher dosing without toxicity,2,3 and (ii) ensuring a specific to the disease of interest; the use of larger constructs or
larger fraction of the delivered agent reaches the physiological drug carriers also limits tissue perfusion, carries risk of off-site
site of need to increase effectiveness of the therapeutic agent.4 accumulation, and may lead to prolonged circulation while
Drug delivery can be achieved through passive accumulation of shedding active drug systemically.16,17 For example, only
drug carriers, sometimes taking advantage of leaky vasculature 0.001−0.01% of an injected monoclonal antibody, and by
that is a pathophysiological hallmark of certain diseases.5,6 The logical extension an antibody−drug conjugate, localizes to a
first FDA-approved nanoscale drug delivery technology, Doxil, tumor site in humans.18,19 Meanwhile, nanoparticles targeted
is a PEGylated liposomal formulation of doxorubicin that
with a clinically validated antibody have demonstrated local
functions through such a mechanism.7 Accordingly, early
accumulation of <1% in vivo.20 As such, there remains a need
efforts in the field of drug delivery often sought to increase the
therapeutic index through a combination of sequestering toxic
agents within nanoscale carriers and leveraging physiologic Received: June 24, 2021
features of diseased tissue to promote preferential accumu- Revised: July 13, 2021
lation. Published: August 20, 2021
Another strategy broadly explored in the field of drug
delivery to increase the therapeutic index is active targeting.
These routes commonly leverage affinity from biological

© 2021 American Chemical Society https://doi.org/10.1021/acs.bioconjchem.1c00326


1935 Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

Figure 1. (A) Schematic of dynamic supramolecular interactions used for in situ association, leveraging complementary motifs with one attached to
an entity for pretargeting and another attached to the desired agent to be delivered. (B) Generalized overview of this two-component approach to
targeting via in situ complex formation. (C) Examples of common covalent bioorthogonal reactions used in pretargeting applications for in situ
covalent bond formation. (D) Overview of the dynamic supramolecular motifs described herein, which form association complexes through
noncovalent molecular recognition.

to explore new technologies in order to more effectively deliver recognition in the body (Figure 1A). These synthetic
therapeutics to sites of need. supramolecular motifs are attractive in the development of
In the field of bioconjugate chemistry, a molecular-scale drug delivery platforms due to their scalable, tunable, and
pretargeting approach has been demonstrated using different molecularly well-defined characteristics.32−36 The various
bioorthogonal ligations to capture circulating agents at specific motifs used for noncovalent recognition in the body include
sites in the body through spontaneous formation of covalent host−guest macrocycle complexes, complementary oligonu-
bonds.21−24 Spatial localization can be achieved within the cleotide segments, and coiled-coil peptide assemblies (Figure
body by covalent bond formation in situ using a two-step 1D). The mechanisms that underlie recognition incorporate
application of a pretargeted entity bearing one component of a pseudospecificity through unnaturally high affinity and/or high
bioorthogonal motif followed by application of the second effective affinity through engineering multivalent motifs to
motif attached to a drug or imaging agent (Figure 1B,C).25−29 enable avidity. While certain of these motifs (e.g., host−guest)
The attachment of a drug to a group for click chemistry offers are subject to competition from naturally occurring com-
certain prodrug benefits of attenuated systemic activity; such pounds and thus not fully bioorthogonal, outcomes resembling
agents also incorporate labile linkages for subsequent release of orthogonality can be realized through motif selection and
the active therapeutic via linker hydrolysis following local design to tune the affinity well in excess of naturally present
accumulation. Others have demonstrated so-called “click-to- competitors, or by engineering avidity to gain advantage.35
release” and “catch and release” chemistries wherein an active Molecular-scale approaches to drug targeting through both
agent releases from its bioorthogonal motif-bearing prodrug in situ covalent bioconjugation and noncovalent recognition
precursor by spontaneous ring isomerization simultaneous to offer certain distinctions relative to drug delivery methods
in situ formation of a covalent bond.30,31 using active biological targeting. In one manifestation of this
The present review focuses on related molecular-scale two-step molecular-scale approach, pretargeting a site of
approaches akin to bioorthogonal click chemistry, instead interest with an antibody or related biomolecule maintains
using noncovalent supramolecular interactions for in situ the benefits of biological recognition of disease biomarkers.
1936 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

However, separating the drug from the antibody reduces the such as azide−DBCO (2.3 M−1 s−1; from ref 39) and
risk of undesired release during prolonged circulation. In other tetrazene−trans-cyclooctene (3100 M−1 s−1; from ref 40).
uses, pretargeting and capture of a circulating therapeutic using Typically, higher affinity interactions will have concomitantly
a localized material suffers from a requirement for a priori slower koff and thus have a longer lifetime of complexation
knowledge in applying the pretargeting material to guide once formed.41
subsequent administration of the agent, and as such may be The effective doses of different therapeutics vary, but an
more limited in its practical application. Small molecule assumption for serum concentrations on the order of ∼[nM]
prodrugs, prepared by modifying a therapeutic with a for most drugs defines (roughly) the target Keq needed for
molecular-scale targeting motif, offer the benefits of attenuated complex formation when considering the use of a particular
activity in systemic circulation, more extensive tissue perfusion, motif in targeting therapeutics; this implies Keq may need to be
and rapid clearance owing to small size relative to antibodies or greater than ∼108 M−1 to drive complex formation in vivo.
even larger nanoscale carriers. As such, the general concept Given this extent of dilution expected for uses in the body, as
introduced here for noncovalent molecular recognition of well as a variety of possible competitors for certain classes of
synthetic motifs in the context of in situ targeting of interactions, monovalent affinity may thus not be sufficient for
therapeutics and imaging agents should be framed with these some recognition motifs to facilitate efficient supramolecular
benefits and drawbacks in mind. With the aim of specifically complexation. Accordingly, other design approaches may
focusing on uses of synthetic noncovalent molecular couple multiple lower affinity interactions on a defined scaffold
recognition motifs, this review will also (by necessity) not to achieve a higher effective affinity, a phenomenon referred to
cover voluminous work in the areas of biomolecular-based as avidity. The complexes formed between antibody and
recognition using antibodies, aptamers, peptides, or other antigen, with multivalent display of a specific binding epitope
common biomolecular affinity agents. Instances where such on the antibody, illustrate the uses of avidity in nature.42
affinity agents are used in the context of pretargeting a Binding events in multivalent systems do not necessarily occur
synthetic motif for subsequent noncovalent recognition- simultaneously, but they are likewise not completely
mediated targeting will be discussed. independent. The physical tethering of multiple binding motifs
creates an elevated local concentration through the close
2. THERMODYNAMICS OF RECOGNITION proximity of binding sites to drive complex formation.43 In
The propensity for a noncovalent complex to form in the dilute other instances, both motifs may be presented on multivalent
environment of the body is governed by thermodynamics. In scaffolds, leading to an overall reduction in the effective koff
the simplest case of a monovalent interaction, the dynamic given the asynchronous time scale of dynamic complex
process of recognition proceeds as follows: exchange for individual binding sites as multiple dynamic
interactions drive greater complexation between the two
[A] + [B] V [A·B] scaffolds.44 In this way, the use of multivalent systems may
where [A] and [B] are the concentrations of the free binding compensate for the low affinity of an individual motif to
pairs and [A·B] is the concentration of the formed complex. facilitate recognition even under the conditions of dilution
From the law of mass action, an equilibrium constant, Keq expected for applications in the body.
(sometimes denoted KA), can be derived as follows:
3. HOST−GUEST RECOGNITION
[A·B] Host−guest chemistry, characterized as the noncovalent
Keq =
[A][B] association of a small molecule guest within the portal of a
This quantity has standard units of [M−1] for a 1:1 monovalent host macrocycle, is among the most recognizable of supra-
interaction. Keq is commonly referred to as the affinity of an molecular motifs. The affinity of different interactions can vary
interaction. It is conventional in some systems to express the substantially, though complexes have been demonstrated that
reciprocal of this value, 1/Keq = KD, yielding units of [M] and form at high affinity (e.g., Keq > 1010 M−1) and are therefore
serving to define the dilution concentration for spontaneous resistant to dilution and native competition, in pursuit of
dissociation of a complex. A number of synthetic host various bioconjugation-based applications.35,45,46 Many syn-
molecules have been reported to bind with an array of thetic macrocyclic host molecules are known, including crown
complementary guest motifs, enabling Keq to be tuned by ethers, cryptands, cyclodextrins (CD), cucurbit[n]urils (CB-
molecular design or in response to a biologically relevant [n]), calix[n]arenes, and pillar[n]arenes.32 High-affinity design-
stimulus.37 A higher value of Keq thus signifies a more stable er molecules have also been revealed from host−guest
complex that exhibits preferential formation even under dilute complexes that form highly fluorescent and stable dyes.47,48
conditions. The value of Keq is also related to the rates of Of the motifs used in the context of drug delivery, CD
dynamic formation, kon, and dissociation, koff, of the complex, macrocycles prepared enzymatically from starch constitute the
as follows: most broadly explored and readily available macrocycles.49,50
CDs have rigid conical geometry comprised of different
kon numbers of glucopyranoside subunits, to include α-CD (6), β-
Keq ≈ CD (7), and γ-CD (8), enabling size-mediated selectivity in
koff
their binding to different guests. Their hydrophobic interiors
For a 1:1 monovalent interaction, kon has units of [M−1 s−1] and hydrophilic exteriors allow guest encapsulation within the
and koff has units of [s−1]. The rate of complex formation for cavity, taking advantage of both hydrophobic and van der
some supramolecular motifs has been found to occur near the Waals interactions.51 Binding between CDs and their guests
diffusion limit (∼108 M−1 s−1);38 this suggests a possible occur with Keq values not typically exceeding ∼105 M−1, the
benefit of fast association for supramolecular motifs when order expected for binding between β-CD and an adamantane
compared to common covalent bioorthogonal conjugations guest.52,53 The CB[n] family of macrocycles, composed of [n]
1937 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

