0% found this document useful (0 votes)
46 views36 pages

The Impact of Nanobodies On GPCR Structural Biology

The family of human G protein-coupled receptors (GPCRs) is comprised of about 800 different members, with about 35% of current pharmaceutical drugs targeting GPCRs. However, GPCR structural biology, necessary for structure-guided drug design, has lagged behind that of other membrane proteins, and it was not until the year 2000 when the first crystal structure of a GPCR (rhodopsin) was solved.

Uploaded by

jose
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
46 views36 pages

The Impact of Nanobodies On GPCR Structural Biology

The family of human G protein-coupled receptors (GPCRs) is comprised of about 800 different members, with about 35% of current pharmaceutical drugs targeting GPCRs. However, GPCR structural biology, necessary for structure-guided drug design, has lagged behind that of other membrane proteins, and it was not until the year 2000 when the first crystal structure of a GPCR (rhodopsin) was solved.

Uploaded by

jose
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 36

Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.

000974
This article has not been copyedited and formatted. The final version may differ from this version.

The impact of nanobodies on GPCR structural biology and their potential as

therapeutic agents

David Salom, Arum Wu, Chang C. Liu, and Krzysztof Palczewski

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


Primary affiliations:

Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of

Ophthalmology (D.S., A.W., K.P), and Department of Biomedical Engineering (C.C.L),

University of California, Irvine, CA 92697, USA

1
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Running title: Nanobodies and GPCRs

Corresponding authors:

1- Krzysztof Palczewski, 837 Health Sciences Road, Gillespie Neuroscience Building,


Room 2105, Irvine, CA 92697-4375; phone 949-824-6527, email: [email protected]
2- David Salom, 837 Health Sciences Rd, Gillespie Neuroscience Building, Room 2216,
Irvine, CA 92617; phone 949-824-5154; email: [email protected]
3- Arum Wu, 837 Health Sciences Rd, Gillespie Neuroscience Building, Room 2216,
Irvine, CA 92617; phone 949-824-5154; email: [email protected]
4- Chang C. Liu, ISEB 4064, 419 South Circle View Dr., Irvine, CA 92697; Phone: 949-
824-8534, email: [email protected]

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


Manuscript pages: 32
Number of tables: 1
Number of figures: 4
Number of references: 77
Abstract word count: 194
Introduction word count: 255
Discussion word count: no discussion

Abbreviations:
ACKR3: atypical chemokine receptor 3
AR: adrenergic receptor
AT1R: Angiotensin II type I receptor
BRIL: thermostabilized apocytochrome b562
CDR: complementarity-determining region
Cryo-EM: cryogenic electron microscopy
ECD: extracellular domain
GPCR: G protein-coupled receptor
GPCRDB: GPCR database
ICD: intracellular domain
ICL3: intracellular loop 3
Nb: nanobody
OR: opioid receptor
scFv: single-chain variable fragment
T4L: T4 lysozyme
Trx: thioredoxin

Keywords: Nanobodies, GPCRs, single domain antibodies, cryo-EM, crystallography,


therapeutic antibodies, structural biology, ligand, VHHs

2
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Abstract

The family of human G protein-coupled receptors (GPCRs) is comprised of about 800

different members, with about 35% of current pharmaceutical drugs targeting GPCRs.

However, GPCR structural biology, necessary for structure-guided drug design, has

lagged behind that of other membrane proteins, and it was not until the year 2000 when

the first crystal structure of a GPCR (rhodopsin) was solved. Starting in 2007, the

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


determination of additional GPCR structures was facilitated by protein engineering, new

crystallization techniques, complexation with antibody fragments, and other strategies.

More recently, the use of camelid heavy-chain-only antibody fragments (nanobodies) as

crystallographic chaperones has revolutionized the field of GPCR structural biology,

aiding in the determination of more than 340 GPCR structures to date. In most cases,

the GPCR structures solved as complexes with nanobodies (Nbs) have revealed the

binding mode of cognate or non-natural ligands; in a few cases, the same Nb has acted

as an orthosteric or an allosteric modulator of GPCR signaling. In this review we

summarize the multiple ingenious strategies that have been conceived and

implemented in the last decade to capitalize on the discovery of nanobodies to study

GPCRs from a structural perspective.

3
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Significance Statement:

G protein-coupled receptors (GPCRs) are major pharmacological targets, and the

determination of their structures at high resolution has been essential for structure-

guided drug design and for insights about their functions. Single domain antibodies

(nanobodies) have greatly facilitated the structural determination of GPCRs, by forming

complexes directly with the receptors or indirectly through protein partners.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024

4
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Introduction:

The history of nanobodies (Nbs) dates back to 1993, when a serendipitous discovery in

the laboratory of Professor R. Hamers (Vrije Universiteit Brussel) revealed that some

camelid antibodies only have heavy chains (Hamers-Casterman et al., 1993). From this

observation, it was quickly realized that their variable region (VVH) could be engineered

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


into a fully functional ~14-kDa antibody fragment with wide potential applications,

including in structural biology (Desmyter et al., 1996; Hamers-Casterman et al., 1993).

Since Nbs have a single domain, they normally cannot bind linear epitopes. Instead,

their three complementarity-determining regions (CDRs) can insert into cavities of the

proteins, making them good at binding difficult-to-access concave epitopes and

stabilizing particular conformations. In the case of GPCRs, several dozen Nbs have

been found to insert one or more CDRs into the GPCR’s ligand-binding pocket, the helix

bundle, or other allosteric sites, stabilizing the active or inactive states.

In the last decade, Nbs have been particularly helpful in structural biology, especially in

obtaining structures of difficult targets such as membrane proteins, in studies

spearheaded by the laboratory of Jan Steyaert in Belgium (Vrije Universiteit Brussel)

(Uchanski et al., 2020). Not surprisingly, the field of GPCR-structural biology has

capitalized on these advances, and the number of structures of GPCRs determined with

the aid of Nbs has been growing since 2011 (Fig. 1). In addition, Nbs are also being

investigated as potential antibody-based therapeutics. Here we review the impact of

Nbs on elucidating structures of GPCRs, either by crystallography or cryo-electron

microscopy (cryo-EM), and the therapeutic potential of Nbs.

5
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024

6
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Modes of binding of nanobodies to GPCRs:

The binding mode of a Nb to a GPCR can be classified as direct or indirect, and

intracellular or extracellular. The GPCR database, GPCRDB [GPCRdb.org], lists all the

GPCR structures, including GPCRs bound to Nbs. Unfortunately, GPCRDB has not

been updated since August 2023. In Table S1, we list the structures of GPCR/Nb

complexes that were made public after that date. In Table S2, we list the PDB entries of

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


Nb-aided GPCR structures in the GPCR database (except for 214 GPCR structures

with Nb35). Here, we summarize the impact of Nbs in the determination of GPCR

structures, grouped by Nb-binding mode, with an emphasis on Nbs that bind directly to

the extracellular domain of the GPCRs.

1- Direct binding to the intracellular domain: The first-ever Nb-aided GPCR structure

was that of the β2-adrenergic receptor (β2-AR) with Nb80 bound to the intracellular

domain (ICD) (PDB 3P0G) (Rasmussen et al., 2011a). The β2-AR construct used in this

work had the flexible intracellular loop 3 (ICL3) replaced by T4 lysozyme (T4L). Fusions

with T4L and other small, thermostable proteins, such as BRIL (a thermostabilized

apocytochrome b562), thioredoxin (Trx) or rubredoxin are used routinely to increase the

expression and stability of GCPRs for structural studies (Chun et al., 2012). In the case

of the crystal structure of the complex between Nb80 and β2-AR/T4L construct, there

was no interpretable electron density for T4L. However, the CDR3 of Nb80 was clearly

visible binding deep inside the 7-helix bundle, mimicking the binding of G protein and

stabilizing the active conformation of the receptor (Fig. 2A). In most of the GPCR/Nb

structures solved to date, the Nbs bind to the intracellular side of the receptor and are

7
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

specific for the particular receptor (see examples in Fig. 2). Other examples of Nb-

CDRs reaching into the intracellular side of the 7-helix bundle, stabilizing the GPCR

active conformation, include additional structures of Nbs in complex with β2-AR with

different ligands (PDB 6N48 and 4QKX), β1-AR (Fig. 2B) (PDB 6IBL and 7BTS), and

the M2 muscarinic acetylcholine receptor (PDB 4MQS). In most cases, it is the CDR3

loop that penetrates the helix bundle; although in several cases, such as μ-opioid

receptor (μ-OR, PDB 5C1M) (Fig. 2C), and κ-OR (PDB 6B73, 7YIT), the Nbs have a

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


different mode of binding, with CDR2 instead being the loop reaching deeper inside the

bundle and stabilizing the active conformation.

