Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Rapid generation of potent antibodies by autonomous hypermutation in yeast

Abstract

The predominant approach for antibody generation remains animal immunization, which can yield exceptionally selective and potent antibody clones owing to the powerful evolutionary process of somatic hypermutation. However, animal immunization is inherently slow, not always accessible and poorly compatible with many antigens. Here, we describe ‘autonomous hypermutation yeast surface display’ (AHEAD), a synthetic recombinant antibody generation technology that imitates somatic hypermutation inside engineered yeast. By encoding antibody fragments on an error-prone orthogonal DNA replication system, surface-displayed antibody repertoires continuously mutate through simple cycles of yeast culturing and enrichment for antigen binding to produce high-affinity clones in as little as two weeks. We applied AHEAD to generate potent nanobodies against the SARS-CoV-2 S glycoprotein, a G-protein-coupled receptor and other targets, offering a template for streamlined antibody generation at large.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Autonomous hypermutation yeast surface display (AHEAD).
Fig. 2: Evolution of anti-AT1R nanobodies.
Fig. 3: Evolution of anti-SARS-CoV-2 nanobodies and activities of potent anti-SARS-CoV-2 nanobodies.
Fig. 4: Epitope mapping using deep mutational scanning libraries of RBD.

Similar content being viewed by others

Data availability

All data generated for the present study are available upon request to the corresponding authors. pAW240 and its sequence are available at Addgene (plasmid 170791). NGS data are available at NCBI’s SRA website https://www.ncbi.nlm.nih.gov/sra?term=SRP320370 (identifier biosample accession numbers SAMN19242322, SAMN19242323, SAMN19242324, SAMN19242325, SAMN19242326, SAMN19242327 and SAMN19242328).

References

  1. Lu, R. M. et al. Development of therapeutic antibodies for the treatment of diseases. J. Biomed. Sci. 27, 1 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gravbrot et al. Therapeutic monoclonal antibodies targeting immune checkpoints for the treatment of solid tumors. Antibodies 8, 51 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  3. Czajka, T. F., Vance, D. J. & Mantis, N. J. Slaying SARS-CoV-2 one (single-domain) antibody at a time. Trends Microbiol. 29, 195–203 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Byrne, B., Stack, E., Gilmartin, N. & O’Kennedy, R. Antibody-based sensors: principles, problems and potential for detection of pathogens and associated toxins. Sensors 9, 4407–4445 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yao, H. et al. Patient-derived SARS-CoV-2 mutations impact viral replication dynamics and infectivity in vitro and with clinical implications in vivo. Cell Discov. 6, 76 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hanke, L. et al. An alpaca nanobody neutralizes SARS-CoV-2 by blocking receptor interaction. Nat. Commun. 11, 4420 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Schoof, M. et al. An ultrapotent synthetic nanobody neutralizes SARS-CoV-2 by stabilizing inactive spike. Science 370, 1473–1479 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gray, A. et al. Animal-free alternatives and the antibody iceberg. Nat. Biotechnol. 38, 1234–1239 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Rajewsky, K. Clonal selection and learning in the antibody system. Nature 381, 751–758 (1996).

    Article  CAS  PubMed  Google Scholar 

  10. Mishra, A. K. & Mariuzza, R. A. Insights into the structural basis of antibody affinity maturation from next-generation sequencing. Front. Immunol. 9, 117 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Teng, G. & Papavasiliou, F. N. Immunoglobulin somatic hypermutation. Annu. Rev. Genet. 41, 107–120 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Boder, E. T., Raeeszadeh-Sarmazdeh, M. & Price, J. V. Engineering antibodies by yeast display. Arch. Biochem. Biophys. 526, 99–106 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Almagro, J. C., Pedraza-Escalona, M., Arrieta, H. I. & Pérez-Tapia, S. M. Phage display libraries for antibody therapeutic discovery and development. Antibodies 8, 44 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  14. Baker, M. Reproducibility crisis: blame it on the antibodies. Nature 521, 274–276 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Voskuil, J. L. A. The challenges with the validation of research antibodies. F1000Res. 6, 161 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Ravikumar, A., Arrieta, A. & Liu, C. C. An orthogonal DNA replication system in yeast. Nat. Chem. Biol. 10, 175–177 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Ravikumar, A., Arzumanyan, G. A., Obadi, M. K. A., Javanpour, A. A. & Liu, C. C. Scalable, continuous evolution of genes at mutation rates above genomic error thresholds. Cell 175, 1946–1957 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Boder, E. T. & Wittrup, K. D. Yeast surface display for screening combinatorial polypeptide libraries. Nat. Biotechnol. 15, 553–557 (1997).