Figure 2. Antibody-based pretargeting for delivery to sites of disease bearing a specific biomarker, coupled with a secondary delivery approach and
in situ targeting driven by (A) monovalent host−guest motifs or (B) complementary oligonucleotide sequences designed for hybridization. (Panel
A inspired by refs 70 and 71; Panel B inspired by ref 100.)

repeating glycoluril subunits, constitutes another useful The types of guest molecules useful for this purpose are amine-
macrocycle for guest binding in aqueous environments.54−57 containing ferrocene and adamantane derivatives, exhibiting
Glycoluril subunits afford a symmetric macrocycle with a rigid Keq values in the range of 1010−15 M−1 in binding CB[7].67−69
hydrophobic cavity and two identical carbonyl-fringed portals. Recognition using these motifs has thus been explored for a
CB[7] macrocycles bind to adamantane-class guests with Keq variety of imaging and therapeutic applications.
up to ∼1017 M−1, well in excess of what is achievable by other The use of supramolecular host−guest motifs for in situ
macrocycles or even natural motifs such as biotin−avidin.58−60 targeting typically comprises a pretargeting step followed by
High-affinity binding is possible through a combination of the subsequent administration of an agent for imaging or therapy.
hydrophobic association and volume-filling of the macrocycle In this context, an antibody may be used for the initial
cavity coupled along with electrostatic interactions between pretargeting to deposit either a host or guest at the site of
aliphatic-adjacent protonating groups and the electronegative interest, followed by subsequent addition of the desired agent
carbonyl-fringed portal.61 Accordingly, the differing spectrum attached to the complementary binder (Figure 2A).70 Using
of affinity offered by CD and CB[n] macrocycles affords pretargeting principles, in situ formation of host−guest
distinct opportunities for host−guest recognition and complex complexes have been explored in live nematodes (C. elegans)
formation in the conditions of the body, as described herein. and mice.71 The studies in nematodes coupled complementary
3.1. Monovalent Host−Guest Recognition. In the FRET pairs to CB[7] and guest, verifying sequential
context of in situ recognition, protein-based motifs have been administration of the motif resulted in complex formation in
extensively explored, yet can exhibit slow biodistribution and situ. This system was then explored in mice for in vivo cancer
clearance.62−64 Long circulation times to reach a target may imaging. CB[7] was covalently attached to cetuximab, an
limit their use to deliver short-lived isotopes for radioimaging antibody recognizing epidermal growth factor receptor that is
and increase the possibility for enzymatic degradation in used clinically in treating colorectal, neck, and lung cancers.
circulation.65,66 Host−guest motifs, with small molecule guests Following pretargeting with the CB[7]−antibody conjugate,
on the order of ∼200 g/mol and macrocycles on the order of adamantane linked to a near-infrared cyanine dye was found to
1200 g/mol, may thus offer a variety of possible benefits. accumulate at the tumor site for selective tumor imaging.71
CB[n] macrocycles, and in particular the water-soluble and Pretargeting has also been achieved by local injection of a
high-affinity CB[7] variant, have been most explored in the polymer hydrogel presenting CB[7], with subsequent systemic
context of monovalent host−guest recognition in the body. administration of a guest-linked agent.72 A series of guests
1938 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

Figure 3. Methodology to assess the affinity needed for in situ recognition and targeting. A hydrogel presenting pendant CB[7] macrocycles was
implanted locally at a site. Subsequently, a near-infrared fluorescent probe (Cy5) modified with ferrocene guests having different affinities for
CB[7] (Fc-N:1012 M−1 vs Fc-O:109 M−1) was administered systemically. Through in vivo imaging, the amount of dye localized and retained at the
site presenting CB[7] was then quantified. Subtle differences in the guest structure, altering their resulting affinity for CB[7], led to dramatic
changes in the effectiveness of in situ complexation. (Figure inspired by ref 72.)

ranging in Keq from ∼109 to 1012 M−1 fused to a near-infrared sating for the relatively low Keq of a monovalent CD host−
fluorescent dye were explored to assess the role of affinity for in guest complex compared to those observed for CB[7]. In one
situ complex formation at the site of the CB[7]-rich depot such design, adamantane-functionalized albumin aggregates
(Figure 3). These studies identified complexes between CB[7] were used to pretarget sites for subsequent delivery of β-CD-
and an amino-ferrocene guest with Keq of ∼1012 M−1 that modified polymers carrying agents for either fluorescence or
achieved substantial localization, whereas the dye bound to a SPECT imaging modalities (Figure 4).73 Pretargeting with the
different ferrocene guest with Keq of ∼109 M−1 showed no multivalent albumin aggregates followed by multivalent agent
accumulation. For the high-affinity case, ∼4% of the delivery offered a ∼16-fold increase in the accumulation of the
administered agent homed within a few hours; the remainder agent in the liver and a 4.5-fold increase in the lungs when
was rapidly cleared over this same time. This figure is
impressive in context of the typical targeting efficiency
achieved by antibodies, as discussed previously here. The
depot site could be serially reloaded, with site retention of the
bound agent for multiple weeks following administration. This
same high-affinity guest motif was then conjugated to the
chemotherapeutic doxorubicin to create a prodrug for
integration with supramolecular targeting. By injecting the
CB[7]-rich hydrogel near a tumor, the therapeutic efficacy of
supramolecular homing was evaluated in comparison to a
prodrug variant with no affinity for CB[7]. In this case, the
targeted prodrug demonstrated a significant reduction in the
tumor growth rate, with the effect extending for weeks Figure 4. Schematic overview of the use of guest-modified serum
albumin aggregates, which following administration preferentially
following initial dosing. accumulate at sites of disease. Subsequently, a cyclodextrin-modified
3.2. Multivalent Host−Guest Recognition. The uses of polymer was administered to enable multivalent in situ complexation
CD for in situ complex formation have primarily leveraged and agent delivery to the site bearing these guest-modified albumin
multivalent constructs to introduce avidity, thereby compen- aggregates. (Figure inspired by ref 73.)