In the case of US28, a human cytomegalovirus-encoded chemokine receptor that was

“stolen” from the human genome and evolved by the virus, single-chain fusions of US28

with intracellularly directed nanobodies were screened against yeast cells displaying

CX3CL1 chemokine libraries on their surface (Miles et al., 2018). For this work, the C-

terminus of US28 was fused to the nanobody Nb7 to enable the production of stable

US28 for staining yeast-displayed chemokine libraries. Crystallization of the

CX3CL1.35/US28-Nb7 complex by lipidic cubic phase required an additional nanobody,

raised by alpaca immunization against apo-US28-Nb7 (Fig. 2D). Comparison of the

structure of US28 bound to CX3CL1.35 and the wild type CX3CL1, as well as its

signaling activity bound to different CX3CL1 variants, suggested that signaling of US28

is largely unaffected by even drastic changes to the interactions within the receptor

binding cavity, providing structural basis for US28’s chemokine promiscuity.

Recently, a potentially universal strategy for Nb/GPCR-structural determination was

devised where ICL3 of other GPCRs could be replaced by the ICL3 from κ-OR, which is

8
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

recognized by the negative allosteric modulator Nb6 (Fig. 2E) (Che et al., 2020)

(Robertson et al., 2022). Using this loop-grafting strategy, the structure of several

GPCRs in inactive conformations have been solved in complex with Nb6 by cryo-EM:

histamine receptor 2 (H2R, PDB 7UL3), somatostatin receptor 2 (SSTR2, PDB 7UL5),

neurotensin 1 receptor (NTSR1, PDB 7UL2), α1A-AR (PDB 8HN1, 7YMJ), and

chemokine receptor CXCR3 (PDB 8K2W, 8HNN). In addition, the structure of μ-OR was

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


obtained with a megabody (see below) based on Nb6 (PDB 7UL4). Except for SSTR2,

the GPCRs in complex with Nb6 were also bound to antagonists.

2- Direct binding to the extracellular domain: Despite being the most interesting

binding mode because of its therapeutic potential, the structure of only six class-A

GPCRs have been solved to date with a nanobody directly bound to their extracellular

domain; in addition, four class-C GPCRs have been solved with Nbs bound to their

large extracellular domain (ECD) (Fig. 3).

The first structure of a GPCR with a bound extracellular Nb was that of APJ, a target for

the treatment of chronic heart failure (Ma et al., 2020) (Fig. 3A). Thus, Nb-JN241 was

found to make critical contacts with the second extracellular loop of APJ and insert

CDR3 into the ligand-binding pocket. Guided by this crystal structure, the authors

converted antagonist Nb-JN241 into a full agonist Nb-JN241-9 just by inserting a

tyrosine into the CDR3.

Next, the cryo-EM structure of the active orexin receptor 2 (OX2R) in complex with its G

protein and a small-molecule agonist was solved by stabilizing the complex with a

bound Nb (Hong et al., 2021) (Fig. 3B). OX2R is a potential target for the treatment of

9
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

narcolepsy; however, in this work, no modulation of activity of the receptor by the Nb

was reported.

μ-Opioid receptor (μ-OR) is the molecular target of opioid analgesics such as morphine

and fentanyl. The cryo-EM structure of μ-OR bound to the antagonist-nanobody NbE

was solved recently (Fig. 3C) (Yu et al., 2023). NbE displays a unique ligand-binding

mode and achieves selectivity for µ-OR through interactions with extracellular receptor

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


loops and deep penetration into the orthosteric pocket. Based on the structure of the

CDR3 loop of NbE, the authors designed short-peptide analogs that retain antagonism

toward µ-OR. In addition to NbE, the μ-OR complex also included an anti-Nb Fab and

an anti-Fab Nb. The work of Yu et al. illustrates the potential of Nbs to uniquely engage

with GPCRs, and it describes novel μ-OR ligands that can serve as the basis for

therapeutic development.

Rhodopsin is the prototypical, class-A GPCR, and multiple mutations in rhodopsin have

been identified to cause retinal degeneration. In the case of rhodopsin, the covalently

bound antagonist (11-cis-retinal) photo-isomerizes into the full agonist all-trans-retinal;

then, through a series of short-lived conformations, rhodopsin transforms into the fully

activated conformation Meta-II. Our lab recently developed a battery of Nbs that bind to

the extracellular domain of rhodopsin and act as allosteric modulators, preventing

rhodopsin to transition from the ground state to the Meta-II state upon photoactivation

(Wu et al., 2023). Furthermore, photoactivation of rhodopsin in isolation followed by

incubation with the Nb, shifts the equilibrium from active meta II-rhodopsin toward the

inactive Meta-I conformation, despite the binding pocket being occupied by the full

agonist all-trans-retinal. This Nb-induced conformational shift is also observed with apo-

10
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

rhodopsin (Fig. 3D), whose conformation in isolation is otherwise similar to that of Meta-

II. The crystal structures of Nb2 in complex with ground-state rhodopsin, photo-activated

rhodopsin and apo-rhodopsin revealed extensive interactions of the three CDRs of Nb2

with the N-terminus, ELC2 and, surprisingly, the two native N-glycans of rhodopsin.

These observations serve as a reminder that post-translational modifications of native

GPCRs like glycosylation, which often are overlooked, need to be taken into

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


consideration in the development of biologics. Additionally, we found out that the non-

variable framework region 2 (FR2) of Nb2 also was involved in extensive interactions

with the native glycans of rhodopsin. There was also another unexpected finding in our

work. Notably, unlike the other available structures of extracellular Nb/class-A GPCR

complexes where the Nb-CDR3 loop binds deep inside the orthosteric pocket of the

GPCR, the antagonistic activity of Nb2 toward rhodopsin is achieved by preventing

small conformational changes in the extracellular domain of rhodopsin that are

concomitant with the transition from ground-state to activated state. This effect is in

contrast with the mode of action of intracellular Nb modulators, since their activity is

facilitated by the major opening of the helix bundle upon GPCR activation.

Angiotensin II type I receptor (AT1R) modulates renal and cardiovascular function in

response to the eight-amino-acid peptide-hormone angiotensin II. Recently, the

structure of two fusion proteins Nb-AT1R/BRIL bound to anti-BRIL Fab have been

solved by cryo-EM (PDBs 8TH3, 8TH4) (Skiba et al., 2024). In both cases, the CDR3s

of the fused Nbs (AT118-H and AT118-L) were found to occupy the orthosteric binding

pocket and act as antagonists by competing with peptide ligands for binding to AT1R;

11
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

however, the two Nbs exhibited different levels of interference with small-molecule

antagonists.

The α1A-AR responds to adrenaline and noradrenaline and is involved in smooth muscle

contraction and cognitive function. Two structures of Nb29/α1A-AR/miniGsq have been

solved (Fig. 3E), in which an agonist (noradrenaline or oxymetazoline) is bound in the

orthosteric binding pocket, while the Nb29-CDR3 loop occupies the extracellular

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


vestibule of this binding pocket (PDB 7YMH and 7YM8). Nb29 covers the extracellular

surface of α1A-AR, analogous to the anti-APJ nanobody JN241 and anti-PAR2 Fab;

whereas the CDR3 loop of Nb29 binds to a site similar to that of LY2119620 (a weak

positive allosteric modulator) binding to M2R (Toyoda et al., 2023).

