You are on page 1of 12

Nanobodies: Prospects of expanding the gamut of neutralizing

antibodies against SARS-CoV-2

1 Rocktotpal Konwarh1,2,*
1
2 Department of Biotechnology, Addis Ababa Science and Technology University, Akaki Kality, P.O.
3 Box: 16417, Addis Ababa, Ethiopia
2
4 Centre of Excellence-Nanotechnology, Addis Ababa Science and Technology University, Akaki
5 Kality, P.O. Box: 16417, Addis Ababa, Ethiopia
6 * Correspondence:
7 rocktotpal.konwarh@aastu.edu.et, rock1311@gmail.com

8 Keywords: COVID-19, SARS-CoV-2, neutralizing antibody, nanobodies, spike protein

10 Number of figures: 1

11 Number of Tables: 0

12

13

14

15

16

17

18

19

20

21

22

23
Nanobodies against SARS-CoV-2

24 With more than 6.9 M confirmed cases and ~400 K deaths as on June 8, 2020 (World Health
25 Organization, 2020), COVID-19, ushered in by the SARS-CoV-2 has projected itself as a
26 microscopic-holocaust, much more sinister than those portrayed in the SciFi movies. Asymptomatic
27 transmission of the virus has been projected as the Achilles’ heel in the context of the current control
28 strategies of the pandemic (Day 2020; Gandhi et al. 2020). Reports on undiagnosed deep vein
29 thrombosis among patients, succumbing to the viral assault (Wichmann et al. 2020) and
30 demonstration of direct infection of human blood vessel and kidney organoids (Monteil et al. 2020)
31 have triggered huge hue and cry. The extreme high transmissibility of the virus, bracketed together
32 with current absence of population immunity and occurrence of stark clinical consequences projects
33 the swift advancement in effective therapeutic stratagems as the need of the hour. Needless to say,
34 researchers, across the globe, are beavering to devise appropriate diagnostic and therapeutic
35 strategies. The various nucleic acid based detection-approaches like PCR, isothermal nucleic acid
36 amplification-based methods, CRISPR/Cas platforms as well as immunoassay based point-of-care
37 lateral flow tests are marked with respective pros and cons (Bai et al. 2020; Shen et al. 2020). On the
38 other hand, strategies of inhibiting the viral fusion/entry, disrupting the replication pathway,
39 suppressing the inflammatory response, using convalescent plasma treatment and vaccine
40 development have been at the forefront of recent research (Li et al. 2020). The success lies in our
41 comprehensive understanding of the ‘biochemically and genetically guileful’ virus. At this juncture,
42 it is relevant to mention that long-term development of appropriate antibody and other protein
43 therapeutics to effectively bind and neutralize the viral infection is imperative. This would be
44 significant in case the researchers need to buy excess time to ensure befitting vaccine discovery and
45 development. Such therapeutics could possibly provide an alternative/additional way to assist those
46 people who might show unresponsiveness to vaccines (as, exemplified by many in the elderly
47 population) or do not obtain vaccine. Amidst the current hay-wired situation, the recent communiqué
48 from Israeli Defense Minister Natfali Bennet about the successful isolation of a ‘monoclonal
49 neutralizing antibody’ with potency to ‘neutralize [disease] inside carriers’ bodies’ by the scientists
50 in the Israel Institute for Biological Research has ushered in new waves of hope (Perper, 2020).

51 Prior to getting ahead, it would be prudent to recapitulate the general aspects of the lifecycle
52 of the highly pathogenic human coronaviruses (CoVs) (Jiang et al. 2020) (Figure 1 A). Talking
53 about the viral pathogenesis, the receptor binding domains (RBD) of the spike (S) glycoprotein
54 interact with the human angiotensin-converting enzyme 2 (ACE2)- the receptor that invites SARS-
55 CoV and SARS-CoV-2 into human cells (Figure 1 B (a)). The presence of a furin cleavage site at
56 interfacial zone of the S1/S2 subunits of the SARS-CoV-2 S glycoprotein demarcates the virus from
57 SARS-CoV and SARS-related CoVs (Walls et al. 2020). Precise understanding of the SARS-CoV-2
58 S ectodomain trimer is envisaged to be instrumental in developing vaccines, therapeutic antibodies
59 and diagnostics. The prospective targets of neutralizing antibodies (nAbs) against human pathogenic
60 CoVs are depicted in Figure 1 B (b). Monoclonal antibodies (mAbs), functional antigen-binding
61 fragment (Fab), single-chain variable region fragment (scFv), and single-domain antibodies
62 (nanobodies or Nbs) have been assessed against various human CoVs (Zhou et al. 2019; Zhu et al.
63 2007; Rockx et al. 2008; Sui et al. 2004; He et al. 2006; Tian et al. 2020). Jiang et al. (2020) have
64 recently reviewed the development of SARS-CoV- and MERS-CoV-specific nAbs, while literature
65 reports on nAbs against SARS-CoV-2 are comparatively scanty. Previous studies on neutralization
66 with anti-SARS-CoV-1 RBD and anti-MERS-CoV RBD antibodies had unveiled a premature
67 switching from the pre-fusion to post-fusion conformation following a closure of the receptor binding
68 site and trapping the RBD in ‘up’ conformation (Hwang et al. 2006; Walls et al. 2019; Wang et al.
69 2018). The structure of CR3022, an antibody derived from a convalescent SARS patient, in complex
70 with the RBD of the S protein at a resolution of 3.1 Å was recently reported (Yuan et al. 2020).
71 Interestingly, a cross-reactive interaction between SARS-CoV-2 and SARS-CoV was evinced by the