    Article  CAS  PubMed  Google Scholar 

  19. Wingler, L. M., McMahon, C., Staus, D. P., Lefkowitz, R. J. & Kruse, A. C. Distinctive activation mechanism for angiotensin receptor revealed by a synthetic nanobody. Cell 176, 479–490 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Neuberger, M. Antibodies: a paradigm for the evolution of molecular recognition. Biochem. Soc. Trans. 30, 341–350 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Muyldermans, S. Nanobodies: natural single-domain antibodies. Annu. Rev. Biochem. 82, 775–797 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Zavrtanik, U., Lukan, J., Loris, R., Lah, J. & Hadži, S. Structural basis of epitope recognition by heavy-chain camelid antibodies. J. Mol. Biol. 430, 4369–4386 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Manglik, A., Kobilka, B. K. & Steyaert, J. Nanobodies to study G protein-coupled receptor structure and function. Annu. Rev. Pharmacol. Toxicol. 57, 19–37 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Gray, A. C., Sidhu, S. S., Chandrasekera, P. C., Hendriksen, C. F. M. & Borrebaeck, C. A. K. Animal-based antibodies: obsolete. Science 353, 452–453 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Wingler, L. M. et al. Angiotensin and biased analogs induce structurally distinct active conformations within a GPCR. Science 367, 888–892 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, Z., Mathias, A., Stavrou, S. & Neville, D. M. A new yeast display vector permitting free scFv amino termini can augment ligand binding affinities. Protein Eng. Des. Sel. 18, 337–343 (2005).

    Article  PubMed  CAS  Google Scholar 

  27. Rakestraw, J. A., Sazinsky, S. L., Piatesi, A., Antipov, E. & Wittrup, K. D. Directed evolution of a secretory leader for the improved expression of heterologous proteins and full-length antibodies in Saccharomyces cerevisiae. Biotechnol. Bioeng. 103, 1192–1201 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhong, Z., Ravikumar, A. & Liu, C. C. Tunable expression systems for orthogonal DNA replication. ACS Synth. Biol. 7, 2930–2934 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Makrides, S. C. et al. Extended in vivo half-life of human soluble complement receptor type 1 fused to a serum albumin-binding receptor. J. Pharmacol. Exp. Ther. 277, 534–542 (1996).

    CAS  PubMed  Google Scholar 

  30. Renier, N. et al. IDISCO: a simple, rapid method to immunolabel large tissue samples for volume imaging. Cell 159, 896–910 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Chung, K. et al. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. McMahon, C. et al. Yeast surface display platform for rapid discovery of conformationally selective nanobodies. Nat. Struct. Mol. Biol. 25, 289–296 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fridy, P. C. et al. A robust pipeline for rapid production of versatile nanobody repertoires. Nat. Methods 11, 1253–1260 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yan, R. et al. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 367, 1444–1448 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cohen, J. ‘Provocative results’ boost hopes of antibody treatment for COVID-19. Science https://doi.org/10.1126/science.abf0591 (2020).

  36. Hansen, J. et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science 369, 1010–1014 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Greaney, A. J. et al. Complete mapping of mutations to the SARS-CoV-2 spike receptor-binding domain that escape antibody recognition. Cell Host Microbe 29, 44–57 (2020).

    Article  PubMed  CAS  Google Scholar 

  38. Starr, T. N. et al. Deep mutational scanning of SARS-CoV-2 receptor binding domain reveals constraints on folding and ACE2 binding. Cell 182, 1295–1310 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lan, J. et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581, 215–220 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Tang, J. W., Toovey, O. T. R., Harvey, K. N. & Hui, D. D. S. Introduction of the South African SARS-CoV-2 variant 501Y.V2 into the UK. J. Infect. 82, e8–e10 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Deng, X. et al. Transmission, infectivity, and neutralization of a spike L452R SARS-CoV-2 variant. Cell https://doi.org/10.1016/j.cell.2021.04.025 (2021).