The Nb-aided structures of dimeric class-C GPCRs, which have large extracellular

domains (ECD), represent a special case. In recent work (Kumar et al., 2023), the cryo-

EM structures of mGlu5 by itself (8T7H) or bound to Nb43 through the ECD (PDB 8T6J,

8T7H, 8T8M, 8TAO) enabled the authors to propose a model for a sequential, multistep

activation mechanism for mGlu5 (Fig. 3F). Other structures of class-C GPCRs with Nb

bound to the extracellular domain are mGlu1 in an intermediate state (PBD 7DGE),

active mGlu2 (PDB 7EPB) and inactive CaSR (PDB 7E6U).

3- Indirect binding to the intracellular domain: The majority (~80%) of the Nb-

facilitated structures of GPCRs consist of complexes with a bound G protein which, in

turn, is bound to Nb35 (Fig. 4A). This nanobody stabilizes the active GPCR/G protein

complex by interacting with the Gαs and Gβ subunits, preventing dissociation of the

nucleotide-free complex by the nonhydrolyzable GTP analog GTPγS (Rasmussen et al.,

12
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

2011b). All GPCR/Gs/Nb35 structures have been determined by cryo-EM, except for

the D1A dopamine receptor (PDB 7JOZ) and β2-AR (PDB 3SN6), which were solved by

X-ray crystallography. In a few cases, a single-chain variable fragment (scFv) also

formed part of the complex.

Another mode of indirect binding in GPCR/Nb complexes is that of an anti-BRIL Fab

fragment bound to a GPCR/BRIL fusion construct, as shown for inactive Frizzled

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


receptor FZD3 (Fig. 4B). Finally, to obtain the cryo-EM structure of the inactive

glucagon receptor/Arr2 complex, without or with glucagon bound (PDB 8JRU and 8JRV,

respectively), multiple modifications were performed on the proteins to stabilize the

complex: the C-terminus of the receptor was replaced by the C-terminus of the

vasopressin V2 receptor to achieve optimal binding to a Cys-free Arr2; Arr2 was fused

to a scFv; and a Nb that binds to active Arr2 was added to the complex (Fig. 4C).

4- Indirect binding to the extracellular domain:

The only Nb-facilitated GPCR structure where the Nb is indirectly bound to the

extracellular domain of the GPCR is of that of atypical chemokine receptor 3 (ACKR3)

bound to chemokine CXCL12 (also called stromal cell-derived factor 1) (PDB 7SK7). In

that case, a Fab fragment that directly contacts the GPCR/ligand complex serves as an

intermediate binding partner between the Nb and the GPCR (Yen et al., 2022). In the

same article, the authors also solved another structure of the ACKR3/CXCL12 complex

with a Fab fragment plus a Nb, but in this second structure the Nb and Fab were bound

to the intracellular domain of the GPCR. These two structures, along with five additional

structures of ACKR3 without Nb but with different ligands and mutations, together with

13
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

functional studies, have provided insights about the ligand-binding promiscuity of

ACKR3, why it fails to couple to G proteins, and why it is biased toward regulation by

arrestin-2.

Use of nanobodies for therapeutic intervention and clinical imaging

Since the discovery of nanobodies, the interest in developing therapeutic nanobodies

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


has been growing rapidly, leading to the introduction of two FDA-approved nanobody

drugs into the clinical market (Morrison, 2019; Natrajan et al., 2024). More than three

dozen nanobodies have been entered into the clinical realm for various human-disease

indications, including solid tumors, chronic inflammatory disease, autoimmune and

infectious disorders, and brain neuroimaging (Table S3). Half of the clinical-stage

nanobody-drug candidates have been targeted to cancer-expressed antigens whose

high expression is often associated with tumor progression and immune response.

Over 370 GPCRs are potential pharmacological targets for drug discovery, as they are

implicated in many diseases and therapeutic strategies. Yet the vast majority of GPCR-

targeting nanobodies are still in the discovery stages of research and development, they

are currently being developed for a broad spectrum of disease indications, including

asthma, cancer, cardiovascular diseases, HIV infection, hyperparathyroidism,

hyperthyroidism, immune dysfunctions, inflammatory diseases, kidney dysfunction,

metabolic syndromes, migraine, neurological and neurodegenerative diseases,

osteoporosis, pain, psychiatric disorders, and vision disorders (Table 1). The selectivity

and regiospecificity of the binding of nanobodies enable diverse pharmacological effects

on the activities of the GPCRs by regulating various interactions, including occupation of

14
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

orthosteric binding pockets or allosteric sites, ligand binding affinity, stabilization of

specific conformations based on the Nb-epitope binding sites, and penetration of Nb-

CDRs into the receptor cores.

A unique feature of nanobodies from a therapeutic perspective is their use as molecular

chaperones for misfolded proteins associated with degenerative diseases, such as

amyloidosis and rhodopsin-associated retinal degeneration. Potentially therapeutic

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


nanobodies have been developed against amyloidogenic targets such as rhodopsin

(Wu et al., 2023), lysozyme (Dumoulin et al., 2003), Aβ-peptides (Habicht et al., 2007;

Lafaye et al., 2009), α-synuclein (Lafaye et al., 2009), β2-microglobulin (Domanska et

al., 2011), and gelsolin, which are involved, respectively, in autosomal dominant retinitis

pigmentosa, systemic amyloid disease, Alzheimer’s Disease, Parkinson’s disease, renal

failure, and gelsolin amyloidosis (Van Overbeke et al., 2014). One of the underlying

mechanisms of anti-amyloidogenic nanobodies is to inhibit amyloid fibril formation by

stabilizing oligomers and trapping them in intermediate fibrils. A recent publication

reported that rhodopsin-targeting Nb2 acted as a molecular chaperone to restore the

proteostasis of the misfolded P23H-mutant rhodopsin protein by stabilizing it in an

intermediate conformation (Wu et al., 2023).

Clinical applications of radiolabeled or fluorescent-labeled nanobodies have emerged

for in vivo molecular imaging methods, such as optical and positron-emission

tomography (PET)/computer tomography (CT) nuclear imaging, particularly in cancer

and brain. High specificity, affinity for the target antigen, and rapid renal clearance of

nanobody tracers are major advantages for in vivo imaging. Due to their small size,

nanobodies are expected to achieve a uniform distribution and deep tissue penetration,

15
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

an important rate-limiting factor in diagnostic imaging (Debie et al., 2020; Zheng et al.,

2022).

Current challenges, knowledge gaps and perspective:

Nbs are commonly generated by immunization of llamas or alpacas with the target

protein. However, large animal immunization has several downsides including cost and

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


accessibility, animal welfare challenges, and limitations on addressable targets since

immunological tolerance of self-antigens makes it difficult to raise nanobodies that bind

conserved epitopes. Over the past few years, several technological developments in

synthetic Nb generation have taken off. For example, in vitro discovery of Nbs using

phage display or yeast surface display (McMahon et al., 2018; Moutel et al., 2016;

Uchanski et al., 2019) have led to the discovery of countless Nbs, including ones

targeting GPCRs such as AT1R on both the intracellular and extracellular interface

(McMahon et al., 2020; Wingler et al., 2019). For such approaches to yield potent Nbs,

large synthetic Nb libraries must be constructed from which target-specific binders can

be enriched. Often, the enriched binders have weak affinity and need to undergo affinity

maturation through repeated rounds of error-prone PCR and selection. More recently, in

vitro Nb discovery was upgraded through the implementation of autonomously

diversifying yeast-displayed Nbs (Wellner et al., 2021) that encode Nbs or Nb libraries

on a rapidly hypermutating plasmid system (Ravikumar et al., 2018; Rix et al., 2023).

These systems allow for Nbs to continuously diversify, and therefore rapidly affinity

mature, as they are subjected to simple rounds of sorting for target binding and growth.