2
Nanobodies against SARS-CoV-2

72 elucidation that a highly conserved but cryptic, epitope, distal from the receptor binding site is
73 targeted by CR3022. However, at least two RBDs on the trimeric S protein in the ‘up’ conformation
74 and slight rotation are prerequisites to access the binding epitope by CR3022. The authors proposed
75 that albeit, the CR3022 fails to neutralize SARS-CoV-2 in vitro, the epitope could plausibly confer in
76 vivo protection. On a similar vein, researchers have resorted to the use of SARS-CoV-2 S murine
77 polyclonal antibodies for the inhibition of SARS-CoV-2 S mediated entrance into cells (Walls et al.
78 2020). The study vouched that vaccination could elicit cross-neutralizing antibodies, targeting the
79 conserved S epitopes.

80 At this juncture, the germaneness of antibody engineering may be comprehended in the


81 context of continual search for high-affinity antibodies, effective against conserved targets as well as
82 novel therapeutics with attributes like better tumor and tissue penetration and efficient launching of
83 immune effector functions (Saeed et al. 2017). Particularly, in the context of antitumor therapeutics,
84 Bannas et al. (2017) had raised concerns about the large-size (150 kDa) dictated practical snag of in
85 vivo delivery of conventional antibodies to tumor cells. On the other hand, aggregation and/or
86 mispairing of V-domains due to lower stability and solubility of engineered antibodies- a
87 consequence of intrinsic hydrophobic interactions of VH and VL domains (that constitute the
88 antigen binding fragment (Fab) of IgG antibodies) have been another pertinent issue. As plausible
89 solutions, nanobodies (15 kDa) and nanobody based human heavy chain antibodies (75kDa) (Bannas
90 et al., 2017) have instigated considerable research impetus. Besides conventional antibodies,
91 camelids produce heavy-chain-only antibodies (HCAbs) with a single variable domain as the target
92 recognition module (Hamers-Casterman et al. 1993; Muyldermans et al. 2013). This single variable
93 domain without an effector domain functions as a single-domain antibody, VHH, or nanobody (Nb)
94 (Figure 1 C). Although the prospects of using nanobodies as research and diagnostic tools have been
95 critically and comprehensively assessed (De Meyer et al. 2014; Steeland et al. 2016) and a plethora
96 of nanobodies are currently being placed under pre-clinical or clinical assessments for various
97 diseases like brain tumours, inflammation, lung diseases, as well as autoimmune diseases, paralleling
98 the performance of classical antibodies with nanobodies for therapeutic applications could be bit
99 fiddly (Jovčevska and Muyldermans, 2019). Nevertheless, studies have attested the advantages of
100 nanobodies in contrast to conventional antibodies with respect to the former’s smaller size,
101 amenability for processing into multiple formats, desirable thermal and chemical stability, high
102 solubility, commendable in vivo tissue penetration and targeting, lower susceptibility to steric
103 hindrances (that may otherwise obstruct optimal binding) as well as ability to display antigenic
104 affinity and specificity at par with conventional antibodies (Bannas et al. 2017; De Vlieger et al.
105 2018; Govaert et al. 2012; Laursen et al. 2018; Fernandes, 2019; Wrapp et al. 2020). Prospects of
106 genetically linking to Fc-domains, peptide tags, or other nanobodies as well as site-specific chemical
107 fusion with nanoscale materials, radionuclides, photosensitizers, etc. widen the spectrum of their
108 applications. Furthermore, the expedient attributes of nanobodies and human Fc domains may be
109 combined in chimeric nanobody-heavy chain antibodies, half the size of the conventional antibodies,
110 as mentioned before (Bannas et al. 2017).