1939 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

Figure 5. Schematic overview of the concept of drug-free macromolecular therapeutics. This concept has been demonstrated for motifs derived
from both oligonucleotide hybridization and coiled-coil peptide association. A cancer cell of interest was first pretargeted with a reagent for
biological recognition, an antibody or antibody fragment, that was modified with one-half of the desired recognition motif. Subsequently, a
multivalent polymer bearing the complementary recognition motif was administered. The polymer scaffold, by simultaneously binding multiple
surface-presented motifs, serves to noncovalently link the receptors on the cell leading to triggered cell death. (Figure inspired by works from
Kopecek and colleagues, such as refs 107 and 131.)

compared using SPECT imaging to pretargeting with sequences for recognition in the body must account for specific
unmodified albumin aggregates. Further studies using albumin operating conditions to ensure reliable complex formation. As
aggregates to pretarget a β-CD-modified polymer leveraged the focus here is on the use of synthetic noncovalent
dual-isotope imaging (99mTc on the albumin particles and 111In recognition motifs for targeting applications, the many
on the polymer) to validate colocalization of the two important uses of aptamers for recognition of biomolecules
components in vivo.74 As such, multivalent scaffolds presenting falls outside the present scope of this review; the reader is
both host and guest enable the use of CD macrocycles in spite encouraged to explore other relevant reviews on this
of its modest monovalent affinity. topic.86−88
4.1. Monovalent Oligonucleotide Hybridization. One
4. OLIGONUCLEOTIDE HYBRIDIZATION benefit of oligonucleotide-based recognition arises from its
The association of complementary strands of DNA, forming its ease of synthetic modification with molecular cargo.89−92 This
canonical double helix, is one of the most recognizable design tool enables an array of therapeutics or imaging agents
noncovalent motifs in the living world. Synthetic oligonucleo- to be appended to oligonucleotide strands. One salient
tides thus offer a tunable and biologically relevant affinity motif example of this approach is found in the field of molecular
for noncovalent complex formation, toward many therapeutic beacons, wherein binding to a target DNA or RNA strand
uses.75−77 This is highlighted by the decades of work triggers a hybridization-mediated unfolding of the beacon and
evaluating the therapeutic potential of small interfering typically an increase in fluorescence relative to a quenched
RNAs (siRNA), where therapeutic function arises specifically state in the folded form.93,94 Early work using this technology
from recognition and binding target mRNA to transiently in vivo relied on aptamer-mediated recognition to facilitate
inhibit protein expression.78,79 Oligonucleotide strand com- beacon rearrangement for imaging.95,96 Related aptamer-
plexation, a process known as hybridization, is driven by targeted technologies have also been used to deliver drugs
Watson−Crick base pairing with lateral hydrogen bond bound via intercalation with double-stranded regions of the
formation between complementary bases offering an enthalpic probe.97 Other technologies evaluated in vitro suggest the
driving force.80−82 The vertical stacking of aromatic bases in possibility that aptamer-based constructs with a pendant
the formed helical structure also contributes a favorable driving oligonucleotide tail can be used for pretargeting, with
force for hybridization via hydrophobic and π−π interac- subsequent delivery of a probe coupled to the complementary
tions.83 The number of base pairs, and by extension the oligonucleotide strand.98 A similar pretargeting approach was
number of hydrogen bonds and π−π interactions, dictates the also demonstrated in vitro using copper-free click chemistry to
binding affinity between oligonucleotide strands; this affinity is modify the cell surface with oligonucleotides, subsequently
highly dependent on environmental conditions such as delivering a complementary strand linked to a probe for
temperature, concentration, and osmolarity.84,85 For example, imaging.99 However, the use of oligonucleotide hybridization
the complexation of model 10-base strands in 3 mM buffer specifically for targeting molecular beacons in vivo has been less
exhibits a Keq of ∼5 × 107 M−1 at 15 °C, reducing to ∼3 × 105 commonly explored.
M−1 at 35 °C as noncovalent interactions become less Targeting via monovalent oligonucleotide hybridization has
favorable.85 Meanwhile, 20-base strands have Keq values been demonstrated in the context of antibody-mediated
(∼108 M−1) that are much less temperature-dependent over pretargeting for PET-CT imaging (Figure 2B).100 In this
the same range. For both lengths, affinity also increases by work, the cetuximab antibody was modified with a 17-mer L-
∼1−2 orders of magnitude for interactions in a buffer of higher DNA segment. Subsequently, a mirror-image 17-mer L-DNA
salt (10 mM). Accordingly, the design of oligonucleotide segment connected to a 64Cu radionuclide chelator was
1940 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

Figure 6. Schematic of in situ surface regeneration using a toehold-mediated strand exchange approach. This general strategy illustrates a route to
use designed recognition motifs to regenerate the presentation of active sites on a device surface. (Figure inspired by ref 113.)

administered for localization by in situ hybridization. Efforts to prepare these constructs with oligonucleotides have
Biodistribution studies performed in vivo demonstrated relied on morpholino oligomers, synthetic analogues of
significant tumor accumulation and contrast enhancement oligonucleotides consisting of DNA bases attached to a
when using this pretargeting approach for radionuclide backbone of methylenemorpholine rings linked through
delivery. phosphorodiamidate groups, intended to facilitate enhanced
In a related context, synthetic oligonucleotide analogues stability in serum.110 The first design leveraged an antibody
known as peptide nucleic acids (PNAs) may also enable fragment (Fab′) against a marker for B-cell lymphoma
recognition in the context of targeting. The nucleobases of (CD20), fusing this to a morpholino strand for pretargeting
PNAs form stable duplexes with DNA or RNA segments and cancer cells. A polymer based on N-(2-hydroxypropyl)-
may also be designed to recognize other PNAs.101−104 methacrylamide (HPMA) with pendant complementary
Accordingly, PNA recognition has been used in the context morpholino strands was then administered to multimerize
of a two-step pretargeting.105 In this work, a PNA-modified the CD20 cell surface receptors and induce apoptosis.107
protein was first administered for passive accumulation at sites These constructs demonstrated therapeutic function in a
of infection or tumors, and subsequently a PNA radiolabeled disseminated B-cell lymphoma model in mice, pointing to a
with 99mTc was administered for localization by in situ key benefit of this approach against metastatic disease.
hybridization. Subsequent work on this concept utilized an intact anti-
4.2. Multivalent Oligonucleotide Hybridization. Ef- CD20 antibody (Obinutuzumab) for pretargeting with
forts to increase the effective binding affinity of complementary morpholino strands and induced multimerization with
oligonucleotide strands have entailed developing multivalent morpholino-modified human serum albumin.108 By combining
scaffolds to introduce avidity into the process of targeting. In the intact antibody with multivalent cross-linking, this
one example, recognition via the hybridization of comple- approach enabled two synergistic modes to induce apoptosis.
mentary oligonucleotides has been demonstrated to refill a 4.3. In Situ Strand Displacement. Oligonucleotide
locally applied hydrogel depot.106 In this design, an alginate complexes can be designed to engage in strand displacement
hydrogel modified with oligodeoxynucleotide (ODN) strands through binding to unhybridized segments flanking a double-
was applied locally. Subsequent systemic application of alginate stranded segment, an approach used to facilitate polymer de-
modified with the complementary ODN strands enabled local gelling, site-specific drug release, and surface regenera-
accumulation at the depot through strand recognition. A tion.111−113 This displacement is often initiated through
control of noncomplementary ODN sequences exhibited no toehold-mediated strand exchange, wherein a single-stranded
increased accumulation. The ODN-targeted platform was oligonucleotide binds to an exposed portion of its comple-
evaluated for functional use in vivo in the delivery of a mentary strand that is otherwise engaged in a double helix,
chemotherapeutic, doxorubicin, to the site of a tumor. Mice triggering dissociation of the initial complex as the replacement
treated weekly by systemic application of ODN-modified strand hybridizes.114 Recently, this mechanism was utilized to
alginate strands conjugated to doxorubicin showed a significant regenerate antithrombotic functionality of a surface (Figure
reduction in tumor growth compared to controls, attributable 6).113 To combat degradation of an antithrombotic agent
to ODN hybridization localizing the drug-modified polymer to presented on the device, strand displacement was designed to
the site of the depot. replace the degraded agent and restore antithrombotic
Certain therapeutic benefits arise when using multivalent functionality of the surface. This approach demonstrated a
scaffolds apart from increasing the effective Keq of recognition. significant reduction in fibrin formation. Though not used in
One such example is found in efforts to pretarget using vivo, recognition-mediated strand displacement offers many
oligonucleotide-modified antibodies followed by subsequent possible opportunities to externally control the properties of
recognition on the cell surface of a multivalent oligonucleotide biomedical device interfaces in situ.
scaffold (Figure 5).107,108 The therapeutic effect of this
approach arises from induction of apoptosis due to receptor 5. PEPTIDE COILED-COIL FORMATION
multimerization on the cell surface, leading to demonstrations Engineered coiled-coil peptides, characterized by the arrange-
for a new class of drug-free macromolecular therapeutic.109 ment of α-helical peptides into a superhelix bundle, afford
1941 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