Another way to produce heavy-chain only single-domain antibodies is to generate

16
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

transgenic mouse lines with either fully humanized or hybrid llama/human variable

domains genes (Eden et al., 2024; Janssens et al., 2006; Teng et al., 2020; Xu et al.,

2021). These methods still rely on animal immunization, but of mice instead of alpacas

or llamas, providing obvious advantages. A remarkable recent advancement is the

possibility of de novo computational design of Nbs in cases where the structure of the

epitope is known (Bennett et al., 2024). Here, diffusion-based generative models for

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


protein design nominate epitope-specific synthetic Nb designs that can be further

screened and improved by yeast display, with the eventual goal of direct design of Nbs

against any epitope without needing animal immunization nor in vitro engineering

approaches.

A particular gap in our knowledge is how Nbs can modulate the activity of GPCRs from

the extracellular side. The presence of PTMs such as glycosylation could be a major

obstacle for translating Nbs from in vitro proof-of-principle status to in vivo therapeutic

application, since most GPCRs are N- and/or O-glycosylated at the N-terminus and

extracellular loops. The discovery and characterization of antibodies targeting GPCRs is

usually done using recombinant GPCRs, in many cases with mutations, deletions, or

deglycosylase treatments to remove N-glycans. The case of a Nb targeting bRho that

does not recognize deglycosylated or hyperglycosylated bRho (Wu et al., 2023)

illustrates how the presence of native glycans in the GPCR is necessary for relevant

development and characterization of Nbs as potential therapeutics.

The future of Nb-facilitated GPCR structural biology seems to be tied to advances in

strategies to increase the molecular weight (MW) of the complexes required for

structural determination by cryo-EM. Once a GPCR-targeted Nb becomes available, it is

17
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

possible to increase its MW by engineering a megabody (Mb), which are generated by

circular permutation of a Nb with a stable, mid-size unrelated protein (Uchanski et al.,

2021). μ-OR/Mb6 was the first GPCR/Mb complex solved, although most of the Mb

structure was not solved (PDB 7UL4) (Robertson et al., 2022). Other similar strategies

involve adding to the protein/Nb complex an anti-Nb Fab fragment plus an MBP/Protein-

A construct (Legobodies) (Wu and Rapoport, 2021). Finally, it is possible to increase the

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


MW of a GPCR/Nb complex by addition of a Fab fragment that recognizes the Nb; and,

then, an anti-Fab Nb (for example, Fig. 3C in (Bloch et al., 2021)).

Another interesting approach is the use of intrabodies in GPCR research (Manglik et al.,

2017), where nanobodies are expressed de novo within cells for multiple applications,

such as to visualize or modulate antigens in the living cell (Wagner and Rothbauer,

2020); or to study protein conformational states (Uchanski et al., 2020). Among the

reported applications of anti-GPCR intrabodies are inhibition of G protein activation,

GRK-mediated receptor phosphorylation, β-arrestin recruitment, and receptor

internalization (Manglik et al., 2017).

18
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Acknowledgements

We thank Dr. Jeffrey L. Benovic, Montserrat Salom and members of the Center

for Translational Vision Research and the Gavin Herbert Eye Institute for their

comments and insights on this manuscript.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024

19
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Data Availability Statement

The authors declare that all the data supporting the findings of this study are available

within the paper and its Supplemental Data

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024

20
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Salom D., Wu, A., Liu C., and

Palczewski K.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024

21
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

References

Bennett NR, Watson JL, Ragotte RJ, Borst AJ, See DL, Weidle C, Biswas R, Shrock EL, Leung PJY, Huang B,
Goreshnik I, Ault R, Carr KD, Singer B, Criswell C, Vafeados D, Sanchez MG, Kim HM, Torres SV,
Chan S and Baker D (2024) Atomically accurate de novo design of single-domain antibodies.
bioRxiv.
Bloch JS, Mukherjee S, Kowal J, Filippova EV, Niederer M, Pardon E, Steyaert J, Kossiakoff AA and Locher
KP (2021) Development of a universal nanobody-binding Fab module for fiducial-assisted cryo-
EM studies of membrane proteins. Proc Natl Acad Sci U S A 118(47).
Bobkov V, Zarca AM, Van Hout A, Arimont M, Doijen J, Bialkowska M, Toffoli E, Klarenbeek A, van der
Woning B, van der Vliet HJ, Van Loy T, de Haard H, Schols D, Heukers R and Smit MJ (2018)
Nanobody-Fc constructs targeting chemokine receptor CXCR4 potently inhibit signaling and

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


CXCR4-mediated HIV-entry and induce antibody effector functions. Biochem Pharmacol 158:
413-424.
Bradley ME, Dombrecht B, Manini J, Willis J, Vlerick D, De Taeye S, Van den Heede K, Roobrouck A, Grot
E, Kent TC, Laeremans T, Steffensen S, Van Heeke G, Brown Z, Charlton SJ and Cromie KD (2015)
Potent and efficacious inhibition of CXCR2 signaling by biparatopic nanobodies combining two
distinct modes of action. Mol Pharmacol 87(2): 251-262.
Burg JS, Ingram JR, Venkatakrishnan AJ, Jude KM, Dukkipati A, Feinberg EN, Angelini A, Waghray D, Dror
RO, Ploegh HL and Garcia KC (2015) Structural basis for chemokine recognition and activation of
a viral G protein-coupled receptor. Science 347(6226): 1113-1117.
Che T, English J, Krumm BE, Kim K, Pardon E, Olsen RHJ, Wang S, Zhang S, Diberto JF, Sciaky N, Carroll FI,
Steyaert J, Wacker D and Roth BL (2020) Nanobody-enabled monitoring of kappa opioid receptor
states. Nat Commun 11(1): 1145.
Che T, Majumdar S, Zaidi SA, Ondachi P, McCorvy JD, Wang S, Mosier PD, Uprety R, Vardy E, Krumm BE,
Han GW, Lee MY, Pardon E, Steyaert J, Huang XP, Strachan RT, Tribo AR, Pasternak GW, Carroll FI,
Stevens RC, Cherezov V, Katritch V, Wacker D and Roth BL (2018) Structure of the Nanobody-
Stabilized Active State of the Kappa Opioid Receptor. Cell 172(1-2): 55-67 e15.
Cheloha RW, Fischer FA, Woodham AW, Daley E, Suminski N, Gardella TJ and Ploegh HL (2020) Improved
GPCR ligands from nanobody tethering. Nat Commun 11(1): 2087.
Chen XC, Wang L, Cui QQ, Ding ZY, Han L, Kou YJ, Zhang WQ, Wang HN, Jia XM, Dai M, Shi ZZ, Li YY, Li XY
and Geng Y (2021) Structural insights into the activation of human calcium-sensing receptor. Elife
10.
Chun E, Thompson AA, Liu W, Roth CB, Griffith MT, Katritch V, Kunken J, Xu F, Cherezov V, Hanson MA and
Stevens RC (2012) Fusion partner toolchest for the stabilization and crystallization of G protein-
coupled receptors. Structure 20(6): 967-976.
Cui QQ, Wang L, Wang HN, Chen XC, Han L, Geng TJ, Kou YJ, Zhang WQ, Dai M, Qiao HR, Sun ZC, Li LY, Lan
ZY, Xu HX, Xu JQ, Dai YY and Geng Y (2024) Nanobodies as negative allosteric modulators for
human calcium sensing receptor. Biochem Bioph Res Co 695.
De Groof TWM, Bergkamp ND, Heukers R, Giap T, Bebelman MP, Goeij-de Haas R, Piersma SR, Jimenez
CR, Garcia KC, Ploegh HL, Siderius M and Smit MJ (2021) Selective targeting of ligand-dependent
and -independent signaling by GPCR conformation-specific anti-US28 intrabodies. Nat Commun
12(1): 4357.
de Wit RH, Heukers R, Brink HJ, Arsova A, Maussang D, Cutolo P, Strubbe B, Vischer HF, Bachelerie F and
Smit MJ (2017) CXCR4-Specific Nanobodies as Potential Therapeutics for WHIM syndrome. J
Pharmacol Exp Ther 363(1): 35-44.