  42. Shin, J. E. et al. Protein design and variant prediction using autoregressive generative models. Nat. Commun. 12, 2403 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wei, L. et al. Overlapping hotspots in CDRs are critical sites for V region diversification. Proc. Natl Acad. Sci. USA 112, E728–E737 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ovchinnikov, V., Louveau, J. E., Barton, J. P., Karplus, M. & Chakraborty, A. K. Role of framework mutations and antibody flexibility in the evolution of broadly neutralizing antibodies. eLife 7, e33038 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hess, G. T. et al. Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat. Methods 13, 1036–1042 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wright, S. The roles of mutation, inbreeding, crossbreeding and selection in evolution. Proc. Sixth Int. Congr. Genet. 1, 356–366 (1932).

    Google Scholar 

  47. Rix, G. et al. Scalable continuous evolution for the generation of diverse enzyme variants encompassing promiscuous activities. Nat. Commun. 11, 5644 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rix, G. & Liu, C. C. Systems for in vivo hypermutation: a quest for scale and depth in directed evolution. Curr. Opin. Chem. Biol. 64, 20–26 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Wang, T., Badran, A. H., Huang, T. P. & Liu, D. R. Continuous directed evolution of proteins with improved soluble expression. Nat. Chem. Biol. 14, 972–980 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Gunge, N. & Sakaguchi, K. Intergeneric transfer of deoxyribonucleic acid killer plasmids, pGKl1 and pGKl2, from Kluyveromyces lactis into Saccharomyces cerevisiae by cell fusion. J. Bacteriol. 147, 155–160 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gietz, R. D. & Schiestl, R. H. High-efficiency yeast transformation using the LiAc/SS carrier DNA/PEG method. Nat. Protoc. 2, 31–34 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Lee, M. E., DeLoache, W. C., Cervantes, B. & Dueber, J. E. A highly characterized yeast toolkit for modular, multipart assembly. ACS Synth. Biol. 4, 975–986 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Radoshitzky, S. R. et al. Transferrin receptor 1 is a cellular receptor for New World haemorrhagic fever arenaviruses. Nature 446, 92–96 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zhang, F. et al. Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat. Biotechnol. 29, 149–153 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Iyer, A. S. et al. Persistence and decay of human antibody responses to the receptor binding domain of SARS-CoV-2 spike protein in COVID-19 patients. Sci. Immunol. 5, eabe0367 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank W. Capel for assistance with nanobody purifications, Z. Zhong, C. Carlson, T. Loveless, A. Banks and other members of the Liu and Kruse groups for experimental assistance, materials and thoughtful discussions, and G. Arzumanyan for the pGA promoter mutations discovered in his OrthoRep continuous protein evolution experiments (unrelated to this study). We thank D. Trono (EPFL), F. Zhang (Broad Institute), H. Mou (Scripps Research) and M. Farzan (Scripps Research) for the gift of plasmids and cells used in our study. We also acknowledge the support of the Center for Macromolecular Interactions at Harvard Medical School. This work was funded by NIH 1DP2GM119163 (C.C.L.), NIH NIGMS 1R35GM136297 (C.C.L.), the Moore Inventor Fellowship (C.C.L.), the UCI COVID-19 Basic, Translational and Clinical Research Fund (C.C.L.), NIH DP5OD021345 (A.C.K.), a Vallee Scholars Award (A.C.K.), NIH NIAID R01AI146779 (A.G.S.), a Massachusetts Consortium on Pathogenesis Readiness (MassCPR; A.G.S.), training grants NIGMS T32GM007753 (B.M.H. and T.M.C.) and T32AI007245 (J.F.) and NIH NCI 1R01CA260415 (C.C.L., A.C.K. and D.S.M.).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to experimental design and data analysis. A.W., C.M., A.C.K. and C.C.L. were responsible for the conception of AHEAD. A.W., M.H.H., V.J.H. and K.M.N. carried out experiments establishing the first generation of AHEAD and made improvements to reach the second-generation AHEAD system. C.M. carried out AHEAD experiments for the evolution of anti-AT1R nanobodies and selected parent anti-SARS-CoV-2 for evolution using AHEAD. A.W., J.R.C. and M.H.H. carried out AHEAD experiments for the evolution of anti-GFP, anti-HSA and anti-SARS-CoV-2 nanobodies. A.W., C.M., M.S.A.G., S.C. and L.M.W. characterized the activities of evolved nanobodies in binding assays (A.W., C.M. and L.M.W.), SPR measurements (C.M. and M.S.A.G.), neutralization assays (S.C.) and ACE2 competition assays (S.C.). J.F., B.M.H., T.M.C. and A.W. were responsible for the expression of RBD used throughout this study. A.W. and V.J.H. were responsible for the RBD mutational scanning experiments and NGS data analysis that mapped target epitopes and RBD escape mutations for anti-RBD nanobodies. J.-E.S. and D.S.M. were responsible for computational design aspects for the naïve ~200,000-member nanobody library and A.W. inserted that library into AHEAD. A.C.K. and C.C.L. oversaw all aspects of the project, D.S.M. supervised computational nanobody library design, J.A. supervised neutralization and ACE2 competition assays, and A.G.S. supervised the preparation of RBD. A.W. carried out the deep mutational scanning analysis. A.W., C.M., A.C.K. and C.C.L. wrote the manuscript, with input and contributions from all authors.