recognition properties with utility in the design of functional outcome of in situ recognition. Herein, a related concept
materials and systems.115,116 Their biological relevance as a leveraging the noncovalent association of synthetic supra-
common structural motif found in nature has inspired molecular motifs is described. Through motif selection and
significant study into both mechanisms of formation and design, high-affinity interactions can be realized to enable de
strategies for sequence manipulation to realize coiled-coil facto specificity and orthogonality in the body. Many of these
motifs comprising a different number (n = 2−6) of both motifs offer kinetic advantages over traditional bioorthogonal
homo- or hetero[n]meric α-helical peptides.117−120 Various chemistries, such as the ability to associate with diffusion-
naturally derived and de novo designs have thus been governed interaction rates. Moreover, the synthetic origins of
demonstrated for coiled-coil recognition, with synthetic these motifs enable tractable multivalent display on polymers,
heterodimeric variants having K eq values up to 10 14 nanoparticles, or related scaffolds to engage avidity and further
M−1.121−124 Such interactions are thus comparable to (or enhance recognition specificity. This approach has even
higher than) high-affinity host−guest or oligonucleotide revealed a new therapeutic class based on drug-free macro-
motifs. The predictable nature of these associations has been molecular architectures.
used to recreate the complex higher-ordered structures of There are remaining challenges that must be navigated to
natural proteins with synthetic variants, for instance, in the more fully exploit the potential of these supramolecular tools
preparation of discrete cage-like assemblies.125,126 In addition, for in situ targeting. The two-step targeting used in many
coiled-coil motifs have been incorporated as a modular systems, while advantageous in limiting off-site accumulation
associating unit in noncovalent preparation of modular drug and systemic drug shedding of often toxic drugs, introduces
carriers.127−130 Accordingly, these interactions offer another complexities and variability with respect to the timing of
class of synthetic noncovalent interactions with promise for in administration of each component. The benefit of broader and
situ recognition in the body. biologically specific systemic surveillance when pretargeting is
5.1. Multivalent Coiled-Coil Recognition. As with work done using antibodies is not captured in cases where a locally
in oligonucleotide systems, coiled-coil interactions have been implanted material depot is used as the pretargeting entity.
explored in conjunction with routes for pretargeting as well as This requirement for a priori knowledge of the desired site of
scaffolds for multivalent presentation toward the concept of action also limits uses for the latter case in disseminated
drug-free macromolecular therapeutics.109 For example, one diseases such as metastatic cancer, yet may remain relevant for
component of a heterodimeric coiled-coil was attached applications in regenerating active signals on implanted
multivalently to HPMA with the complementary α-helical biomedical devices. There are also remaining challenges to
segment attached to a Fab′ with reactivity against CD20 better integrate supramolecular targeting motifs with relevant
(Figure 5).131 This platform showed in vivo efficacy in a mouse methods in prodrug chemistry, such as incorporating analyte-
model of B-cell lymphoma, functioning by cross-linking the or enzyme-sensitive linkers for site-specific drug activation
surface-bound Fab′ on cell surface receptors to induce following homing.
apoptosis.132 The immunogenicity of this platform was studied The emerging concept to use noncovalent association of
in vitro and in vivo, pointing to no specific immunogenicity for supramolecular motifs offers inspiration to reimagine the
the coil-forming peptide motif; this study explored the same diagnosis and treatment of disease. With nature as inspiration
motif prepared from D-isomer peptides and found the for specific noncovalent recognition in physiological con-
enantiomeric peptide coiled-coils to behave similarly to the ditions, recreating these concepts using synthetic tools is a path
originally used L-amino acids.133 Multifluorophore imaging of primed for many possible applications. Accordingly, the
this system further verified in situ assembly of the two concept of supramolecular “click chemistry” for in situ targeting
components on B-cell membranes when administered by this offers a promising direction ripe for further evaluation.


two-step pretargeting approach, noting the importance of the
delay time between administration of the first and second AUTHOR INFORMATION
component to enable localization.134 This system was also
Corresponding Author
found to function when the multivalent HPMA component
was replaced with human serum albumin modified with Matthew J. Webber − University of Notre Dame, Department
multiple copies of one of the coil-forming peptide segments.135 of Chemical & Biomolecular Engineering, Notre Dame,
Related work demonstrated the ability to target cancer cells Indiana 46556, United States; orcid.org/0000-0003-
presenting one-half of a coiled-coil motif with liposomes 3111-6228; Email: mwebber@nd.edu
presenting the complementary peptide, demonstrating in situ Authors
homing in a zebrafish model.136 Accordingly, systems based on Christopher J. Addonizio − University of Notre Dame,
pretargeting and multivalent recognition may also use synthetic Department of Chemical & Biomolecular Engineering, Notre
coiled-coil motifs to facilitate recognition in the body. Dame, Indiana 46556, United States
Brant D. Gates − University of Notre Dame, Department of
6. CONCLUSIONS Chemical & Biomolecular Engineering, Notre Dame, Indiana
In the continued pursuit of new routes to enhance efficacy in 46556, United States
diagnosing and treating disease, strategies for recognition on
Complete contact information is available at:
the molecular scale hold promise. In particular, the use of
https://pubs.acs.org/10.1021/acs.bioconjchem.1c00326
synthetic motifs offers new routes to reliably and efficiently
perform in situ conjugation under dilute conditions in the
Author Contributions
body, even in the presence of salts, proteins, lipids, and other ‡
“sticky” biological entities. The use of small molecules affords C.J.A. and B.D.G. contributed equally to this work.
rapid and extensive tissue perfusion. To date, bioorthogonal Notes
covalent conjugations have offered one means of achieving this The authors declare no competing financial interest.
1942 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry


pubs.acs.org/bc Review

ACKNOWLEDGMENTS (18) Epenetos, A. A., Snook, D., Durbin, H., Johnson, P. M., and
Taylor-Papadimitriou, J. (1986) Limitations of Radiolabeled Mono-
M.J.W. gratefully acknowledges funding support for this work clonal Antibodies for Localization of Human Neoplasms. Cancer Res.
from the National Institutes of Health (R35GM137987), the 46 (6), 3183−3191.
National Science Foundation (BMAT, 1944875), a 3M Non- (19) Bornstein, G. G. (2015) Antibody Drug Conjugates: Preclinical
Tenured Faculty Award (3M Company), and the University of Considerations. AAPS J. 17 (3), 525−534.
Notre Dame “Advancing our Vision” initiative. All schematics (20) Dai, Q., Wilhelm, S., Ding, D., Syed, A. M., Sindhwani, S.,
and graphics were created using BioRender.com and used with Zhang, Y., Chen, Y. Y., MacMillan, P., and Chan, W. C. W. (2018)
permission. Quantifying the Ligand-Coated Nanoparticle Delivery to Cancer Cells

■ REFERENCES
(1) Langer, R. (1998) Drug Delivery and Targeting. Nature 392
in Solid Tumors. ACS Nano 12 (8), 8423−8435.
(21) Devaraj, N. K. (2018) The Future of Bioorthogonal Chemistry.
ACS Cent. Sci. 4, 952−959.
(22) Chang, P. V., Prescher, J. A., Sletten, E. M., Baskin, J. M., Miller,
(6679 Suppl), 5−10.
(2) Tonga, G. Y., Jeong, Y., Duncan, B., Mizuhara, T., Mout, R., Das, I. A., Agard, N. J., Lo, A., and Bertozzi, C. R. (2010) Copper-Free
R., Kim, S. T., Yeh, Y.-C., Yan, B., Hou, S., et al. (2015) Click Chemistry in Living Animals. Proc. Natl. Acad. Sci. U. S. A. 107
Supramolecular Regulation of Bioorthogonal Catalysis in Cells (5), 1821−1826.
Using Nanoparticle-Embedded Transition Metal Catalysts. Nat. (23) Prescher, J. A., Dube, D. H., and Bertozzi, C. R. (2004)
Chem. 7 (7), 597−603. Chemical Remodelling of Cell Surfaces in Living Animals. Nature 430
(3) Rautio, J., Kumpulainen, H., Heimbach, T., Oliyai, R., Oh, D., (7002), 873−877.
Järvinen, T., and Savolainen, J. (2008) Prodrugs: Design and Clinical (24) Baskin, J. M., Prescher, J. A., Laughlin, S. T., Agard, N. J.,
Applications. Nat. Rev. Drug Discovery 7, 255−270. Chang, P. V., Miller, I. A., Lo, A., Codelli, J. A., and Bertozzi, C. R.
(4) Pattni, B. S., Chupin, V. V., and Torchilin, V. P. (2015) New (2007) Copper-Free Click Chemistry for Dynamic in Vivo Imaging.
Developments in Liposomal Drug Delivery. Chem. Rev. 115, 10938− Proc. Natl. Acad. Sci. U. S. A. 104 (43), 16793−16797.
10966. (25) Brudno, Y., Pezone, M. J., Snyder, T. K., Uzun, O., Moody, C.
(5) Izci, M., Maksoudian, C., Manshian, B. B., and Soenen, S. J. T., Aizenberg, M., and Mooney, D. J. (2018) Replenishable Drug
(2021) The Use of Alternative Strategies for Enhanced Nanoparticle Depot to Combat Post-Resection Cancer Recurrence. Biomaterials
Delivery to Solid Tumors. Chem. Rev. 121 (3), 1746−1803. 178, 373−382.
(6) Golombek, S. K., May, J.-N., Theek, B., Appold, L., Drude, N., (26) Evans, H. L., Slade, R. L., Carroll, L., Smith, G., Nguyen, Q.-D.,
Kiessling, F., and Lammers, T. (2018) Tumor Targeting via EPR: Iddon, L., Kamaly, N., Stöckmann, H., Leeper, F. J., Aboagye, E. O.,
Strategies to Enhance Patient Responses. Adv. Drug Delivery Rev. 130, et al. (2012) Copper-Free Clicka Promising Tool for Pre-Targeted
17−38. PET Imaging. Chem. Commun. 48, 991−993.
(7) Barenholz, Y. C. (2012) Doxil  The First FDA-Approved (27) Wang, H., Sobral, M. C., Snyder, T., Brudno, Y., Gorantla, V. S.,
Nano-Drug: Lessons Learned. J. Controlled Release 160 (2), 117−134. and Mooney, D. J. (2020) Clickable, Acid Labile Immunosuppressive
(8) Alley, S. C., Okeley, N. M., and Senter, P. D. (2010) Antibody- Prodrugs for in Vivo Targeting. Biomater. Sci. 8 (1), 266−277.
Drug Conjugates: Targeted Drug Delivery for Cancer. Curr. Opin. (28) Palvai, S., Bhangu, J., Akgun, B., Moody, C. T., Hall, D. G., and
Chem. Biol. 14 (4), 529−537. Brudno, Y. (2020) In Vivo Targeting Using Arylboronate/Nopoldiol
(9) Emerich, D. F., and Thanos, C. G. (2007) Targeted Click Conjugation. Bioconjugate Chem. 31, 2288−2292.
Nanoparticle-Based Drug Delivery and Diagnosis. J. Drug Target. 15 (29) Rossin, R., Verkerk, P. R., van den Bosch, S. M., Vulders, R. C.
(3), 163−183. M., Verel, I., Lub, J., and Robillard, M. S. (2010) In Vivo Chemistry
(10) Omstead, D. T., Sjoerdsma, J., and Bilgicer, B. (2019) for Pretargeted Tumor Imaging in Live Mice. Angew. Chem., Int. Ed.
Polyvalent Nanoobjects for Precision Diagnostics. Annu. Rev. Anal. 49 (19), 3375−3378.
Chem. 12 (1), 69−88. (30) Versteegen, R. M., Rossin, R., Ten Hoeve, W., Janssen, H. M.,
(11) Drago, J. Z., Modi, S., and Chandarlapaty, S. (2021) Unlocking and Robillard, M. S. (2013) Click to Release: Instantaneous
the Potential of Antibody−drug Conjugates for Cancer Therapy. Nat.
Doxorubicin Elimination upon Tetrazine Ligation. Angew. Chem.,
Rev. Clin. Oncol. 18, 327−344.
Int. Ed. 52 (52), 14112−14116.
(12) Beck, A., Goetsch, L., Dumontet, C., and Corvaïa, N. (2017)
(31) Mejia Oneto, J. M., Khan, I., Seebald, L., and Royzen, M.
Strategies and Challenges for the next Generation of Antibody-Drug
(2016) In Vivo Bioorthogonal Chemistry Enables Local Hydrogel and
Conjugates. Nat. Rev. Drug Discovery 16 (5), 315−337.
(13) Lv, S., Sylvestre, M., Prossnitz, A. N., Yang, L. F., and Pun, S. H. Systemic Pro-Drug To Treat Soft Tissue Sarcoma. ACS Cent. Sci. 2
(2021) Design of Polymeric Carriers for Intracellular Peptide Delivery (7), 476−482.
in Oncology Applications. Chem. Rev., 1 DOI: 10.1021/acs.chem- (32) Liu, Z., Nalluri, S. K. M., and Stoddart, J. F. (2017) Surveying
rev.0c00963. Macrocyclic Chemistry: From Flexible Crown Ethers to Rigid
(14) Kunjachan, S., Pola, R., Gremse, F., Theek, B., Ehling, J., Cyclophanes. Chem. Soc. Rev. 46 (9), 2459−2478.
Moeckel, D., Hermanns-Sachweh, B., Pechar, M., Ulbrich, K., (33) Khvorova, A., and Watts, J. K. (2017) The Chemical Evolution
Hennink, W. E., et al. (2014) Passive versus Active Tumor Targeting of Oligonucleotide Therapies of Clinical Utility. Nat. Biotechnol. 35
Using RGD- and NGR-Modified Polymeric Nanomedicines. Nano (3), 238−248.
Lett. 14 (2), 972−981. (34) Webber, M. J., and Langer, R. (2017) Drug Delivery by
(15) Omstead, D. T., Mejia, F., Sjoerdsma, J., Kim, B., Shin, J., Khan, Supramolecular Design. Chem. Soc. Rev. 46 (21), 6600−6620.
S., Wu, J., Kiziltepe, T., Littlepage, L. E., and Bilgicer, B. (2020) In (35) Schreiber, C. L., and Smith, B. D. (2019) Molecular
Vivo Evaluation of CD38 and CD138 as Targets for Nanoparticle- Conjugation Using Non-Covalent Click Chemistry. Nat. Rev. Chem.
Based Drug Delivery in Multiple Myeloma. J. Hematol. Oncol. 13 (1), 3 (6), 393−400.
145. (36) Webber, M. J., Appel, E. A., Meijer, E. W., and Langer, R.
(16) Wang, W., Singh, S., Zeng, D. L., King, K., and Nema, S. (2007) (2016) Supramolecular Biomaterials. Nat. Mater. 15 (1), 13−26.
Antibody Structure, Instability, and Formulation. J. Pharm. Sci. 96 (1), (37) Braegelman, A. S., and Webber, M. J. (2019) Integrating
1−26. Stimuli-Responsive Properties in Host-Guest Supramolecular Drug
(17) Coats, S., Williams, M., Kebble, B., Dixit, R., Tseng, L., Yao, N.- Delivery Systems. Theranostics 9 (11), 3017−3040.
S., Tice, D. A., and Soria, J.-C. (2019) Antibody-Drug Conjugates: (38) Tang, H., Fuentealba, D., Ko, Y. H., Selvapalam, N., Kim, K.,
Future Directions in Clinical and Translational Strategies to Improve and Bohne, C. (2011) Guest Binding Dynamics with cucurbit[7]uril
the Therapeutic Index. Clin. Cancer Res. 25 (18), 5441−5448. in the Presence of Cations. J. Am. Chem. Soc. 133 (50), 20623−20633.