22
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Debie P, Lafont C, Defrise M, Hansen I, van Willigen DM, van Leeuwen FWB, Gijsbers R, D'Huyvetter M,
Devoogdt N, Lahoutte T, Mollard P and Hernot S (2020) Size and affinity kinetics of nanobodies
influence targeting and penetration of solid tumours. J Control Release 317: 34-42.
Deshpande I, Liang J, Hedeen D, Roberts KJ, Zhang Y, Ha B, Latorraca NR, Faust B, Dror RO, Beachy PA,
Myers BR and Manglik A (2019) Smoothened stimulation by membrane sterols drives Hedgehog
pathway activity. Nature 571(7764): 284-288.
Desmyter A, Transue TR, Ghahroudi MA, Thi MH, Poortmans F, Hamers R, Muyldermans S and Wyns L
(1996) Crystal structure of a camel single-domain VH antibody fragment in complex with
lysozyme. Nat Struct Biol 3(9): 803-811.
Domanska K, Vanderhaegen S, Srinivasan V, Pardon E, Dupeux F, Marquez JA, Giorgetti S, Stoppini M,
Wyns L, Bellotti V and Steyaert J (2011) Atomic structure of a nanobody-trapped domain-
swapped dimer of an amyloidogenic beta2-microglobulin variant. Proc Natl Acad Sci U S A
108(4): 1314-1319.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


Dumoulin M, Last AM, Desmyter A, Decanniere K, Canet D, Larsson G, Spencer A, Archer DB, Sasse J,
Muyldermans S, Wyns L, Redfield C, Matagne A, Robinson CV and Dobson CM (2003) A camelid
antibody fragment inhibits the formation of amyloid fibrils by human lysozyme. Nature
424(6950): 783-788.
Eden T, Schaffrath AZ, Wesolowski J, Stahler T, Tode N, Richter N, Schafer W, Hambach J, Hermans-
Borgmeyer I, Woens J, Le Gall CM, Wendler S, Linke-Winnebeck C, Stobbe M, Budnicki I, Wanney
A, Heitz Y, Schimmelpfennig L, Schweitzer L, Zimmer D, Stahl E, Seyfried F, Gebhardt AJ, Dieckow
L, Riecken K, Fehse B, Bannas P, Magnus T, Verdoes M, Figdor CG, Hartlepp KF, Schleer H, Funer J,
Tomas NM, Haag F, Rissiek B, Mann AM, Menzel S and Koch-Nolte F (2024) Generation of
nanobodies from transgenic 'LamaMice' lacking an endogenous immunoglobulin repertoire. Nat
Commun 15(1): 4728.
English JG, Olsen RHJ, Lansu K, Patel M, White K, Cockrell AS, Singh D, Strachan RT, Wacker D and Roth BL
(2019) VEGAS as a Platform for Facile Directed Evolution in Mammalian Cells (vol 178, pg 748,
2019). Cell 178(4): 1030-1030.
Eshak F, Pion L, Scholler P, Nevoltris D, Chames P, Rondard P, Pin JP, Acher FC and Goupil-Lamy A (2024)
Epitope Identification of an mGlu5 Receptor Nanobody Using Physics-Based Molecular Modeling
and Deep Learning Techniques. Journal of Chemical Information and Modeling.
Habicht G, Haupt C, Friedrich RP, Hortschansky P, Sachse C, Meinhardt J, Wieligmann K, Gellermann GP,
Brodhun M, Götz J, Halbhuber KJ, Röcken C, Horn U and Fändrich M (2007) Directed selection of
a conformational antibody domain that prevents mature amyloid fibril formation by stabilizing
Aβ protofibrils. P Natl Acad Sci USA 104(49): 19232-19237.
Haffke M, Fehlmann D, Rummel G, Boivineau J, Duckely M, Gommermann N, Cotesta S, Sirockin F,
Freuler F, Littlewood-Evans A, Kaupmann K and Jaakola VP (2019) Structural basis of species-
selective antagonist binding to the succinate receptor. Nature 574(7779): 581-585.
Hamers-Casterman C, Atarhouch T, Muyldermans S, Robinson G, Hamers C, Songa EB, Bendahman N and
Hamers R (1993) Naturally occurring antibodies devoid of light chains. Nature 363(6428): 446-
448.
Hao S, Xu S, Li L, Li Y, Zhao M, Chen J, Zhu S, Xie Y, Jiang H, Zhu J and Wu M (2022) Tumour inhibitory
activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4. BMC Cancer
22(1): 1092.
Haubrich J, Font J, Quast RB, Goupil-Lamy A, Scholler P, Nevoltris D, Acher F, Chames P, Rondard P,
Prézeau L and Pin JP (2021) A nanobody activating metabotropic glutamate receptor 4
discriminates between homo-and heterodimers. P Natl Acad Sci USA 118(33).
Heukers R, De Groof TWM and Smit MJ (2019) Nanobodies detecting and modulating GPCRs outside in
and inside out. Curr Opin Cell Biol 57: 115-122.

23
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Heukers R, Fan TS, de Wit RH, van Senten JR, De Groof TWM, Bebelman MP, Lagerweij T, Vieira J, de
Munnik SM, Smits-de Vries L, van Offenbeek J, Rahbar A, van Hoorick D, Soderberg-Naucler C,
Wurdinger T, Leurs R, Siderius M, Vischer HF and Smit MJ (2018) The constitutive activity of the
virally encoded chemokine receptor US28 accelerates glioblastoma growth. Oncogene 37(30):
4110-4121.
Hong C, Byrne NJ, Zamlynny B, Tummala S, Xiao L, Shipman JM, Partridge AT, Minnick C, Breslin MJ, Rudd
MT, Stachel SJ, Rada VL, Kern JC, Armacost KA, Hollingsworth SA, O'Brien JA, Hall DL, McDonald
TP, Strickland C, Brooun A, Soisson SM and Hollenstein K (2021) Structures of active-state orexin
receptor 2 rationalize peptide and small-molecule agonist recognition and receptor activation.
Nat Commun 12(1): 815.
Huang W, Manglik A, Venkatakrishnan AJ, Laeremans T, Feinberg EN, Sanborn AL, Kato HE, Livingston KE,
Thorsen TS, Kling RC, Granier S, Gmeiner P, Husbands SM, Traynor JR, Weis WI, Steyaert J, Dror
RO and Kobilka BK (2015) Structural insights into micro-opioid receptor activation. Nature