111 Post perusal of the afore-stated, harnessing VHHs as therapeutics against various viral
112 infectious agents seems to be an interesting proposition (Sroga et al. 2020). In this respect, use of
113 VHH against dengue virus (Fatima et al. 2014); hepatitis C virus (Tarr et al. 2013); multiple VHH
114 monovalent candidates against poliovirus (Thys et al. 2010) and norovirus (Koromyslova
115 and Hansman, 2017); anti-CXCR4 monovalent and bivalent (Jähnichen et al. 2010) as well as anti-
116 p24 monovalent and bivalent (Gray et al. 2017) nanobodies against HIV; VHH bivalent/albumin-
117 linked nanobody against rabies virus (Terryn et al. 2014) and anti-VP6 VHH as an effective
118 prophylactic treatment against rotavirus A-associated diarrhea (Maffey et al. 2016) have been

3
Nanobodies against SARS-CoV-2

119 documented. Investigations on the application of nanobodies against respiratory pathogens has also
120 gained pace in recent years. Use of H5N1-HA bivalent nanobody against influenza virus (Ibanez et
121 al. 2011), as well as the application of multi-domain antibody MD3606 (generated using diverse
122 camelid single-domain antibodies to influenza virus hemagglutinin) to protect mice against influenza
123 A and B infection post intravenous administration or expression using recombinant adeno-associated
124 vector (Laursen et al. 2018), merit special mention. Similarly, two llama-derived single-domain
125 antibodies with human respiratory syncytial virus (RSV) - neutralizing action have been reported to
126 selectively bind to RSV fusion protein (F) in its pre-fusion state with picomolar affinity (Rossey et
127 al. 2017). Delivering a trimeric nanobody, ALX-0171 (that interacted with antigenic site II of RSV F
128 protein at subnanomolar affinity), prophylactically or therapeutically directly to lungs of cotton rats
129 was effective in down-scaling both nasal and lung RSV titers (Detalle et al. 2015). Stalin Raj et al.
130 (2018) had resorted to direct cloning and expression of VHHs of HCAbs from the bone marrow of
131 MERS-CoV–infected Arabian camels and identified several MERS-CoV–specific VHHs or
132 nanobodies. With a prolonged half-life in serum, camel/human chimeric HCAbs were efficacious in
133 endowing protection to mice against MERS-CoV challenge. In a similar vein, the efficacy to target
134 MERS-CoV S RBD using novel neutralizing Nb (NbMS10) and its human-Fc-fused version
135 (NbMS10-Fc) has been documented (Zhao et al. 2018). Remarkably, the Nbs were able to cross-
136 neutralize infections caused by diverse MERS-CoV strains isolated from humans and camels. The
137 Fc-tagged Nb was able to confer complete protection of humanized mice from lethal MERS-CoV
138 assault.

139 A concerted effort of biologist Michael Rout and chemist Brian Chait has been directed
140 towards selecting high affinity and effective neutralizing nanobodies, interacting with the various
141 non-overlapping target-epitopes of SARS-CoV-2 S (Rockefeller University, 2020). The researchers
142 envisage to set-up the appropriate nanobodies as increased level multimers to augment affinity and
143 eventually tune them at the molecular level to better their neutralizing potency. Similarly, researchers
144 from Protein Production UK, a project hosted by the Rosalind Franklin Institute in association with
145 Diamond Light Source, UK, have made nanobodies (exhibiting high affinity to the S protein of the
146 SARS-CoV-2), available to scientist at the University of Oxford for deeper delving into the structure
147 of the virus (Rosalind Franklin Institute, 2020). On a stimulating note, scientists from the University
148 of Texas (UT) at Austin, the National Institutes of Health and Ghent University in Belgium have
149 documented the isolation of two potently neutralizing VHHs, targeting the SARS-CoV-1 and MERS-
150 CoV RBDs, respectively (Wrapp et al. 2020). Wrapp et al. (2020) had resorted to sequential
151 immunization of a llama subcutaneously multiple times with SARS-CoV-1 S and MERS-CoV S
152 protein. Two sequential rounds of panning were executed by phage display using either SARS-CoV-
153 1 S or MERS-CoV S proteins to procure VHHs directed against the S proteins. The researchers
154 successfully isolated seven unique MERS-CoV S and five SARS-CoV-1 S specific VHHs post-
155 sequencing of the positive clones, multiple sequence alignment, and phylogenetic analysis. Following
156 expression in Pichia pastoris and purification from yeast medium, the interaction of the purified
157 VHHs with the perfusion-stabilized MERS-CoV S and SARS-CoV-1 S was attested by ELISA.
158 Pertinently, the SARS-CoV-1 RBD-directed VHH could cross-react with the SARS-CoV-2 RBD. A
159 fascinating dimension to the work was the neutralization of the SARS-CoV-2 S pseudotyped viruses
160 by the cross reactive VHH, engineered as a bivalent human IgG Fc-fusion. The plausible scaled up
161 production of the VHH-Fc fusion was attested in a commercial-standard CHO cell system. The
162 MERS VHH-55, SARS VHH-72 and VHH-72-Fc, exhibiting desirable biophysical attributes and
163 potent neutralization potency, could be prospective therapeutic candidates. However, appropriate in
164 vivo experimentations as part of preclinical studies are prerequisite.

4
Nanobodies against SARS-CoV-2

165 Retrieval of information from the preprint at BioRxiv evinces the successful endeavors of
166 Swiss researchers Walter et al. (2020) in identifying 63 unique anti-RBD synthetic nanobodies or
167 sybodies, interacting in the context of the full-length SARS-CoV-2 spike ectodomain. Assisted by a
168 prompt in vitro selection platform (encompassing ribosome and phage display), the task of selecting
169 the sybodies was accomplished within twelve days. Six of the selected sybodies displayed double-
170 digit nanomolar binding affinity with the viral spike while five of them could inhibit RBD interaction
171 with ACE2. Furthermore, the researchers identified a pair of anti-RBD sybodies that could
172 concomitantly interact with the RBD. It would be interesting to peruse the outcomes of the authors’
173 previously reported NestLink technology (Egloff et al. 2019) based delving of the selection pools to
174 unearth unique sybodies with little off-rates and capacity to identify rare epitopes. The authors are
175 upbeat about plausible therapeutic exploitation of the sybodies for the development of an inhalable
176 drug as useful prophylaxis against COVID-19.

177 To speak about yet another development, Beroni Group (an international biopharmaceutical
178 enterprise) in concert with Tianjin University in China has recently identified 24 types of nanobodies
179 (post-screening a library with one billion-plus nanobody sequences) for prompt detection and
180 treatment of SARS-CoV-2 (Beroni Group, 2020). Eight of them are directed against the S protein
181 while sixteen of them target the nucleocapsid (N) protein- the latter could find application as a
182 marker in diagnostic assays. Based on approaches of structural biology, computational biology, and
183 protein engineering, the researchers are gearing up to optimize the properties of the nanobodies
184 besides endeavoring to reduce their immunogenicity and augment the therapeutic efficiency by
185 humanizing them. By the same token, researchers from Fudan University and Biomissile
186 Corporation, China have directed their endeavors towards the development of a phage-displayed
187 single-domain antibody library based on embedding naive complementarity-determining regions
188 (CDRs) into framework sites of a human germline immunoglobulin heavy chain variable region
189 (IGHV) allele (Wu et al. 2020). Their study, encompassing the library-biopanning against SARS-
190 CoV-2 RBD and S1 subunit led to the revelation of fully human single-domain antibodies, displaying
191 low-nanomolar/subnanomolar range affinities towards five distinct epitopes on SARS-CoV-2 RBD
192 (Figure 1 D). Amongst the groups of A, B, C, D, and E neutralizing antibodies, the group D
193 members, n3088 and n3130 could target a ‘cryptic’ epitope, positioned in the spike trimeric interface,
194 resulting in effective neutralization of SARS-CoV-2. The researchers are buoyant about the apt
195 application of these, either alone or in synergy with other SARS-CoV-2 neutralizing antibodies,
196 especially the ACE2-competing neutralizing antibodies. They may also be employed as integrant for
197 creating bispecific or multispecific antibodies (Wu et al. 2020). Previously, He et al. (2019) had
198 demonstrated an augmented efficacy of oligomeric nanobodies, relative to monomeric nanobodies
199 against MERS coronavirus RBD. Investigating the potential of such oligomeric nanobodies in the
200 case of SARS-CoV-2 would be attention-grabbing.

201 These studies spark obvious anticipations and hopes for the potential application of
202 nanobodies against COVID-19. The attributes of small size (almost one-fourth of the size of human
203 antibodies) and simple structure, ease and comparatively lower cost, low immunogenicity and ability
204 to display high affinity have endowed them with a special niche in the realm of therapeutics and rapid
205 point-of-care diagnostics. Nanobodies seem to be quite efficient in trapping and stabilizing
206 conformation-switchable targets in specific conformations, facilitating greater insight into
207 biomolecular mechanisms and interactions. This could be of immense relevance to mine information
208 on SARS-CoV-2 pathogenesis. Most importantly, highly stable VHHs could be nebulized and
209 exploited for the development of inhalable prophylactic formulations, thereby ensuring straight
210 delivery to the lungs- the combat zone. Another merit lies in the plausibility of stockpiling the VHHs
211 without trade-off in their stability even after extended storages and using them as therapeutic choices

5
Nanobodies against SARS-CoV-2

212 in case of disasters like COVID-19. To conclude, I do hope that the incessant and concerted research
213 endeavors would surely pave the way to a safer world, liberated from the grasp of SARS-CoV-2 and
214 akin.

215 Conflict of Interest

216 The author declares the absence of any commercial or financial relationships that could be construed
217 as a potential conflict of interest.

218 Author’s Contribution

219 RK reviewed the literature, critically analyzed it and authored the article.

220 Funding

221 RK did not receive any specific grant from funding agencies in the public, commercial or not-for-
222 profit sectors to compile and submit this article.