1943 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

(39) Ning, X., Guo, J., Wolfert, M. A., and Boons, G.-J. (2008) Attomolar Dissociation Constant. Angew. Chem., Int. Ed. 53 (4), 988−
Visualizing Metabolically Labeled Glycoconjugates of Living Cells by 993.
Copper-Free and Fast HuisgenCycloadditions. Angew. Chem., Int. Ed. (60) Rekharsky, M. V., Mori, T., Yang, C., Ko, Y. H., Selvapalam, N.,
47 (12), 2253−2255. Kim, H., Sobransingh, D., Kaifer, A. E., Liu, S., Isaacs, L., et al. (2007)
(40) Taylor, M. T., Blackman, M. L., Dmitrenko, O., and Fox, J. M. A Synthetic Host-Guest System Achieves Avidin-Biotin Affinity by
(2011) Design and Synthesis of Highly Reactive Dienophiles for the Overcoming Enthalpy-Entropy Compensation. Proc. Natl. Acad. Sci.
Tetrazine-Trans-Cyclooctene Ligation. J. Am. Chem. Soc. 133 (25), U. S. A. 104 (52), 20737−20742.
9646−9649. (61) Sigwalt, D., Š ekutor, M., Cao, L., Zavalij, P. Y., Hostaš, J., Ajani,
(41) Maikawa, C. L., d’Aquino, A. I., Vuong, E. T., Su, B., Zou, L., H., Hobza, P., Mlinarić-Majerski, K., Glaser, R., and Isaacs, L. (2017)
Chen, P. C., Nguyen, L. T., Autzen, A. A. A., Mann, J. L., Webber, M. Unraveling the Structure-Affinity Relationship between Cucurbit[n]-
J., and Appel, E. A. (2021) Affinity-Directed Dynamics of urils (n = 7, 8) and Cationic Diamondoids. J. Am. Chem. Soc. 139 (8),
Host−Guest Motifs for Pharmacokinetic Modulation via Supra- 3249−3258.
molecular PEGylation. Biomacromolecules 22 (8), 3565−3573. (62) Herce, H. D., Schumacher, D., Schneider, A. F. L., Ludwig, A.
(42) Sela-Culang, I., Kunik, V., and Ofran, Y. (2013) The Structural K., Mann, F. A., Fillies, M., Kasper, M.-A., Reinke, S., Krause, E.,
Basis of Antibody-Antigen Recognition. Front. Immunol. 4, 302. Leonhardt, H., et al. (2017) Cell-Permeable Nanobodies for Targeted
(43) Erlendsson, S., and Teilum, K. (2021) Binding Revisited- Immunolabelling and Antigen Manipulation in Living Cells. Nat.
Avidity in Cellular Function and Signaling. Front MolBiosci 7, 615565. Chem. 9 (8), 762−771.
(44) Vauquelin, G., and Charlton, S. J. (2013) Exploring Avidity: (63) Marquez, B. V., and Lapi, S. E. (2016) PretargetedImmuno-
Understanding the Potential Gains in Functional Affinity and Target PET: Overcoming Limitations of Space and Time. J. Nucl. Med. 57
Residence Time of Bivalent and Heterobivalent Ligands. Br. J. (3), 332−333.
Pharmacol. 168 (8), 1771−1785. (64) Hnatowich, D. J., Virzi, F., and Rusckowski, M. (1987)
(45) Liu, W., Samanta, S. K., Smith, B. D., and Isaacs, L. (2017) Investigations of Avidin and Biotin for Imaging Applications. J. Nucl.
Synthetic Mimics of Biotin/(strept)avidin. Chem. Soc. Rev. 46 (9), Med. 28 (8), 1294−1302.
2391−2403. (65) Beck, A., Wurch, T., Bailly, C., and Corvaia, N. (2010)
(46) Zou, L., VandenBerg, M. A., and Webber, M. J. (2020) Single- Strategies and Challenges for the next Generation of Therapeutic
Molecule Nanoscale Drug Carriers with Quantitative Supramolecular Antibodies. Nat. Rev. Immunol. 10 (5), 345−352.
Loading. Molecular Systems Design & Engineering 5, 197−204. (66) Rothbauer, U., Zolghadr, K., Tillib, S., Nowak, D., Schermelleh,
(47) Liu, W., Johnson, A., and Smith, B. D. (2018) Guest Back- L., Gahl, A., Backmann, N., Conrath, K., Muyldermans, S., Cardoso,
Folding: A Molecular Design Strategy That Produces a Deep-Red M. C., et al. (2006) Targeting and Tracing Antigens in Live Cells with
Fluorescent Host/Guest Pair with Picomolar Affinity in Water. J. Am. Fluorescent Nanobodies. Nat. Methods 3 (11), 887−889.
Chem. Soc. 140 (9), 3361−3370. (67) Moghaddam, S., Yang, C., Rekharsky, M., Ko, Y. H., Kim, K.,
(48) Liu, W., McGarraugh, H., and Smith, B. (2018) Fluorescent Inoue, Y., and Gilson, M. K. (2011) New Ultrahigh Affinity Host-
Thienothiophene-Containing Squaraine Dyes and Threaded Supra- Guest Complexes of cucurbit[7]uril with bicyclo[2.2.2]octane and
molecular Complexes with Tunable Wavelengths between 600−800 Adamantane Guests: Thermodynamic Analysis and Evaluation of M2
Nm. Molecules 23, 2229. Affinity Calculations. J. Am. Chem. Soc. 133 (10), 3570−3581.
(49) Crini, G. (2014) Review: A History of Cyclodextrins. Chem. (68) Jeon, W. S., Moon, K., Park, S. H., Chun, H., Ko, Y. H., Lee, J.
Rev. 114, 10940−10975. Y., Lee, E. S., Samal, S., Selvapalam, N., Rekharsky, M. V., et al.
(50) Jansook, P., Ogawa, N., and Loftsson, T. (2018) Cyclodextrins: (2005) Complexation of Ferrocene Derivatives by the cucurbit[7]uril
Structure, Physicochemical Properties and Pharmaceutical Applica- Host: A Comparative Study of the Cucurbituril and Cyclodextrin
tions. Int. J. Pharm. 535 (1−2), 272−284. Host Families. J. Am. Chem. Soc. 127 (37), 12984−12989.
(51) Hu, Q.-D., Tang, G.-P., and Chu, P. K. (2014) Cyclodextrin- (69) Zou, L., Braegelman, A. S., and Webber, M. J. (2019) Dynamic
Based Host-Guest Supramolecular Nanoparticles for Delivery: From Supramolecular Hydrogels Spanning an Unprecedented Range of
Design to Applications. Acc. Chem. Res. 47 (7), 2017−2025. Host-Guest Affinity. ACS Appl. Mater. Interfaces 11 (6), 5695−5700.
(52) Eftink, M. R., Andy, M. L., Bystrom, K., Perlmutter, H. D., and (70) Strebl, M. G., Yang, J., Isaacs, L., and Hooker, J. M. (2018)
Kristol, D. S. (1989) Cyclodextrin Inclusion Complexes: Studies of Adamantane/Cucurbituril: A Potential Pretargeted Imaging Strategy
the Variation in the Size of Alicyclic Guests. J. Am. Chem. Soc. 111, in Immuno-PET. Mol. Imaging 17, 153601211879983.
6765−6772. (71) Li, M., Kim, S., Lee, A., Shrinidhi, A., Ko, Y. H., Lim, H. G.,
(53) Chen, G., and Jiang, M. (2011) Cyclodextrin-Based Inclusion Kim, H. H., Bae, K. B., Park, K. M., and Kim, K. (2019) Bio-
Complexation Bridging Supramolecular Chemistry and Macro- Orthogonal Supramolecular Latching inside Live Animals and Its
molecular Self-Assembly. Chem. Soc. Rev. 40, 2254. Application for in Vivo Cancer Imaging. ACS Appl. Mater. Interfaces
(54) Lee, J. W., Samal, S., Selvapalam, N., Kim, H.-J., and Kim, K. 11 (47), 43920−43927.
(2003) Cucurbituril Homologues and Derivatives: New Opportu- (72) Zou, L., Braegelman, A. S., and Webber, M. J. (2019) Spatially
nities in Supramolecular Chemistry. Acc. Chem. Res. 36 (8), 621−630. Defined Drug Targeting by in Situ Host−Guest Chemistry in a Living
(55) Kim, J., Jung, I.-S., Kim, S.-Y., Lee, E., Kang, J.-K., Sakamoto, S., Animal. ACS Cent. Sci. 5, 1035−1043.
Yamaguchi, K., and Kim, K. (2000) New Cucurbituril Homologues: (73) Spa, S. J., Welling, M. M., van Oosterom, M. N., Rietbergen, D.
Syntheses, Isolation, Characterization, and X-Ray Crystal Structures D. D., Burgmans, M. C., Verboom, W., Huskens, J., Buckle, T., and
of Cucurbit[n]uril (n = 5, 7, and 8). J. Am. Chem. Soc. 122, 540−541. van Leeuwen, F. W. B. (2018) A Supramolecular Approach for Liver
(56) Lagona, J., Mukhopadhyay, P., Chakrabarti, S., and Isaacs, L. Radioembolization. Theranostics 8 (9), 2377−2386.
(2005) The Cucurbit[n]uril Family. Angew. Chem., Int. Ed. 44 (31), (74) Welling, M. M., Spa, S. J., van Willigen, D. M., Rietbergen, D.
4844−4870. D. D., Roestenberg, M., Buckle, T., and van Leeuwen, F. W. B. (2019)
(57) Barrow, S. J., Kasera, S., Rowland, M. J., del Barrio, J., and In Vivo Stability of Supramolecular Host−guest Complexes
Scherman, O. A. (2015) Cucurbituril-Based Molecular Recognition. Monitored by Dual-Isotope Multiplexing in a Pre-Targeting Model
Chem. Rev. 115 (22), 12320−12406. of Experimental Liver Radioembolization. J. Controlled Release 293,
(58) Shetty, D., Khedkar, J. K., Park, K. M., and Kim, K. (2015) Can 126−134.
We Beat the Biotin-Avidin Pair?: cucurbit[7]uril-Based Ultrahigh (75) Mehta, M., Deeksha Tewari, D., Gupta, G., Awasthi, R., Singh,
Affinity Host-Guest Complexes and Their Applications. Chem. Soc. H., Pandey, P., Chellappan, D. K., Wadhwa, R., Collet, T., et al.
Rev. 44 (23), 8747−8761. (2019) Oligonucleotide Therapy: An Emerging Focus Area for Drug
(59) Cao, L., Śekutor, M., Zavalij, P. Y., Mlinarić-Majerski, K., Delivery in Chronic Inflammatory Respiratory Diseases. Chem.-Biol.
Glaser, R., and Isaacs, L. (2014) Cucurbit[7]uril·guest Pair with an Interact. 308, 206−215.