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


524(7565): 315-321.
Jahnichen S, Blanchetot C, Maussang D, Gonzalez-Pajuelo M, Chow KY, Bosch L, De Vrieze S, Serruys B,
Ulrichts H, Vandevelde W, Saunders M, De Haard HJ, Schols D, Leurs R, Vanlandschoot P, Verrips
T and Smit MJ (2010) CXCR4 nanobodies (VHH-based single variable domains) potently inhibit
chemotaxis and HIV-1 replication and mobilize stem cells. Proc Natl Acad Sci U S A 107(47):
20565-20570.
Janssens R, Dekker S, Hendriks RW, Panayotou G, van Remoortere A, San JK, Grosveld F and Drabek D
(2006) Generation of heavy-chain-only antibodies in mice. Proc Natl Acad Sci U S A 103(41):
15130-15135.
Kleist AB, Jenjak S, Sente A, Laskowski LJ, Szpakowska M, Calkins MM, Anderson EI, McNally LM, Heukers
R, Bobkov V, Peterson FC, Thomas MA, Chevigne A, Smit MJ, McCorvy JD, Babu MM and Volkman
BF (2022) Conformational selection guides beta-arrestin recruitment at a biased G protein-
coupled receptor. Science 377(6602): 222-228.
Koehl A, Hu H, Feng D, Sun B, Zhang Y, Robertson MJ, Chu M, Kobilka TS, Laeremans T, Steyaert J,
Tarrasch J, Dutta S, Fonseca R, Weis WI, Mathiesen JM, Skiniotis G and Kobilka BK (2019)
Structural insights into the activation of metabotropic glutamate receptors. Nature 566(7742):
79-84.
Kruse AC, Ring AM, Manglik A, Hu J, Hu K, Eitel K, Hubner H, Pardon E, Valant C, Sexton PM,
Christopoulos A, Felder CC, Gmeiner P, Steyaert J, Weis WI, Garcia KC, Wess J and Kobilka BK
(2013) Activation and allosteric modulation of a muscarinic acetylcholine receptor. Nature
504(7478): 101-106.
Kumar KK, Wang H, Habrian C, Latorraca NR, Xu J, O'Brien ES, Zhang C, Montabana E, Koehl A, Marqusee
S, Isacoff EY and Kobilka BK (2023) Step-wise activation of a Family C GPCR. bioRxiv.
Lafaye P, Achour I, England P, Duyckaerts C and Rougeon F (2009) Single-domain antibodies recognize
selectively small oligomeric forms of amyloid β, prevent Aβ-induced neurotoxicity and inhibit
fibril formation. Mol Immunol 46(4): 695-704.
Low S, Wu H, Jerath K, Tibolla A, Fogal B, Conrad R, MacDougall M, Kerr S, Berger V, Dave R, Villalona J,
Pantages L, Ahlberg J, Li H, Van Hoorick D, Ververken C, Broadwater J, Waterman A, Singh S and
Kroe-Barrett R (2020) VHH antibody targeting the chemokine receptor CX3CR1 inhibits
progression of atherosclerosis. MAbs 12(1): 1709322.
Lv Z, He Y, Xiang Y, Li J, Zhang S, Meng F, Lan B, Guo H, He D, Wang Y, Zhao H, Zhuo W, Liu Y, Liu X, Ni X and
Heng J (2022) Cryo-EM complex structure of active GPR75 with a nanobody. bioRxiv.
Ma Y, Ding Y, Song X, Ma X, Li X, Zhang N, Song Y, Sun Y, Shen Y, Zhong W, Hu LA, Ma Y and Zhang MY
(2020) Structure-guided discovery of a single-domain antibody agonist against human apelin
receptor. Sci Adv 6(3): eaax7379.

24
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Manglik A, Kobilka BK and Steyaert J (2017) Nanobodies to Study G Protein-Coupled Receptor Structure
and Function. Annu Rev Pharmacol Toxicol 57: 19-37.
Maussang D, Mujic-Delic A, Descamps FJ, Stortelers C, Vanlandschoot P, Stigter-van Walsum M, Vischer
HF, van Roy M, Vosjan M, Gonzalez-Pajuelo M, van Dongen GA, Merchiers P, van Rompaey P and
Smit MJ (2013) Llama-derived single variable domains (nanobodies) directed against chemokine
receptor CXCR7 reduce head and neck cancer cell growth in vivo. J Biol Chem 288(41): 29562-
29572.
McMahon C, Baier AS, Pascolutti R, Wegrecki M, Zheng S, Ong JX, Erlandson SC, Hilger D, Rasmussen SGF,
Ring AM, Manglik A and Kruse AC (2018) Yeast surface display platform for rapid discovery of
conformationally selective nanobodies. Nat Struct Mol Biol 25(3): 289-296.
McMahon C, Staus DP, Wingler LM, Wang JL, Skiba MA, Elgeti M, Hubbell WL, Rockman HA, Kruse AC and
Lefkowitz RJ (2020) Synthetic nanobodies as angiotensin receptor blockers. P Natl Acad Sci USA
117(33): 20284-20291.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


Meng J, Xu C, Lafon PA, Roux S, Mathieu M, Zhou R, Scholler P, Blanc E, Becker JAJ, Le Merrer J, Gonzalez-
Maeso J, Chames P, Liu J, Pin JP and Rondard P (2022) Nanobody-based sensors reveal a high
proportion of mGlu heterodimers in the brain. Nat Chem Biol 18(8): 894-903.
Miles TF, Spiess K, Jude KM, Tsutsumi N, Burg JS, Ingram JR, Waghray D, Hjorto GM, Larsen O, Ploegh HL,
Rosenkilde MM and Garcia KC (2018) Viral GPCR US28 can signal in response to chemokine
agonists of nearly unlimited structural degeneracy. Elife 7.
Morrison C (2019) Nanobody approval gives domain antibodies a boost. Nat Rev Drug Discov 18(7): 485-
487.
Moutel S, Bery N, Bernard V, Keller L, Lemesre E, de Marco A, Ligat L, Rain JC, Favre G, Olichon A and
Perez F (2016) NaLi-H1: A universal synthetic library of humanized nanobodies providing highly
functional antibodies and intrabodies. Elife 5.
Natrajan K, Kaushal M, George B, Kanapuru B and Theoret MR (2024) FDA Approval Summary:
Ciltacabtagene Autoleucel for Relapsed or Refractory Multiple Myeloma. Clin Cancer Res.
Peyrassol X, Laeremans T, Gouwy M, Lahura V, Debulpaep M, Van Damme J, Steyaert J, Parmentier M
and Langer I (2016) Development by Genetic Immunization of Monovalent Antibodies
(Nanobodies) Behaving as Antagonists of the Human ChemR23 Receptor. J Immunol 196(6):
2893-2901.
Rasmussen SG, Choi HJ, Fung JJ, Pardon E, Casarosa P, Chae PS, Devree BT, Rosenbaum DM, Thian FS,
Kobilka TS, Schnapp A, Konetzki I, Sunahara RK, Gellman SH, Pautsch A, Steyaert J, Weis WI and
Kobilka BK (2011a) Structure of a nanobody-stabilized active state of the beta(2) adrenoceptor.
Nature 469(7329): 175-180.
Rasmussen SG, DeVree BT, Zou Y, Kruse AC, Chung KY, Kobilka TS, Thian FS, Chae PS, Pardon E, Calinski D,
Mathiesen JM, Shah ST, Lyons JA, Caffrey M, Gellman SH, Steyaert J, Skiniotis G, Weis WI,
Sunahara RK and Kobilka BK (2011b) Crystal structure of the beta2 adrenergic receptor-Gs
protein complex. Nature 477(7366): 549-555.
Ravikumar A, Arzumanyan GA, Obadi MKA, Javanpour AA and Liu CC (2018) Scalable, Continuous
Evolution of Genes at Mutation Rates above Genomic Error Thresholds. Cell 175(7): 1946-+.
Ring AM, Manglik A, Kruse AC, Enos MD, Weis WI, Garcia KC and Kobilka BK (2013) Adrenaline-activated
structure of beta2-adrenoceptor stabilized by an engineered nanobody. Nature 502(7472): 575-
579.
Rix G, Williams RL, Spinner H, Hu VJ, Marks DS and Liu CC (2023) Continuous evolution of user-defined
genes at 1-million-times the genomic mutation rate. bioRxiv.
Robertson MJ, Papasergi-Scott MM, He F, Seven AB, Meyerowitz JG, Panova O, Peroto MC, Che T and
Skiniotis G (2022) Structure determination of inactive-state GPCRs with a universal nanobody.
Nat Struct Mol Biol 29(12): 1188-1195.