223 Acknowledgments

224 A bouquet of gratitude is extended to all the researchers and frontline warriors, engaged in the battle
225 against COVID-19.

226

227 References

228 Bai, H., Cai, X., Zhang, X. (2020). Landscape coronavirus disease 2019 test (COVID-19 test) in vitro- A
229 comparison of PCR vs Immunoassay vs CRISPR-based test.
230 https://doi.org/10.31219/osf.io/6eagn
231 Bannas, P., Hambach, J., Koch-Nolte, F. (2017). Nanobodies and nanobody-based human heavy chain
232 antibodies as antitumor therapeutics. Front. Immunol 8, 1603.
233 Beroni Group (2020). Beroni Group and Tianjin University have made a significant discovery in
234 developing a medical solution using nanobody technology for the novel coronavirus.
235 https://www.beronigroup.com/2020/05/19/beroni-group-and-tianjin-university-have-made-
236 a-significant-discovery-in-developing-a-medical-solution-using-nanobody-technology-for-the-
237 novel-coronavirus/ [Accessed on June 3, 2020]
238 Day, M. (2020). COVID-19: four fifths of cases are asymptomatic, China figures indicate. BMJ 369,
239 m1375. https://doi.org/10.1136/bmj.m1375
240 De Meyer, T., Muyldermans, S., Depicker, A. (2014). Nanobody-based products as research and
241 diagnostic tools. Trends Biotechnol. 32, 263-270.
242 De Vlieger, D., Ballegeer, M., Rossey, I., Schepens, B., Saelens, X. (2019). Single-domain antibodies and
243 their formatting to combat viral infections. Antibodies 8(1), 1.
244 Detalle, L., Stohr, T., Palomo, C., Piedra, P.A., Gilbert, B.E., Mas, V., et al. (2016). Generation and
245 characterization of ALX-0171, a potent novel therapeutic nanobody for the treatment of
246 respiratory syncytial virus infection. Antimicrob. Agents Chemother. 60, 6-13.

6
Nanobodies against SARS-CoV-2

247 Egloff, P., Zimmermann, I., Arnold, F.M., Hutter, C.A., Morger, D., Opitz, L., et al. (2019). Engineered
248 peptide barcodes for in-depth analyses of binding protein libraries. Nat. Methods 16(5), 421-
249 428.
250 Fatima, A., Wang, H., Kang, K., Xia, L., Wang, Y., Ye, W., et al. (2014). Development of VHH antibodies
251 against dengue virus type 2 NS1 and comparison with monoclonal antibodies for use in
252 immunological diagnosis. PloS One, 9(4). 1–12
253 Fernandes, J.C. (2018). Therapeutic application of antibody fragments in autoimmune diseases: Current
254 state and prospects. Drug Discov. Today, 23(12), 1996-2002.
255 Gandhi, M., Yokoe, D.S., Havlir, D.V. (2020). Asymptomatic transmission, the Achilles’ heel of current
256 strategies to control COVID-19. N. Engl. J. Med. 382, 2158-2160
257 Govaert, J., Pellis, M., Deschacht, N., Vincke, C., Conrath, K., Muyldermans, S., et al. (2012). Dual beneficial
258 effect of interloop disulfide bond for single domain antibody fragments. J. Biol. Chem. 287(3),
259 1970-1979.
260 Gray, E.R., Brookes, J.C., Caillat, C., Turbé, V., Webb, B.L., Granger, L.A., et al. (2017). Unravelling the
261 molecular basis of high affinity nanobodies against HIV p24: in vitro functional, structural, and
262 in silico insights. ACS Infect. Dis. 3(7), 479-491.
263 Hamers-Casterman, C.T.S.G., Atarhouch, T., Muyldermans, S., Robinson, G., Hammers, C., Songa, E.B., et al.
264 (1993). Naturally occurring antibodies devoid of light chains. Nature 363(6428), 446-448.
265 He, L., Tai, W., Li, J., Chen, Y., Gao, Y., Li, J., Sun, S., Zhou, Y., Du, L., Zhao, G. (2019). Enhanced ability of
266 oligomeric nanobodies targeting MERS coronavirus receptor-binding domain. Viruses 11(2),
267 166.
268 He, Y., Li, J., Li, W., Lustigman, S., Farzan, M., Jiang, S. (2006). Cross-neutralization of human and palm
269 civet severe acute respiratory syndrome coronaviruses by antibodies targeting the receptor-
270 binding domain of spike protein. J. Immunol. 176(10), 6085-6092.
271 Hwang, W.C., Lin, Y., Santelli, E., Sui, J., Jaroszewski, L., Stec, B. et al. (2006). Structural basis of
272 neutralization by a human anti-severe acute respiratory syndrome spike protein antibody,
273 80R. J. Biol. Chem. 281(45), 34610-34616.
274 Jähnichen, S., Blanchetot, C., Maussang, D., Gonzalez-Pajuelo, M., Chow, K.Y., Bosch, L., et al. (2010).
275 CXCR4 nanobodies (VHH-based single variable domains) potently inhibit chemotaxis and HIV-1
276 replication and mobilize stem cells. Proc. Natl. Acad. Sci. U.S.A. 107(47), 20565-20570.
277 Jiang, S., Hillyer, C., Du, L. (2020). Neutralizing antibodies against SARS-CoV-2 and other human
278 coronaviruses. Trends Immunol. 41, 355-359. https://doi.org/10.1016/j.it.2020.03.007
279 Jovčevska, I., Muyldermans, S. (2019). The Therapeutic Potential of Nanobodies. BioDrugs 34, 11-16.
280 Koromyslova, A.D., Hansman, G.S. (2017). Nanobodies targeting norovirus capsid reveal functional
281 epitopes and potential mechanisms of neutralization. PLoS Pathog. 13(11), e1006636
282 Laursen, N.S., Friesen, R.H., Zhu, X., Jongeneelen, M., Blokland, S., Vermond, J., et al. (2018). Universal
283 protection against influenza infection by a multidomain antibody to influenza
284 hemagglutinin. Science 362(6414), 598-602.
285 Li, H., Zhou, Y., Zhang, M., Wang, H., Zhao, Q., Liu, J. (2020). Updated approaches against SARS-CoV-2.
286 Antimicrob. Agents Chemother. doi:10.1128/AAC.00483-20
287 Maffey, L., Vega, C.G., Miño, S., Garaicoechea, L., Parreño, V., 2016. Anti-VP6 VHH: an experimental
288 treatment for rotavirus A-associated disease. PloS One 11(9), e0162351

7
Nanobodies against SARS-CoV-2

289 Monteil, V., Kwon, H., Prado, P., Hagelkrüys, A., Wimmer, R.A., Stahl, M., et al. (2020). Inhibition of SARS-
290 CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell
291 https://doi.org/10.1016/j.cell.2020.04.004
292 Muyldermans, S. (2013). Nanobodies: natural single-domain antibodies. Annu. Rev. Biochem. 82, 775-
293 797.
294 Perper, R. (2020). Israel's government research agency says it successfully isolated a key coronavirus
295 antibody, paving the way for a possible breakthrough treatment.
296 https://www.pulselive.co.ke/bi/politics/israels-government-research-agency-says-it-
297 successfully-isolated-a-key-coronavirus/fyfyz3q [Accessed on 8 May, 2020]
298 Rockefeller University (2020) Research Program on COVID-19/SARS-CoV-2.
299 https://www.rockefeller.edu/coronavirus/research-program-covid-19-sars-cov-2/ [Accessed
300 on May 8, 2020]
301 Rockx, B., Corti, D., Donaldson, E., Sheahan, T., Stadler, K., Lanzavecchia, A. et al. (2008). Structural basis
302 for potent cross-neutralizing human monoclonal antibody protection against lethal human and
303 zoonotic severe acute respiratory syndrome coronavirus challenge. J. Virol. 82(7), 3220-3235.
304 Rossey, I., Gilman, M.S., Kabeche, S.C., Sedeyn, K., Wrapp, D., Kanekiyo, M., et al. (2017). Potent single-
305 domain antibodies that arrest respiratory syncytial virus fusion protein in its prefusion state.
306 Nat. Commun. 8, 14158. doi: 10.1038/ncomms14158
307 Saeed, A.F., Wang, R., Ling, S., Wang, S. (2017). Antibody engineering for pursuing a healthier
308 future. Front. Microbiol. 8, 495.
309 Shen, M., Zhou, Y., Ye, J., AL-maskri, A.A.A., Kang, Y., Zeng, S., Cai, S. (2020). Recent advances and
310 perspectives of nucleic acid detection for coronavirus. J. Pharm. Anal. 10, 97-101.
311 https://doi.org/10.1016/j.jpha.2020.02.010
312 Sroga, P., Safronetz, D., Stein, D.R. (2020). Nanobodies: a new approach for the diagnosis and treatment
313 of viral infectious diseases. Future Virol. 15, 195-205.
314 Stalin Raj, V., Okba, N.M., Gutierrez-Alvarez, J., Drabek, D., van Dieren, B., Widagdo, W., et al. (2018).
315 Chimeric camel/human heavy-chain antibodies protect against MERS-CoV infection. Sci. Adv.
316 4(8), p.eaas9667.
317 Steeland, S., Vandenbroucke, R.E. and Libert, C. (2016). Nanobodies as therapeutics: big opportunities
318 for small antibodies. Drug Discov. Today 21(7), 1076-1113.
319 Sui, J., Li, W., Murakami, A., Tamin, A., Matthews, L.J., Wong, S.K. et al. (2004) Potent neutralization of
320 severe acute respiratory syndrome (SARS) coronavirus by a human mAb to S1 protein that
321 blocks receptor association. PNAS 101(8), 2536-2541.
322 Tarr, A.W., Lafaye, P., Meredith, L., Damier‐Piolle, L., Urbanowicz, R.A., Meola, A., et al. (2013). An alpaca
323 nanobody inhibits hepatitis C virus entry and cell‐to‐cell transmission. Hepatol. 58(3), pp.932-
324 939.
325 Terryn, S., Francart, A., Rommelaere, H., Stortelers, C., Van Gucht, S. (2016). Post-exposure treatment
326 with anti-rabies VHH and vaccine significantly improves protection of mice from lethal rabies
327 infection. PLoS Neglect. Trop. D. 10(8), e0004902
328 The Rosalind Franklin Institute. (2020). Science minister hails nanobody breakthrough at Franklin.
329 https://www.rfi.ac.uk/science-minister-hails-nanobody-breakthrough/ [Accessed on May 8,
330 2020]
331 Thys, B., Schotte, L., Muyldermans, S., Wernery, U., Hassanzadeh-Ghassabeh, G., Rombaut, B. (2010). In
332 vitro antiviral activity of single domain antibody fragments against poliovirus. Antivir.
333 Res. 87(2), 257-264.