1944 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

(76) Crooke, S. T. (1992) Therapeutic Applications of Oligonucleo- (98) Zhou, J., Soontornworajit, B., Snipes, M. P., and Wang, Y.
tides. Nat. Biotechnol. 10 (8), 882−886. (2009) Development of a Novel Pretargeting System with Bifunc-
(77) Juliano, R. L. (2016) The Delivery of Therapeutic tional Nucleic Acid Molecules. Biochem. Biophys. Res. Commun. 386
Oligonucleotides. Nucleic Acids Res. 44, 6518−6548. (3), 521−525.
(78) Reynolds, A., Leake, D., Boese, Q., Scaringe, S., Marshall, W. S., (99) Shi, P., Zhao, N., Lai, J., Coyne, J., Gaddes, E. R., and Wang, Y.
and Khvorova, A. (2004) Rational siRNA Design for RNA (2018) Polyvalent Display of Biomolecules on Live Cells. Angew.
Interference. Nat. Biotechnol. 22, 326−330. Chem., Int. Ed. 57 (23), 6800−6804.
(79) Whitehead, K. A., Langer, R., and Anderson, D. G. (2009) (100) Schubert, M., Bergmann, R., Förster, C., Sihver, W., Vonhoff,
Knocking down Barriers: Advances in siRNA Delivery. Nat. Rev. Drug S., Klussmann, S., Bethge, L., Walther, M., Schlesinger, J., Pietzsch, J.,
Discovery 8 (2), 129−138. et al. (2017) Novel Tumor Pretargeting System Based on
(80) Carlon, E., and Heim, T. (2006) Thermodynamics of RNA/ Complementary L-Configured Oligonucleotides. Bioconjugate Chem.
DNA Hybridization in High-Density Oligonucleotide Microarrays. 28 (4), 1176−1188.
Phys. A 362, 433−449. (101) Nielsen, P. E., Egholm, M., Berg, R. H., and Buchardt, O.
(81) SantaLucia, J., and Hicks, D. (2004) The Thermodynamics of (1991) Sequence-Selective Recognition of DNA by Strand Displace-
DNA Structural Motifs. Annu. Rev. Biophys. Biomol. Struct. 33, 415− ment with a Thymine-Substituted Polyamide. Science 254 (5037),
440. 1497−1500.
(82) SantaLucia, J. (1998) A Unified View of Polymer, Dumbbell, (102) Goldman, J. M., Zhang, L. A., Manna, A., Armitage, B. A., Ly,
and Oligonucleotide DNA Nearest-Neighbor Thermodynamics. Proc. D. H., and Schneider, J. W. (2013) High Affinity γPNA Sandwich
Natl. Acad. Sci. U. S. A. 95, 1460−1465. Hybridization Assay for Rapid Detection of Short Nucleic Acid
(83) Elder, R. M., Pfaendtner, J., and Jayaraman, A. (2015) Effect of Targets with Single Mismatch Discrimination. Biomacromolecules 14
Hydrophobic and Hydrophilic Surfaces on the Stability of Double- (7), 2253−2261.
Stranded DNA. Biomacromolecules 16, 1862−1869. (103) Gupta, A., Mishra, A., and Puri, N. (2017) Peptide Nucleic
(84) Owczarzy, R., Dunietz, I., Behlke, M. A., Klotz, I. M., and Acids: Advanced Tools for Biomedical Applications. J. Biotechnol. 259,
Walder, J. A. (2003) Thermodynamic Treatment of Oligonucleotide 148−159.
Duplex-Simplex Equilibria. Proc. Natl. Acad. Sci. U. S. A. 100, 14840− (104) Nielsen, P. E. (1999) Peptide Nucleic Acid. A Molecule with
14845. Two Identities. Acc. Chem. Res. 32, 624−630.
(85) Bielec, K., Sozanski, K., Seynen, M., Dziekan, Z., Ten Wolde, P. (105) Rusckowski, M., Qu, T., Chang, F., and Hnatowich, D. J.
R., and Holyst, R. (2019) Kinetics and Equilibrium Constants of (1997) Pretargeting Using Peptide Nucleic Acid. Cancer 80 (S12),
Oligonucleotides at Low Concentrations. Hybridization and Melting 2699−2705.
Study. Phys. Chem. Chem. Phys. 21 (20), 10798−10807. (106) Brudno, Y., Silva, E. A., Kearney, C. J., Lewin, S. A., Miller, A.,
(86) Keefe, A. D., Pai, S., and Ellington, A. (2010) Aptamers as Martinick, K. D., Aizenberg, M., and Mooney, D. J. (2014) Refilling
Therapeutics. Nat. Rev. Drug Discovery 9 (7), 537−550. Drug Delivery Depots through the Blood. Proc. Natl. Acad. Sci. U. S. A.
(87) Hermann, T., and Patel, D. J. (2000) Adaptive Recognition by 111 (35), 12722−12727.
Nucleic Acid Aptamers. Science 287 (5454), 820−825. (107) Chu, T.-W., Yang, J., Zhang, R., Sima, M., and Kopeček, J.
(88) Ni, S., Zhuo, Z., Pan, Y., Yu, Y., Li, F., Liu, J., Wang, L., Wu, X., (2014) Cell Surface Self-Assembly of Hybrid Nanoconjugates via
Li, D., Wan, Y., et al. (2021) Recent Progress in Aptamer Discoveries Oligonucleotide Hybridization Induces Apoptosis. ACS Nano 8 (1),
and Modifications for Therapeutic Applications. ACS Appl. Mater. 719−730.
Interfaces 13 (8), 9500−9519. (108) Li, L., Wang, J., Li, Y., Radford, D. C., Yang, J., and Kopeček, J.
(89) Roberts, T. C., Langer, R., and Wood, M. J. A. (2020) Advances (2019) Broadening and Enhancing Functions of Antibodies by Self-
in Oligonucleotide Drug Delivery. Nat. Rev. Drug Discovery 19, 673− Assembling Multimerization at Cell Surface. ACS Nano 13 (10),
694. 11422−11432.
(90) Urban, E., and Noe, C. R. (2003) Structural Modifications of (109) Yang, J., Li, L., and Kopeček, J. (2019) Biorecognition: A Key
Antisense Oligonucleotides. Farmaco 58, 243−258. to Drug-Free Macromolecular Therapeutics. Biomaterials 190−191,
(91) Glazier, D. A., Liao, J., Roberts, B. L., Li, X., Yang, K., Stevens, 11−23.
C. M., and Tang, W. (2020) Chemical Synthesis and Biological (110) Summerton, J., and Weller, D. (1997) Morpholino Antisense
Application of Modified Oligonucleotides. Bioconjugate Chem. 31 (5), Oligomers: Design, Preparation, and Properties. Antisense Nucleic Acid
1213−1233. Drug Dev. 7, 187−195.
(92) Hsu, N.-S., Lee, C.-C., Kuo, W.-C., Chang, Y.-W., Lo, S.-Y., and (111) Murakami, Y., and Maeda, M. (2005) DNA-Responsive
Wang, A. H.-J. (2020) Development of a Versatile and Modular Hydrogels That Can Shrink or Swell. Biomacromolecules 6 (6), 2927−
Linker for Antibody-Drug Conjugates Based on Oligonucleotide 2929.
Strand Pairing. Bioconjugate Chem. 31 (7), 1804−1811. (112) Battig, M. R., Soontornworajit, B., and Wang, Y. (2012)
(93) Tan, W., Wang, K., and Drake, T. J. (2004) Molecular Beacons. Programmable Release of Multiple Protein Drugs from Aptamer-
Curr. Opin. Chem. Biol. 8 (5), 547−553. Functionalized Hydrogels via Nucleic Acid Hybridization. J. Am.
(94) Tyagi, S., and Kramer, F. R. (1996) Molecular Beacons: Probes Chem. Soc. 134 (30), 12410−12413.
That Fluoresce upon Hybridization. Nat. Biotechnol. 14 (3), 303−308. (113) McNamara, S. L., Brudno, Y., Miller, A. B., Ham, H. O.,
(95) Shi, H., He, X., Wang, K., Wu, X., Ye, X., Guo, Q., Tan, W., Aizenberg, M., Chaikof, E. L., and Mooney, D. J. (2020) Regenerating
Qing, Z., Yang, X., and Zhou, B. (2011) Activatable Aptamer Probe Antithrombotic Surfaces through Nucleic Acid Displacement. ACS
for Contrast-Enhanced in Vivo Cancer Imaging Based on Cell Biomater. Sci. Eng. 6 (4), 2159−2166.
Membrane Protein-Triggered Conformation Alteration. Proc. Natl. (114) Zhang, D. Y., and Winfree, E. (2009) Control of DNA Strand
Acad. Sci. U. S. A. 108 (10), 3900−3905. Displacement Kinetics Using Toehold Exchange. J. Am. Chem. Soc.
(96) Yan, L.’a., Shi, H., He, X., Wang, K., Tang, J., Chen, M., Ye, X., 131, 17303−17314.
Xu, F., and Lei, Y. (2014) A Versatile Activatable Fluorescence (115) Utterström, J., Naeimipour, S., Selegård, R., and Aili, D.
Probing Platform for Cancer Cells in Vitro and in Vivo Based on Self- (2021) Coiled Coil-Based Therapeutics and Drug Delivery Systems.
Assembled Aptamer/carbon Nanotube Ensembles. Anal. Chem. 86 Adv. Drug Delivery Rev. 170, 26−43.
(18), 9271−9277. (116) Yu, Y. B. (2002) Coiled-Coils: Stability, Specificity, and Drug
(97) Zhu, G., Zheng, J., Song, E., Donovan, M., Zhang, K., Liu, C., Delivery Potential. Adv. Drug Delivery Rev. 54 (8), 1113−1129.
and Tan, W. (2013) Self-Assembled, Aptamer-Tethered DNA (117) Burkhard, P., Stetefeld, J., and Strelkov, S. V. (2001) Coiled
Nanotrains for Targeted Transport of Molecular Drugs in Cancer Coils: A Highly Versatile Protein Folding Motif. Trends Cell Biol. 11
Theranostics. Proc. Natl. Acad. Sci. U. S. A. 110 (20), 7998−8003. (2), 82−88.