25
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Scholler P, Nevoltris D, de Bundel D, Bossi S, Moreno-Delgado D, Rovira X, Moller TC, El Moustaine D,


Mathieu M, Blanc E, McLean H, Dupuis E, Mathis G, Trinquet E, Daniel H, Valjent E, Baty D,
Chames P, Rondard P and Pin JP (2017) Allosteric nanobodies uncover a role of hippocampal
mGlu2 receptor homodimers in contextual fear consolidation. Nat Commun 8(1): 1967.
Skiba MA, Sterling SM, Rawson S, Zhang S, Xu H, Jiang H, Nemeth GR, Gilman MSA, Hurley JD, Shen P,
Staus DP, Kim J, McMahon C, Lehtinen MK, Rockman HA, Barth P, Wingler LM and Kruse AC
(2024) Antibodies expand the scope of angiotensin receptor pharmacology. Nat Chem Biol.
Staus DP, Wingler LM, Strachan RT, Rasmussen SGF, Pardon E, Ahn S, Steyaert J, Kobilka BK and Lefkowitz
RJ (2014) Regulation of β-Adrenergic Receptor Function by Conformationally Selective Single-
Domain Intrabodies. Molecular Pharmacology 85(3): 472-481.
Teng Y, Young JL, Edwards B, Hayes P, Thompson L, Johnston C, Edwards C, Sanders Y, Writer M, Pinto D,
Zhang Y, Roode M, Chovanec P, Matheson L, Corcoran AE, Fernandez A, Montoliu L, Rossi B,
Tosato V, Gjuracic K, Nikitin D, Bruschi C, McGuinness B, Sandal T and Romanos M (2020) Diverse

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


human V(H) antibody fragments with bio-therapeutic properties from the Crescendo Mouse. N
Biotechnol 55: 65-76.
Toyoda Y, Zhu A, Kong F, Shan S, Zhao J, Wang N, Sun X, Zhang L, Yan C, Kobilka BK and Liu X (2023)
Structural basis of alpha(1A)-adrenergic receptor activation and recognition by an extracellular
nanobody. Nat Commun 14(1): 3655.
Uchanski T, Masiulis S, Fischer B, Kalichuk V, Lopez-Sanchez U, Zarkadas E, Weckener M, Sente A, Ward P,
Wohlkonig A, Zogg T, Remaut H, Naismith JH, Nury H, Vranken W, Aricescu AR, Pardon E and
Steyaert J (2021) Megabodies expand the nanobody toolkit for protein structure determination
by single-particle cryo-EM. Nat Methods 18(1): 60-68.
Uchanski T, Pardon E and Steyaert J (2020) Nanobodies to study protein conformational states. Curr Opin
Struct Biol 60: 117-123.
Uchanski T, Zogg T, Yin J, Yuan D, Wohlkonig A, Fischer B, Rosenbaum DM, Kobilka BK, Pardon E and
Steyaert J (2019) An improved yeast surface display platform for the screening of nanobody
immune libraries. Sci Rep 9(1): 382.
Van Hout A, Klarenbeek A, Bobkov V, Doijen J, Arimont M, Zhao C, Heukers R, Rimkunas R, de Graaf C,
Verrips T, van der Woning B, de Haard H, Rucker JB, Vermeire K, Handel T, Van Loy T, Smit MJ and
Schols D (2018) CXCR4-targeting nanobodies differentially inhibit CXCR4 function and HIV entry.
Biochem Pharmacol 158: 402-412.
Van Overbeke W, Verhelle A, Everaert I, Zwaenepoel O, Vandekerckhove J, Cuvelier C, Derave W and
Gettemans J (2014) Chaperone nanobodies protect gelsolin against MT1-MMP degradation and
alleviate amyloid burden in the gelsolin amyloidosis mouse model. Mol Ther 22(10): 1768-1778.
Wagner TR and Rothbauer U (2020) Nanobodies Right in the Middle: Intrabodies as Toolbox to Visualize
and Modulate Antigens in the Living Cell. Biomolecules 10(12).
Warne T, Edwards PC, Dore AS, Leslie AGW and Tate CG (2019) Molecular basis for high-affinity agonist
binding in GPCRs. Science 364(6442): 775-778.
Wellner A, McMahon C, Gilman MSA, Clements JR, Clark S, Nguyen KM, Ho MH, Hu VJ, Shin JE, Feldman
J, Hauser BM, Caradonna TM, Wingler LM, Schmidt AG, Marks DS, Abraham J, Kruse AC and Liu
CC (2021) Rapid generation of potent antibodies by autonomous hypermutation in yeast. Nat
Chem Biol 17(10): 1057-1064.
Wingler LM and Feld AP (2022) Nanobodies as Probes and Modulators of Cardiovascular G Protein-
Coupled Receptors. J Cardiovasc Pharm 80(3): 342-353.
Wingler LM, McMahon C, Staus DP, Lefkowitz RJ and Kruse AC (2019) Distinctive Activation Mechanism
for Angiotensin Receptor Revealed by a Synthetic Nanobody. Cell 176(3): 479-490 e412.

26
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Wu A, Salom D, Hong JD, Tworak A, Watanabe K, Pardon E, Steyaert J, Kandori H, Katayama K, Kiser PD
and Palczewski K (2023) Structural basis for the allosteric modulation of rhodopsin by nanobody
binding to its extracellular domain. Nat Commun 14(1): 5209.
Wu X and Rapoport TA (2021) Cryo-EM structure determination of small proteins by nanobody-binding
scaffolds (Legobodies). Proc Natl Acad Sci U S A 118(41).
Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Lorenzi JCC, Park S, Schmidt F, Wang Z, Huang Y, Luo
Y, Nair MS, Wang P, Schulz JE, Tessarollo L, Bylund T, Chuang GY, Olia AS, Stephens T, Teng IT,
Tsybovsky Y, Zhou T, Munster V, Ho DD, Hatziioannou T, Bieniasz PD, Nussenzweig MC, Kwong PD
and Casellas R (2021) Nanobodies from camelid mice and llamas neutralize SARS-CoV-2 variants.
Nature 595(7866): 278-282.
Yen YC, Schafer CT, Gustavsson M, Eberle SA, Dominik PK, Deneka D, Zhang P, Schall TJ, Kossiakoff AA,
Tesmer JJG and Handel TM (2022) Structures of atypical chemokine receptor 3 reveal the basis
for its promiscuity and signaling bias. Sci Adv 8(28): eabn8063.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


Yu J, Kumar A, Zhang X, Martin C, Raia P, Koehl A, Laeremans T, Steyaert J, Manglik A, Ballet S, Boland A
and Stoeber M (2023) Structural Basis of mu-Opioid Receptor-Targeting by a Nanobody
Antagonist. bioRxiv.
Zheng F, Pang Y, Li L, Pang Y, Zhang J, Wang X and Raes G (2022) Applications of nanobodies in brain
diseases. Front Immunol 13: 978513.

27
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Footnotes

Financial support: This work was supported in part by grants from the National

Institutes of Health [grants R01EY009339, R01EY0034519, P30EY034070] (K.P., K.P.

and core grant to Department of Ophthalmology, Gavin Herbert Eye Institute at the

University of California, Irvine). The authors acknowledge support to the Department of

Ophthalmology Gavin Herbert Eye Institute at the University of California, Irvine from an

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


unrestricted Research to Prevent Blindness award. The authors also acknowledge the

Institute for Rapid Antibody Engineering and Evolution, part of the Engineering+Health

Initiative of the UCI Samueli School of Engineering, for additional support.

Conflict of interest:

CCL is a co-founder of K2 Biotechnologies, Inc., which uses OrthoRep for protein engineering.

K.P. is a consultant for Polgenix Inc. and serves on the Scientific Advisory Board at Hyperion

Eye Ltd.

Reprint requests: Krzysztof Palczewski, 837 Health Sciences Road, Gillespie

Neuroscience Building, Room 2105, Irvine, CA 92697-4375, email: [email protected]

28
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Figure Legends

Figure 1: Number of PDB entries of nanobody-facilitated GPCR structures, solved

either via X-ray crystallography or cryo-EM. The GPCR database

(gpcrdb.org/structure) was used to compile the structures released up to 8-16-2023, and

the Protein Data Base (rcsb.org) for structures released after that date. Note that some

GPCRs are represented by multiple PDB entries.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


* The value of PDB entries for 2024 is estimated based on the number of PDBs

released at the time of submission of this review

Figure 2: Five representative crystal structures of nanobodies in direct contact with the

intracellular domain of a GPCR. A) β2-adrenergic receptor (PDB 3P0G); B) T4L/β1-

adrenergic receptor fusion protein (PDB 7BTS); C) µ-opioid receptor (PDB 5C1M); D)

US28/Nb7 fusion protein (PDB 5WB2); E) κ-opioid receptor (PDB 6VI4).

Figure 3: Six (crystal or cryo-EM) structures of GPCRs with nanobodies directly bound

to their extracellular domains. A) APJ/rubredoxin fusion protein (PDB 6KNM); B) orexin

receptor 2/Gs protein complex (PDB 7L1V); C) µ-opioid receptor (PDB 8QOT); D) rod

opsin (PDB 8FD0); E) α1-adrenergic receptor in complex with an engineered minimal

Gsq protein (PDB 7YM8); F) metabotropic glutamate receptor mGlu5 (PDB 8TAO).

Figure 4: Three examples of cryo-EM structures of GPCRs in complex with different

proteins, facilitated by an Nb not in direct contact with the GPCR. A) Adenosine-A2A

29
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

receptor in complex with a mini-Gs protein (PDB 6GDG); B) Frizzled receptor

FZD3/BRIL fusion protein bound to anti-BRIL Fab fragment (PDB 8JHC); C) glucagon

receptor in complex with arrestin-2 fused to a scFv (PDB 8JRV).

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024

30
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Table 1. GPCR-targeting nanobodies (as of June 2024)

Target Nb clones Epitope Pharmacology References


Rho Nb2 ECD Antagonist (Wu et al., 2023)
CXCR2 2B2, 127D1, 54B12, 97A9, ECD Antagonist, inverse agonist (Bradley et al., 2015)
163E3, 163D2,
bivalent 2B2-35GS-2B2,
163E3-35GS-163E3
and 127D1-35GS-163E3
CXCR4 238D2, 238D4, bivalent ECD Antagonist, inverse agonist (Jahnichen et al., 2010)
L3-L8
CXCR4 10A10, bivalent 10A10 ECD Antagonist, inverse agonist (de Wit et al., 2017)
CXCR4 VUN401-VUN409 ECD Antagonist (Van Hout et al., 2018)
CXCR4 VUN400-Fc, VUN401-Fc, ECD Antagonist (Bobkov et al., 2018)

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


VUN402-Fc
CXCR4 Bispecific Nb PX4 ECD Antagonist (Hao et al., 2022)
(PD-V1 and CXCR4)
ACKR3 NB1-5, Nb38 ECD Antagonist, inverse agonist (Maussang et al., 2013)
(CXCR7)
ACKR3 VUN701 ECD Antagonist (Kleist et al., 2022)
(CXCR7)
CX3CR1 54A12, 54D05, 66B02 ECD Antagonist (Low et al., 2020)
66G01, BI655088, BI655089
US28 Nb7 ICD Active (Burg et al., 2015)
US28 VUN103 ICD Antagonist (De Groof et al., 2021)
US28 US28-Nb, bivalent US28-Nb ECD Antagonist, inverse agonist (Heukers et al., 2018)
ChemR23 CA4910, CA5183, ECD Antagonist (Peyrassol et al., 2016)
bivalent CA4910
mGlu2 DN1, DN10, DN13 ECD Partial, full agonist PAM (Scholler et al., 2017)
mGlu2/3
mGlu2/4
mGlu4 DN42 ECD Nanobody-based TR-FRET sensors (Meng et al., 2022)
mGlu2/4
mGlu4 DN45 ECD Agonist, PAM (Haubrich et al., 2021)
mGlu2/4
mGlu5 Nb43 ECD PAM (Koehl et al., 2019)
mGlu5 Nb5A ICD PAM (Eshak et al., 2024)
M2R Nb9-1, Nb9-8, Nb9-20 ICD Active (Kruse et al., 2013)
μ-OR NbE ECD Antagonist (Yu et al., 2023)
μ-OR Nb39 ICD Active (Huang et al., 2015)
κ-OR Nb6/7, Nb39 ICD Active (Che et al., 2018)
β1-AR Nb80, Nb6B9 ICD Active (Warne et al., 2019)
β2-AR Nb80 ICD Active (Rasmussen et al., 2011a)
β2-AR Nb6B9 ICD Active (Ring et al., 2013)
β2-AR Nb60, Nb61, Nb64, Nb65, ICD Active or Inactive (Staus et al., 2014)
Nb71
β2-AR Nb.b201, Nb.c200 ICD Active (McMahon et al., 2018)
Nb.c203
α1A-AR Nb29 ECD Weak PAM or neutral (Toyoda et al., 2023)

31
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

AT1R Nb.AT110, Nb.AT110i1 ICD Active (Wingler et al., 2019)


AT1R AT118, AT118i4 ECD Antagonist, inverse agonist (McMahon et al., 2020)
AT1R AT118-H, AT118-L ECD Antagonist (Skiba et al., 2024)
A2AR Nb.AD101, Nb.AD102 ICD Active (McMahon et al., 2018)
APJ JN241, JN241-9 ECD Antagonist, full agonist (Ma et al., 2020)
OX2R Sb51 ECD Active (Hong et al., 2021)
PTHR1 VHH∙PTH ECD Antibody ligand conjugation (Cheloha et al., 2020)
SMO NbSmo8 ICD Active (Deshpande et al., 2019)
CaSR Nb32 ECD NAM (Cui et al., 2024)
CaSR Nb-2D11, Nb88 ECD Inactive (Chen et al., 2021)
SUCNR1 Nanobody6 ICD NAM (Haffke et al., 2019)
5-HT2AR VGS-Nb2 ICD PAM (English et al., 2019)

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


GPR68 VGS-Nb6 ICD PAM (English et al., 2019)
GPR75 NbH3 ICD Active (Lv et al., 2022)

The list of nanobodies were compiled through a literature search, including two review articles
(Heukers et al., 2019; Wingler and Feld, 2022). This table only includes nanobodies that bind directly
to GPCR proteins, and it cites the original publications that first identified the nanobodies.
Abbreviations: α1A-AR, α1A adrenergic receptor; A2AR, A2A adenosine receptor; ACKR, atypical
chemokine receptor; APJ, apelin receptor; AT1R, angiotensin II type 1 receptor; β1-AR, β1-
adrenergic receptor; β2-AR, β2-adrenergic receptor; CaSR, Calcium Sensing Receptor; ChemR23,
Chemerin receptor 23; CXCR, C-X-C motif chemokine receptor; GPR75, G protein-coupled receptor
75; GRP68, G protein-coupled receptor 68; 5-HT2AR, serotonin 2A receptor; κ-OR, kappa opioid
receptor; M2R, M2 muscarinic acetylcholine receptor; mGlu, metabotropic glutamate receptor; μ-OR,
μ-opioid receptor; NAM, negative allosteric modulator; OX2R, orexin receptor type 2; PAM, positive
allosteric modulator; PTHR1, parathyroid hormone receptor 1; Rho, rhodopsin; SMO,
transmembrane transducer smoothened.

32
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


Figure 1
Number of PDB entries with Nbs

100

80

60 Cryo-EM
X-ray
40

20

0
2010 2015 2020 2025
Year of PDB release*
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

)LJXUH

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


A B C D E
&;&/
T4L ȕ$5
fusion

ȕ$5 ȝ25 ț25

86Nb7
fusion
Nb6B9
1E

Nb39 Nb6

1E%
Molecular Pharmacology Fast Forward. Published on August 6, 2024 as DOI: 10.1124/molpharm.124.000974
This article has not been copyedited and formatted. The final version may differ from this version.

Figure 3

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


A B C D E F
Nb43
anti-Fab Nb

1
Nb JN241 anti-Nb Fab
Nb2 Nb29
Sb
Nb Sb51
NbE

OX2R ȝ25 rod Į$5


opsin
svFv16

mGlu5

APJ/Rubredoxin Gs
fusion heterotr
heterotrimer miniGsq
Figure 4

Downloaded from molpharm.aspetjournals.org at ASPET Journals on August 10, 2024


A B C glucagon

anti-Fab Nb
glucagon
AA2AR receptor

FZD3/BRIL
miniGs
fusion
heterotrimer

Arr2/scFv30
anti-BRIL fusion
Fab

Nb35 Nb32

You might also like