Corresponding authors

Correspondence to Andrew C. Kruse or Chang C. Liu.

Ethics declarations

Competing interests

Provisional patents (US Patent Application No. 63/123,558 and US Patent Application No. 63/111,860) have been filed on this work. A.C.K. is a co-founder and advisor of Tectonic Therapeutic, Inc., and of the Institute for Protein Innovation. C.C.L. is a co-founder of K2 Biotechnologies, Inc., which focuses on the use of continuous evolution technologies applied to antibody engineering.

Additional information

Peer review information Nature Chemical Biology thanks Theam Soon Lim and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Antibody fragments.

Single-chain variable fragments and nanobodies are displayed on the surface of yeast in this study. Their relationships to conventional antibodies are depicted.

Extended Data Fig. 2 Evolution of anti-AT1R nanobodies by AHEAD.

a, Contributions of individual mutations fixed during the evolution of AT110 by AHEAD. Affinity (EC50) of each nanobody for AT1R was determined by measuring binding of yeast-displayed nanobodies to each concentration of AT1R-angiotensin II complex (X-axis) in a single replicate and fitting the resulting binding curve. b, Amino acid sequence of AT110 and evolved variants. Mutations that were discovered using AHEAD are underlined in bold. Mutations that were discovered in a previous AT110 evolution experiment using a standard error prone PCR library approach19 are highlighted in yellow.

Extended Data Fig. 3 Optimization of antibody display in AHEAD.

a, Maps of orthogonal p1 plasmids containing OrthoRep parts driving expression of nanobodies in the first-generation AHEAD 1.0 and improved second-generation AHEAD 2.0 systems. Nb = nanobody, tAHD1 = ADH1 terminator, polyA = polyadenosine tail. b, Increased functional expression of nanobody AT110 using all AHEAD 2.0 parts as determined by FACS. The induced population in AHEAD 2.0 shows an ~25-fold increase in nanobody display levels (determined by mean fluorescence intensity of the cell population) compared to AHEAD 1.0.

Extended Data Fig. 4 Optimization of antibody display in AHEAD and evolution of anti-GFP and anti-HSA antibodies using the optimized second-generation AHEAD 2.0 system.

a, Architectures for nanobody display in the first-generation AHEAD 1.0 and improved second-generation AHEAD 2.0 systems. b, Selection of a new leader sequence for higher nanobody display. FACS plots showing the progressive enrichment of higher efficiency leader sequences across 3 rounds of selection (left panel). Nanobody display level using app8 compared to the selected app8i1 variant (right panel). n = 6, error bars represent ± s.d. c, Selected FACS plots showing affinity maturation of Nb.b201 through AHEAD cycles. d, Selected FACS plots showing affinity maturation of Lag42 through AHEAD cycles. e, (left) Affinities (EC50) of improved high-affinity anti-HSA nanobodies evolved using AHEAD. Binding of yeast-displayed nanobodies by each concentration of HSA was measured in replicate (n = 3, error bars represent ± s.d.) and EC50s were determined by fitting each binding curve. (right) Affinities (EC50) of improved high-affinity anti-GFP nanobodies evolved using AHEAD. Binding of yeast-displayed nanobodies by each concentration of GFP was measured in replicate (n = 3, error bars represent ± s.d.) and EC50s were determined by fitting each binding curve.