1945 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946
Bioconjugate Chemistry pubs.acs.org/bc Review

(118) Harbury, P. B., Zhang, T., Kim, P. S., and Alber, T. (1993) A Peptides in Liposomal Anticancer Drug Delivery Using a Zebrafish
Switch between Two-, Three-, and Four-Stranded Coiled Coils in Xenograft Model. ACS Nano 10 (8), 7428−7435.
GCN4 Leucine Zipper Mutants. Science 262 (5138), 1401−1407.
(119) Slovic, A. M., Lear, J. D., and DeGrado, W. F. (2005) De
Novo Design of a Pentameric Coiled-Coil: Decoding the Motif for
Tetramer versus Pentamer Formation in Water-Soluble Phospholam-
ban. J. Pept. Res. 65 (3), 312−321.
(120) Zaccai, N. R., Chi, B., Thomson, A. R., Boyle, A. L., Bartlett,
G. J., Bruning, M., Linden, N., Sessions, R. B., Booth, P. J., Leo Brady,
R., et al. (2011) A de Novo Peptide Hexamer with a Mutable
Channel. Nat. Chem. Biol. 7, 935−941.
(121) Moll, J. R., Ruvinov, S. B., Pastan, I., and Vinson, C. (2001)
Designed Heterodimerizing Leucine Zippers with a Ranger of pIs and
Stabilities up to 10(−15) M. Protein Sci. 10 (3), 649−655.
(122) Reinke, A. W., Grant, R. A., and Keating, A. E. (2010) A
Synthetic Coiled-Coil Interactome Provides Heterospecific Modules
for Molecular Engineering. J. Am. Chem. Soc. 132 (17), 6025−6031.
(123) Woolfson, D. N. (2005) The Design of Coiled-Coil Structures
and Assemblies. Adv. Protein Chem. 70, 79−112.
(124) Fletcher, J. M., Boyle, A. L., Bruning, M., Bartlett, G. J.,
Vincent, T. L., Zaccai, N. R., Armstrong, C. T., Bromley, E. H. C.,
Booth, P. J., Brady, R. L., et al. (2012) A Basis Set of de Novo Coiled-
Coil Peptide Oligomers for Rational Protein Design and Synthetic
Biology. ACS Synth. Biol. 1 (6), 240−250.
(125) Fletcher, J. M., Harniman, R. L., Barnes, F. R. H., Boyle, A. L.,
Collins, A., Mantell, J., Sharp, T. H., Antognozzi, M., Booth, P. J.,
Linden, N., et al. (2013) Self-Assembling Cages from Coiled-Coil
Peptide Modules. Science 340 (6132), 595−599.
(126) Gradišar, H., Božič, S., Doles, T., Vengust, D., Hafner-
Bratkovič, I., Mertelj, A., Webb, B., Š ali, A., Klavžar, S., and Jerala, R.
(2013) Design of a Single-Chain Polypeptide Tetrahedron Assembled
from Coiled-Coil Segments. Nat. Chem. Biol. 9 (6), 362−366.
(127) Pola, R., Laga, R., Ulbrich, K., Sieglová, I., Král, V., Fábry, M.,
Kabešová, M., Kovář, M., and Pechar, M. (2013) Polymer
Therapeutics with a Coiled Coil Motif Targeted against Murine
bcl1 Leukemia. Biomacromolecules 14 (3), 881−889.
(128) Pechar, M., Pola, R., Laga, R., Ulbrich, K., Bednárová, L.,
Maloň, P., Sieglová, I., Král, V., Fábry, M., and Vaněk, O. (2011)
Coiled Coil Peptides as Universal Linkers for the Attachment of
Recombinant Proteins to Polymer Therapeutics. Biomacromolecules 12
(10), 3645−3655.
(129) Park, W. M., and Champion, J. A. (2014) Thermally Triggered
Self-Assembly of Folded Proteins into Vesicles. J. Am. Chem. Soc. 136
(52), 17906−17909.
(130) Dhankher, A., Hernandez, M. E., Howard, H. C., and
Champion, J. A. (2020) Characterization and Control of Dynamic
Rearrangement in a Self-Assembled Antibody Carrier. Biomacromo-
lecules 21, 1407−1416.
(131) Wu, K., Liu, J., Johnson, R. N., Yang, J., and Kopecek, J.
(2010) Drug-Free Macromolecular Therapeutics: Induction of
Apoptosis by Coiled-Coil-Mediated Cross-Linking of Antigens on
the Cell Surface. Angew. Chem., Int. Ed. 49 (8), 1451−1455.
(132) Wu, K., Yang, J., Liu, J., and Kopeček, J. (2012) Coiled-Coil
Based Drug-Free Macromolecular Therapeutics: In Vivo Efficacy. J.
Controlled Release 157 (1), 126−131.
(133) Kverka, M., Hartley, J. M., Chu, T.-W., Yang, J., Heidchen, R.,
and Kopeček, J. (2014) Immunogenicity of Coiled-Coil Based Drug-
Free Macromolecular Therapeutics. Biomaterials 35 (22), 5886−5896.
(134) Zhang, R., Yang, J., Chu, T.-W., Hartley, J. M., and Kopeček, J.
(2015) Multimodality Imaging of Coiled-Coil Mediated Self-
Assembly in a “Drug-Free” Therapeutic System. Adv. Healthcare
Mater. 4, 1054−1065.
(135) Zhang, L., Fang, Y., Li, L., Yang, J., Radford, D. C., and
Kopeček, J. (2018) Human Serum Albumin-Based Drug-Free
Macromolecular Therapeutics: Apoptosis Induction by Coiled-Coil-
Mediated Cross-Linking of CD20 Antigens on Lymphoma B Cell
Surface. Macromol. Biosci. 18 (11), No. e1800224.
(136) Yang, J., Shimada, Y., Olsthoorn, R. C. L., Snaar-Jagalska, B.
E., Spaink, H. P., and Kros, A. (2016) Application of Coiled Coil

1946 https://doi.org/10.1021/acs.bioconjchem.1c00326
Bioconjugate Chem. 2021, 32, 1935−1946

You might also like