8
Nanobodies against SARS-CoV-2

334 Tian, X., Li, C., Huang, A., Xia, S., Lu, S., Shi, Z. (2020). Potent binding of 2019 novel coronavirus spike
335 protein by a SARS coronavirus-specific human monoclonal
336 antibody. Emerg. Microbes Infect. 9(1), 382-385.
337 Walls, A.C., Park, Y.J., Tortorici, M.A., Wall, A., McGuire, A.T., Veesler, D. (2020). Structure, function, and
338 antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 181, 281-292.e6
339 Walls, A.C., Xiong, X., Park, Y.J., Tortorici, M.A., Snijder, J., Quispe, J., et al. (2019). Unexpected receptor
340 functional mimicry elucidates activation of coronavirus fusion. Cell 176(5), 1026-1039.
341 Walter, J.D., Hutter, C.A., Zimmermann, I., Earp, J., Egloff, P., Sorgenfrei, M., et al. (2020). Synthetic
342 nanobodies targeting the SARS-CoV-2 receptor-binding domain. Preprint at BioRxiv. doi:
343 https://doi.org/10.1101/2020.04.16.045419
344 Wang, L., Shi, W., Chappell, J.D., Joyce, M.G., Zhang, Y., Kanekiyo, M., et al. (2018). Importance of
345 neutralizing monoclonal antibodies targeting multiple antigenic sites on the Middle East
346 respiratory syndrome coronavirus spike glycoprotein to avoid neutralization escape. J.
347 Virol. 92(10), e02002-17.
348 Wichmann, D., Sperhake, J.P., Lütgehetmann, M., Steurer, S., Edler, C., Heinemann, A., et al. (2020).
349 Autopsy findings and venous thromboembolism in patients with COVID-19: A prospective
350 cohort study. Ann. Intern. Med. Doi: 10.7326/M20-2003.
351 World Health Organization. (2020). WHO Coronavirus Disease (COVID-19) Dashboard.
352 https://covid19.who.int/?gclid=Cj0KCQjww_f2BRC-
353 ARIsAP3zarEfohyPnXxXij4jnp412SSC_7kKj9J3pEly4tpE21VVLydZtb3KaZsaAlH-
354 EALw_wcB [Accessed on June 9, 2020]
355 Wrapp, D., De Vlieger, D., Corbett, K.S., Torres, G.M., Wang, N., Van Breedam, W., et al. (2020). Structural
356 basis for potent neutralization of betacoronaviruses by single-domain camelid antibodies. Cell
357 181, 1–12
358 Wu, Y., Li, C., Xia, S., Tian, X., Kong, Y., Wang, Z., et al. (2020) Identification of human single-domain
359 antibodies against SARS-CoV-2. Cell Host Microbe https://doi.org/10.1016/j.chom.2020.04.023
360 Yuan, M., Wu, N.C., Zhu, X., Lee, C.C.D., So, R.T., Lv, H., et al. (2020). A highly conserved cryptic epitope in
361 the receptor-binding domains of SARS-CoV-2 and SARS-CoV. Science 368, 630-633.
362 Zhao, G., He, L., Sun, S., Qiu, H., Tai, W., Chen, J., et al. (2018). A novel nanobody targeting Middle East
363 respiratory syndrome coronavirus (MERS-CoV) receptor-binding domain has potent cross-
364 neutralizing activity and protective efficacy against MERS-CoV. J. Virol. 92(18), e00837-18.
365 Zhou, Y., Yang, Y., Huang, J., Jiang, S. Du, L. (2019). Advances in MERS-CoV vaccines and therapeutics
366 based on the receptor-binding domain. Viruses 11(1), 60.
367 Zhu, Z., Chakraborti, S., He, Y., Roberts, A., Sheahan, T., Xiao, X., et al. (2007) Potent cross-reactive
368 neutralization of SARS coronavirus isolates by human monoclonal antibodies. PNAS 104(29),
369 12123-12128.
370

371

372

373

374

9
Nanobodies against SARS-CoV-2

375 Figure Captions

376 Figure 1. Life cycle of highly pathogenic human coronaviruses (CoVs) and specific neutralizing
377 antibodies (nAbs) against these coronaviruses.

378 (A) Life cycle of highly pathogenic human CoVs. These CoVs enter host cells by first binding to
379 their respective cellular receptors [angiotensin-converting enzyme 2 (ACE2) for severe acute
380 respiratory syndrome (SARS)-CoV-2 or SARS-CoV and dipeptidyl peptidase 4 (DPP4) for Middle
381 East respiratory syndrome (MERS)-CoV] on the membranes of host cells expressing ACE2 (e.g.,
382 pneumocytes, enterocytes) or DPP4 (e.g., liver or lung cells including Huh-7, MRC-5, and Calu-3)
383 via the surface spike (S) protein, which mediates virus–cell membrane fusion and viral entry. Viral
384 genomic RNA is released and translated into viral polymerase proteins. The negative (−)-sense
385 genomic RNA is synthesized and used as a template to form sub-genomic or genomic positive (+)-
386 sense RNA. Viral RNA and nucleocapsid (N) structural protein are replicated, transcribed, or
387 synthesized in the cytoplasm, whereas other viral structural proteins, including S, membrane (M),
388 and envelope (E), are transcribed then translated in the endoplasmic reticulum (ER) and transported
389 to the Golgi. The viral RNA-N complex and S, M, and E proteins are further assembled in the ER–
390 Golgi intermediate compartment (ERGIC) to form a mature virion, then released from host cells.

391 (B) Potential targets of nAbs against SARS-CoV-2 and other pathogenic human CoVs.

392 (a) Human CoV receptor binding and membrane fusion process. The CoV first binds a viral
393 receptor (ACE2 or DPP4) through the receptor-binding domain (RBD) in the S protein, followed by
394 fusion of the virus with cell membranes via the formation of a six-helix bundle (6-HB) fusion core.
395 NTD, N-terminal domain.

396 (b) Potential targets of nAbs on the S protein of human CoVs. Monoclonal antibody (mAb),
397 antigen-binding fragment (Fab), single-chain variable region fragment (scFv), or single-domain
398 antibody [nanobody (Nb) or VHH derived from camelid heavy chain antibody (HcAb)] binds to the
399 RBD, S1 subunit (non-RBD, including NTD), or S2 of the viral S protein, blocking binding between
400 the RBD and the respective receptor (for RBD-targeting nAbs), interfering with the conformational
401 change of S (for S1-targeting nAbs), or hindering S2-mediated membrane fusion (for S2-targeting
402 nAbs), leading to the inhibition of infection with pathogenic human CoVs in the host cells. The
403 figure was created using BioRender (https://biorender.com/).

404 (Reproduced from Jiang et al. 2020, under the provisions of Creative Commons License, CC
405 BY 4.0, Copyright © 2020 The Author(s). Published by Elsevier Ltd.)

406 (C) Advantageous features of camelid heavy chain antibodies. Heavy chain antibodies are composed
407 of two heavy chains. The target-binding module is composed of a single VHH domain. A
408 recombinant VHH domain, designated nanobody (Nb) is highly soluble and does not show any
409 tendency to associate with other hydrophobic protein surfaces. Conventional antibodies are
410 composed of two heavy and two light chains. The target-binding module is composed of two non-
411 covalently associated variable domains VH and VL. In intact antibodies, the proper orientation of
412 these domains is mediated by a hydrophobic interface and is further stabilized by the disulfide-linked
413 CL and CH1 domains. A pair of VH and VL domains can be linked genetically into a single-chain
414 variable fragment (scFv) in which the proper orientation of domains is mediated alone by the
415 hydrophobic interface between the two V-domains.

10
Nanobodies against SARS-CoV-2

416 (Reproduced from Bannas et al., 2017, under the provisions of Creative Commons Attribution
417 License (CC BY). Copyright © 2017 Bannas, Hambach and Koch-Nolte)

418 (D) Targeting of diverse epitopes within the SARS-CoV-2 spike protein receptor binding domain
419 (RBD) by human single-domain antibodies, potential therapeutic candidates for COVID-19.

420 (Reproduced from Wu et al. (2020). Copyright ©2020 Elsevier Inc., based on the reuse-provisions of
421 Elsevier’s COVID-19 Resource Centre)

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

11
Nanobodies against SARS-CoV-2

444
445
12

You might also like