Extended Data Fig. 5 Evolution of anti-RBD nanobodies.

a, Isolation of parent anti-RBD nanobodies. (left) FACS plot showing enrichment of initial anti-RBD nanobody clones from a naïve nanobody library32. The green polygon corresponds to the gate used for sorting. (right) Schematic showing the separation of parent clones into different AHEAD experiments in order to minimize competition among parents and their lineages, avoiding early loss of weak parents that have the potential to yield superior descendants later during affinity maturation. b, Selected FACS plots showing anti-RBD affinity maturation by cycles of AHEAD in 8 independent experiments, each starting from one of the 8 parent clones identified from the naïve nanobody library (see Extended Data Fig. 5a). Red polygons correspond to the gates used for sorting.

Extended Data Fig. 6 Affinities of anti-RBD nanobodies determined by surface plasmon resonance (SPR) or EC50 measurements.

SPR or EC50 binding curves are shown for each anti-RBD nanobody characterized in this study. For SPR measurements (Y-axis = Response), kinetic fits are shown where available and steady-state affinity fits are shown for nanobodies for which the on and off rates could not be determined. For EC50 affinities (Y-axis = Normalized Fluorescence), binding of yeast-displayed nanobodies by each concentration of RBD was determined in biological triplicate (n = 3, error bars represent ± s.d.) and EC50s were determined by fitting each binding curve.

Extended Data Fig. 7 Neutralization assays and ACE2 competition assays for anti-RBD nanobodies evolved with AHEAD.

a, Neutralization plots for all anti-RBD nanobodies characterized in this study. Each nanobody concentration (X-axis) was tested in replicate. n = 6, error bars represent ± s.d. b, Biolayer interferometry (BLI) traces measuring ACE2 competition for anti-RBD nanobodies. CR3022 is an anti-RBD antibody that does not compete with ACE2 binding (no competition control) whereas SC1A-B12 is an anti-RBD antibody that competes strongly with RBD binding.

Extended Data Fig. 8 Evolution of an anti-GFP nanobody from a computationally-designed 200,000-member naïve nanobody library encoded on AHEAD.

a, Representative FACS plots showing enrichment of a GFP-binding clone from the nanobody library and subsequent emergence and fixation of a mutation that increases GFP binding across AHEAD cycles. b, Affinity (EC50) of the AHEAD-evolved anti-GFP nanobody, NbG1i1, isolated from AHEAD cycle 6 as compared to its parent, NbG1, that fixed in AHEAD cycle 3. Binding of yeast-displayed nanobodies by each concentration of GFP was determined in relicate (n = 3, error bars represent ± s.d.) and EC50s were determined by fitting each binding curve.

Extended Data Fig. 9 Gating strategy for singlets in all FACS experiments.

(left) Forward scatter (horizontal axes) versus side scatter (vertical axes) of a representative population of yeast cells. Red circle represents cells passing the gate. (right) Forward scatter area (horizontal axes) vs. forward scatter height (vertical axes) gating of cells that passed through the previous gate. Green boundary represents cells passing the gate. For all FACS experiments, only cells sorted through both gates were used in nanobody expression and binding gates and.

Supplementary information

Supplementary Information

Supplementary Tables 1–4.

Reporting Summary

Supplementary Data 1

Information and activities for all anti-SARS-CoV-2 nanobodies characterized in this study.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wellner, A., McMahon, C., Gilman, M.S.A. et al. Rapid generation of potent antibodies by autonomous hypermutation in yeast. Nat Chem Biol 17, 1057–1064 (2021). https://doi.org/10.1038/s41589-021-00832-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41589-021-00832-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing