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:

Reprogramming human B cells with custom heavy-chain antibodies

Abstract

The immunoglobulin locus of B cells can be reprogrammed by genome editing to produce custom or non-natural antibodies that are not induced by immunization. However, current strategies for antibody reprogramming require complex expression cassettes and do not allow for customization of the constant region of the antibody. Here we show that human B cells can be edited at the immunoglobulin heavy-chain locus to express heavy-chain-only antibodies that support alterations to both the fragment crystallizable domain and the antigen-binding domain, which can be based on both antibody and non-antibody components. Using the envelope protein (Env) from the human immunodeficiency virus as a model antigen, we show that B cells edited to express heavy-chain antibodies to Env support the regulated expression of B cell receptors and antibodies through alternative splicing and that the cells respond to the Env antigen in a tonsil organoid model of immunization. This strategy allows for the reprogramming of human B cells to retain the potential for in vivo amplification while producing molecules with flexibility of composition beyond that of standard antibodies.

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: Genome editing at the constant region of the IgH locus.
Fig. 2: Engineering B cell lines to express anti-HIV HCAbs.
Fig. 3: Engineering primary human B cells.
Fig. 4: Differentiation of engineered B cells and regulation of HCAb expression.
Fig. 5: Antigen-specific expansion of HCAb-engineered B cells.
Fig. 6: Editing with alternative HCAb structures and modified Fc domains.

Similar content being viewed by others

Data availability

Sequences of all guide RNAs, expression plasmids, homology-donor plasmids, AAV vectors and primers are provided with this paper and can be found in Supplementary Tables 1 and 36, respectively. Sequences of all insert plasmid features are also described in Supplementary Tables 3 and 4. The antibodies used are listed in Supplementary Table 7. Uncropped gel images for Extended Data Figs. 1c, 2b, 5b and Extended Data Fig. 6d can be found in Supplementary Fig. 3. Representative gating schemes for the flow cytometry analysis of peripheral blood B cells in Figs. 3a,b, 6a,d,i and Extended Data Figs. 4f, 5c are in Supplementary Fig. 8. Gating schemes for Fig. 3g–j are in Supplementary Fig. 10. Gating schemes for Fig. 4a, Extended Data Fig. 6b–d and Supplementary Fig. 12 are in Supplementary Fig. 11. The gating scheme for Fig. 5c–e is in Supplementary Fig. 14. Deep sequencing data have been deposited in the National Center for Biotechnology Information’s Sequence Read Archive database: sequencing data of primary B cells treated with Cas9-sg05 RNPs in Fig. 1d and Extended Data Fig. 1d,e can be found under accession code PRJNA994591 and sequencing data of SHM in edited Raji cells in Supplementary Figs. 4 and 5 can be found under accession code PRJNA1095559. Source data are provided with this paper.

Code availability

Core functions of NGS analysis were performed using publicly available open-source software, as described in the Methods section. R scripts used for Cas9-sg05 RNP activity and SHM in Raji cells are available on reasonable request.

References

  1. Qerqez, A. N., Silva, R. P. & Maynard, J. A. Outsmarting pathogens with antibody engineering. Annu. Rev. Chem. Biomol. Eng. 14, 217–241 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Carter, P. J. & Rajpal, A. Designing antibodies as therapeutics. Cell 185, 2789–2805 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. Page, A., Fusil, F. & Cosset, F. L. Towards physiologically and tightly regulated vectored antibody therapies. Cancers 12, 962 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhan, W., Muhuri, M., Tai, P. W. L. & Gao, G. Vectored immunotherapeutics for infectious diseases: can rAAVs be the game changers for fighting transmissible pathogens? Front. Immunol. 12, 673699 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Martinez-Navio, J. M. et al. Long-term delivery of an anti-SIV monoclonal antibody with AAV. Front. Immunol. 11, 449 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gardner, M. R. et al. Anti-drug antibody responses impair prophylaxis mediated by AAV-delivered HIV-1 broadly neutralizing antibodies. Mol. Ther. 27, 650–660 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fuchs, S. P., Martinez-Navio, J. M., Rakasz, E. G., Gao, G. & Desrosiers, R. C. Liver-directed but not muscle-directed AAV-antibody gene transfer limits humoral immune responses in rhesus monkeys. Mol. Ther. Methods Clin. Dev. 16, 94–102 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Priddy, F. H. et al. Adeno-associated virus vectored immunoprophylaxis to prevent HIV in healthy adults: a phase 1 randomised controlled trial. Lancet HIV 6, e230–e239 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Casazza, J. P. et al. Safety and tolerability of AAV8 delivery of a broadly neutralizing antibody in adults living with HIV: a phase 1, dose-escalation trial. Nat. Med. 28, 1022–1030 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rogers, G. L. & Cannon, P. M. Genome edited B cells: a new frontier in immune cell therapies. Mol. Ther. 29, 3192–3204 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nahmad, A. D. et al. Engineered B cells expressing an anti-HIV antibody enable memory retention, isotype switching and clonal expansion. Nat. Commun. 11, 5851 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Huang, D. et al. Vaccine elicitation of HIV broadly neutralizing antibodies from engineered B cells. Nat. Commun. 11, 5850 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Moffett, H. F. et al. B cells engineered to express pathogen-specific antibodies protect against infection. Sci Immunol 4, eaax0644 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hartweger, H. et al. HIV-specific humoral immune responses by CRISPR/Cas9-edited B cells. J. Exp. Med. 216, 1301–1310 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Nahmad, A. D. et al. In vivo engineered B cells secrete high titers of broadly neutralizing anti-HIV antibodies in mice. Nat. Biotechnol. 40, 1241–1249 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ou, T. et al. Reprogramming of the heavy-chain CDR3 regions of a human antibody repertoire. Mol. Ther. 30, 184–197 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Voss, J. E. et al. Reprogramming the antigen specificity of B cells using genome-editing technologies. eLife 8, e42995 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Greiner, V. et al. CRISPR-mediated editing of the B cell receptor in primary human B cells. iScience 12, 369–378 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Saunders, K. O. Conceptual approaches to modulating antibody effector functions and circulation half-life. Front. Immunol. 10, 1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Delidakis, G., Kim, J. E., George, K. & Georgiou, G. Improving antibody therapeutics by manipulating the Fc domain: immunological and structural considerations. Annu. Rev. Biomed. Eng. 24, 249–274 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  22. Yong Joon Kim, J., Sang, Z., Xiang, Y., Shen, Z. & Shi, Y. Nanobodies: robust miniprotein binders in biomedicine. Adv. Drug Deliv. Rev. 195, 114726 (2023).

    Article  CAS  PubMed  Google Scholar 

  23. Peterson, M. L. Immunoglobulin heavy chain gene regulation through polyadenylation and splicing competition. Wiley Interdiscip. Rev. RNA 2, 92–105 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. McCoy, L. E. et al. Potent and broad neutralization of HIV-1 by a llama antibody elicited by immunization. J. Exp. Med. 209, 1091–1103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Koch, K. et al. Selection of nanobodies with broad neutralizing potential against primary HIV-1 strains using soluble subtype C gp140 envelope trimers. Sci. Rep. 7, 8390 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Singer, P. A. & Williamson, A. R. Cell surface immunoglobulin μ and γ chains of human lymphoid cells are of higher apparent molecular weight than their secreted counterparts. Eur. J. Immunol. 10, 180–186 (1980).

    Article  CAS  PubMed  Google Scholar 

  27. Ford, G. S., Yin, C. H., Barnhart, B., Sztam, K. & Covey, L. R. CD40 ligand exerts differential effects on the expression of Iγ transcripts in subclones of an IgM+ human B cell lymphoma line. J. Immunol. 160, 595–605 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Zhou, T. et al. Structural basis for llama nanobody recognition and neutralization of HIV-1 at the CD4-binding site. Structure 30, 862–875 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Luo, X. M. et al. Engineering human hematopoietic stem/progenitor cells to produce a broadly neutralizing anti-HIV antibody after in vitro maturation to human B lymphocytes. Blood 113, 1422–1431 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Maul, R. W. & Gearhart, P. J. AID and somatic hypermutation. Adv. Immunol. 105, 159–191 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ruckerl, F., Busse, B. & Bachl, J. Episomal vectors to monitor and induce somatic hypermutation in human Burkitt-lymphoma cell lines. Mol. Immunol. 43, 1645–1652 (2006).

    Article  PubMed  Google Scholar 

  32. Vaisman-Mentesh, A., Gutierrez-Gonzalez, M., DeKosky, B. J. & Wine, Y. The molecular mechanisms that underlie the immune biology of anti-drug antibody formation following treatment with monoclonal antibodies. Front. Immunol. 11, 1951 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cohen, Y. Z. et al. Safety, pharmacokinetics, and immunogenicity of the combination of the broadly neutralizing anti-HIV-1 antibodies 3BNC117 and 10-1074 in healthy adults: a randomized, phase 1 study. PLoS ONE 14, e0219142 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Asaadi, Y., Jouneghani, F. F., Janani, S. & Rahbarizadeh, F. A comprehensive comparison between camelid nanobodies and single chain variable fragments. Biomark. Res. 9, 87 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Vincke, C. et al. General strategy to humanize a camelid single-domain antibody and identification of a universal humanized nanobody scaffold. J. Biol. Chem. 284, 3273–3284 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Belanger, K. & Tanha, J. High-efficacy, high-manufacturability human VH domain antibody therapeutics from transgenic sources. Protein Eng. Des. Sel. 34, gzab012 (2021).

    Article  PubMed  Google Scholar 

  37. Sonoda, E., Hochegger, H., Saberi, A., Taniguchi, Y. & Takeda, S. Differential usage of non-homologous end-joining and homologous recombination in double strand break repair. DNA Repair 5, 1021–1029 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Rogers, G. L. et al. Optimization of AAV6 transduction enhances site-specific genome editing of primary human lymphocytes. Mol. Ther. Methods Clin. Dev. 23, 198–209 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hung, K. L. et al. Engineering protein-secreting plasma cells by homology-directed repair in primary human B cells. Mol. Ther. 26, 456–467 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Sanz, I. et al. Challenges and opportunities for consistent classification of human B cell and plasma cell populations. Front. Immunol. 10, 2458 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Schiroli, G. et al. Precise gene editing preserves hematopoietic stem cell function following transient p53-mediated DNA damage response. Cell Stem Cell 24, 551–565 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Vettermann, C. & Schlissel, M. S. Allelic exclusion of immunoglobulin genes: models and mechanisms. Immunol. Rev. 237, 22–42 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. He, J. S. et al. The distinctive germinal center phase of IgE+ B lymphocytes limits their contribution to the classical memory response. J. Exp. Med. 210, 2755–2771 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wagar, L. E. et al. Modeling human adaptive immune responses with tonsil organoids. Nat. Med. 27, 125–135 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kastenschmidt, J. M. et al. Influenza vaccine format mediates distinct cellular and antibody responses in human immune organoids. Immunity 56, 1910–1926 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen, W. et al. Exceptionally potent and broadly cross-reactive, bispecific multivalent HIV-1 inhibitors based on single human CD4 and antibody domains. J. Virol. 88, 1125–1139 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Walker, L. M. et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477, 466–470 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bates, A. & Power, C. A. David vs. Goliath: the structure, function, and clinical prospects of antibody fragments. Antibodies 8, 28 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Finck, A. V., Blanchard, T., Roselle, C. P., Golinelli, G. & June, C. H. Engineered cellular immunotherapies in cancer and beyond. Nat. Med. 28, 678–689 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hibi, T. & Dosch, H. M. Limiting dilution analysis of the B cell compartment in human bone marrow. Eur. J. Immunol. 16, 139–145 (1986).

    Article  CAS  PubMed  Google Scholar 

  51. Hammarlund, E. et al. Plasma cell survival in the absence of B cell memory. Nat. Commun. 8, 1781 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Cheng, R. Y. et al. Ex vivo engineered human plasma cells exhibit robust protein secretion and long-term engraftment in vivo. Nat. Commun. 13, 6110 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Davis, C. W. et al. Influenza vaccine-induced human bone marrow plasma cells decline within a year after vaccination. Science 370, 237–241 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Monzon-Posadas, W. O. et al. Longitudinal monitoring of mRNA-vaccine-induced immunity against SARS-CoV-2. Front. Immunol. 14, 1066123 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Koike, T., Harada, K., Horiuchi, S. & Kitamura, D. The quantity of CD40 signaling determines the differentiation of B cells into functionally distinct memory cell subsets. eLife 8, e44245 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Joshi, K. K. et al. Elucidating heavy/light chain pairing preferences to facilitate the assembly of bispecific IgG in single cells. mAbs 11, 1254–1265 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Jin, B. K., Odongo, S., Radwanska, M. & Magez, S. NANOBODIES®: a review of generation, diagnostics and therapeutics. Int. J. Mol. Sci. 24, 5994 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hamers-Casterman, C. et al. Naturally occurring antibodies devoid of light chains. Nature 363, 446–448 (1993).

    Article  CAS  PubMed  Google Scholar 

  59. Blanc, M. R. et al. A one-step exclusion-binding procedure for the purification of functional heavy-chain and mammalian-type gamma-globulins from camelid sera. Biotechnol. Appl. Biochem. 54, 207–212 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Daley-Bauer, L. P. et al. Contributions of conventional and heavy-chain IgG to immunity in fetal, neonatal, and adult alpacas. Clin. Vaccine Immunol. 17, 2007–2015 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Llewellyn, G. N. et al. Comparison of SARS-CoV-2 entry inhibitors based on ACE2 receptor or engineered Spike-binding peptides. J. Virol. 97, e0068423 (2023).

    Article  PubMed  Google Scholar 

  62. Laursen, N. S. et al. Universal protection against influenza infection by a multidomain antibody to influenza hemagglutinin. Science 362, 598–602 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Smith, P., DiLillo, D. J., Bournazos, S., Li, F. & Ravetch, J. V. Mouse model recapitulating human Fcγ receptor structural and functional diversity. Proc. Natl Acad. Sci. USA 109, 6181–6186 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zalevsky, J. et al. Enhanced antibody half-life improves in vivo activity. Nat. Biotechnol. 28, 157–159 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yu, J. et al. A rational approach to enhancing antibody Fc homodimer formation for robust production of antibody mixture in a single cell line. J. Biol. Chem. 292, 17885–17896 (2017).

    Article  CAS  PubMed  Google Scholar 

  66. Vamva, E. et al. A lentiviral vector B cell gene therapy platform for the delivery of the anti-HIV-1 eCD4-Ig-knob-in-hole-reversed immunoadhesin. Mol. Ther. Methods Clin. Dev. 28, 366–384 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Irrgang, P. et al. Class switch toward noninflammatory, spike-specific IgG4 antibodies after repeated SARS-CoV-2 mRNA vaccination. Sci. Immunol. 8, eade2798 (2023).

    Article  CAS  PubMed  Google Scholar 

  68. Roco, J. A. et al. Class-switch recombination occurs infrequently in germinal centers. Immunity 51, 337–350 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Wardemann, H. et al. Predominant autoantibody production by early human B cell precursors. Science 301, 1374–1377 (2003).

    Article  CAS  PubMed  Google Scholar 

  70. Casellas, R. et al. Igκ allelic inclusion is a consequence of receptor editing. J. Exp. Med. 204, 153–160 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Pelanda, R. Dual immunoglobulin light chain B cells: Trojan horses of autoimmunity? Curr. Opin. Immunol. 27, 53–59 (2014).

    Article  CAS  PubMed  Google Scholar 

  72. Sirac, C., Carrion, C., Duchez, S., Comte, I. & Cogne, M. Light chain inclusion permits terminal B cell differentiation and does not necessarily result in autoreactivity. Proc. Natl Acad. Sci. USA 103, 7747–7752 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Velez, M. G. et al. Ig allotypic inclusion does not prevent B cell development or response. J. Immunol. 179, 1049–1057 (2007).

    Article  CAS  PubMed  Google Scholar 

  74. Makdasi, E. & Eilat, D. L chain allelic inclusion does not increase autoreactivity in lupus-prone New Zealand Black/New Zealand White mice. J. Immunol. 190, 1472–1480 (2013).

    Article  CAS  PubMed  Google Scholar 

  75. Peterson, J. N. et al. Elevated detection of dual antibody B cells identifies lupus patients with B cell-reactive VH4-34 autoantibodies. Front. Immunol. 13, 795209 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Rogers, G. L., Chen, H. Y., Morales, H. & Cannon, P. M. Homologous recombination-based genome editing by clade F AAVs is inefficient in the absence of a targeted DNA break. Mol. Ther. 27, 1726–1736 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Lock, M. et al. Characterization of a recombinant adeno-associated virus type 2 Reference Standard Material. Hum. Gene Ther. 21, 1273–1285 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Rogers, G. L. et al. Unique roles of TLR9- and MyD88-dependent and -independent pathways in adaptive immune responses to AAV-mediated gene transfer. J. Innate Immun. 7, 302–314 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Tatiossian, K. J. et al. Rational selection of CRISPR–Cas9 guide RNAs for homology-directed genome editing. Mol. Ther. 29, 1057–1069 (2021).

    Article  CAS  PubMed  Google Scholar 

  80. Llewellyn, G. N. et al. Humanized mouse model of HIV-1 latency with enrichment of latent virus in PD-1+ and TIGIT+ CD4 T cells. J. Virol. 93, e02086-18 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Sarzotti-Kelsoe, M. et al. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. J. Immunol. Methods 409, 131–146 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Korbie, D. J. & Mattick, J. S. Touchdown PCR for increased specificity and sensitivity in PCR amplification. Nat. Protoc. 3, 1452–1456 (2008).

    Article  CAS  PubMed  Google Scholar 

  83. Conant, D. et al. Inference of CRISPR edits from Sanger trace data. CRISPR J. 5, 123–130 (2022).

    Article  CAS  PubMed  Google Scholar 

  84. Hill, J. T. et al. Poly peak parser: method and software for identification of unknown indels using sanger sequencing of polymerase chain reaction products. Dev. Dyn. 243, 1632–1636 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Rohland, N., Harney, E., Mallick, S., Nordenfelt, S. & Reich, D. Partial uracil-DNA-glycosylase treatment for screening of ancient DNA. Philos. Trans. R Soc. Lond B 370, 20130624 (2015).

    Article  Google Scholar 

  86. Masella, A. P., Bartram, A. K., Truszkowski, J. M., Brown, D. G. & Neufeld, J. D. PANDAseq: paired-end assembler for illumina sequences. BMC Bioinform. 13, 31 (2012).

    Article  CAS  Google Scholar 

  87. Morgan, M. et al. ShortRead: a bioconductor package for input, quality assessment and exploration of high-throughput sequence data. Bioinformatics 25, 2607–2608 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Pagès, H., Aboyoun, P., Gentleman, R. & DebRoy, S. Biostrings: efficient manipulation of biological strings. R package version 2.62.0 https://bioconductor.org/packages/Biostrings (2021).

  89. Schauberger, P. & Walker, A. openxlsx: read, write and edit xlsx files. R package version 4.2.5.1 https://CRAN.R-project.org/package=openxlsx (2022).

  90. Fields, B., Moeskjaer, S., Friman, V. P., Andersen, S. U. & Young, J. P. W. MAUI-seq: metabarcoding using amplicons with unique molecular identifiers to improve error correction. Mol. Ecol. Resour. 21, 703–720 (2021).

    Article  CAS  PubMed  Google Scholar 

  91. Karst, S. M. et al. High-accuracy long-read amplicon sequences using unique molecular identifiers with Nanopore or PacBio sequencing. Nat. Methods 18, 165–169 (2021).

    Article  CAS  PubMed  Google Scholar 

  92. Rogozin, I. B. & Diaz, M. Cutting edge: DGYW/WRCH is a better predictor of mutability at G:C bases in Ig hypermutation than the widely accepted RGYW/WRCY motif and probably reflects a two-step activation-induced cytidine deaminase-triggered process. J. Immunol. 172, 3382–3384 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Wagih, O. ggseqlogo: A ‘ggplot2’ extension for drawing publication-ready sequence logos. R package version 0.1 https://CRAN.R-project.org/package=ggseqlogo (2017).

  94. Abhinandan, K. R. & Martin, A. C. Analyzing the ‘degree of humanness’ of antibody sequences. J. Mol. Biol. 369, 852–862 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank G.N. Llewellyn and C. Diadhiou for critical discussions and assistance with reviewing the manuscript. P.M.C. discloses support for the research described in this study from National Institutes of Health grants HL156274, AI164561, AI164556 and MH130178. G.L.R. discloses support for the research described in this study from a Career Development Award from the American Society of Gene & Cell Therapy. H.-Y.C. discloses support for the research described in this study from a Taiwan USC scholarship. Tonsil material was provided by the Norris Comprehensive Cancer Center Translational Pathology Core, supported in part by National Institutes of Health grant CA014089 from the National Cancer Institute. The content of this study is solely the responsibility of the authors and does not necessarily represent the official views of the American Society of Gene & Cell Therapy, National Cancer Institute or National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

G.L.R., C.H., K.S., A.M., H.M. and H.-Y.C. developed editing reagents and performed cell-editing experiments. G.L.R., C.H., A.M., X.H., K.S. and C.-H.C. established assays and performed antibody and sample analyses. G.L.R. performed NGS experiments, analysed data and visualized results. E.J.K. supervised the collection of tonsil tissue. G.L.R. and P.M.C. conceived the study, designed experiments, interpreted data and wrote the manuscript with input from other authors. P.M.C. supervised the study.

Corresponding author

Correspondence to Paula M. Cannon.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Extended analyses of genome editing at the constant region of the IgH locus.

(a) K562 cells were electroporated with Cas9 RNPs containing indicated gRNAs and matched ssODN homology donors to insert an XhoI restriction site (n = 3). HDR editing was measured by Sanger sequencing and ICE analysis. (b) K562 cells were electroporated with Cas9 RNPs for indicated gRNAs and a matched plasmid homology donor containing a GFP expression cassette (n = 3). HDR editing was measured by flow cytometry for GFP expression after 3 weeks. (c) Site-specific insertion of GFP expression cassettes in AAV6-edited K562 cells was confirmed by in-out PCR for each tested gRNA. Uncropped gel is available in Supplementary Fig. 3a. Created with Biorender.com. (d) On- and off-target activity of sg05 was measured at indicated IGHG genes in primary human B cells, 5 days after editing, by targeted amplicon deep sequencing. Aggregate mutations at each base in a 50 bp window surrounding the sg05 cut site (0; orange dotted line) are shown for each gene. (e) Percentage mutated reads at each IGHG gene calculated as for all changes (≥1 bp changed), which gives a higher background than when a cutoff of ≥2 bp is selected, as shown in Fig. 1d (n = 2). Error bars show mean ± SEM. Statistics were calculated by 2-way ANOVA with Fisher’s LSD test.

Source data

Extended Data Fig. 2 Extended analyses of engineered B cell lines expressing anti-HIV HCAbs.

(a) Raji B cells were edited with sg05 Cas9 RNPs plus a plasmid homology donor to insert a GFP expression cassette and editing rates were measured by flow cytometry. (b) Ramos B cells were edited with sg05 Cas9 RNPs and plasmid homology donors for J3, A6 or a control GFP expression cassette. Editing rates were determined by flow cytometry. (c-d) Raji (c) and Ramos (d) cells edited by sg05 Cas9 RNPs and J3, A6, or GFP plasmid homology donors, post sorting by FACS. (e) GFP-edited Raji cells were sorted by FACS for GFP expression, and the enriched population was subjected to in-out PCR and Sanger sequencing of PCR bands to confirm precise insertion. The dotted line indicates the predicted sg05 cut site. Uncropped gel is available in Supplementary Fig. 3b. (f) Secretion of HCAbs was detected by total IgG ELISA from J3 or A6-edited Raji and Ramos cells, but not from GFP-edited control cells.

Source data

Extended Data Fig. 3 Lack of pairing between HCAb and co-expressed light chain.

(a) Assay schematic. 293T cells were transfected with expression plasmids for the J3 VHH HCAb, a full-length (FL) conventional human IgG1 derived from Ofatumumab, or its Igκ L chain only (LC), or combinations as indicated. Supernatants, including mixed supernatants as controls, were evaluated by ELISAs based on HIV gp120 binding by the J3 VHH and detection with an anti-VHH antibody (control), or anti-Igκ L chain antibodies to detect pairings between the J3 HCAb and either the FL or LC components. FabALACTICA digestion is expected to cleave FL antibodies into Fab and Fc fragments and may also cleave HCAbs. Created with Biorender.com. (b-c) Results of gp120-VHH (b) and gp120-Igκ (c) ELISAs, with or without FabALACTICA digestion (n = 4 J3 and J3/FL; n = 2 FL, LC, J3/LC, L3+LC, and J3 + FL). Cross-pairing was only observed after co-transfection of J3 HCAb and FL antibody, consistent with H chain interactions that were released by FabALACTICA digestion. In contrast, the L chain alone did not pair with the J3 HCAb. Error bars show mean ± SEM. Statistics were performed using 2-way ANOVA with Fisher’s LSD test.

Source data

Extended Data Fig. 4 Comparison of culture conditions for primary human B cells.

Primary human B cells were cultured with the indicated stimulation conditions (DP, BAC, RP105) and basal media (IMDM, XF, XF plus FBS) as described in Supplementary Fig. 7. Cultures were pre-activated for 3 days before measurements were started on day 0. (a) Fold expansion of cells over time (n = 3). (b) Viability of cells over time (n = 3). (c) Cell size, measured by flow cytometry as forward scatter (FSC) median fluorescence intensity (MFI) on day 0 (n = 3). (d) Total IgG secreted into supernatants was measured over time by ELISA (n = 4). (e) Total IgG amounts normalized for viable cell counts (n = 4). (f) B cell phenotypes were assessed at day 8 in indicated cultures by flow cytometry, as shown in Supplementary Fig. 8 (n = 3). For BAC + DP, the cells were started in BAC and treated for a total of 5 days, then switched to the DP protocol for the remaining 6 days, as shown in Supplementary Fig. 7. (g) Comparison of total IgG secretion and IgG/cell at day 8 for cells treated with indicated stimulation protocols (n = 3). DP was in IMDM, BAC and BAC + DP were in XF without FBS. Error bars show mean ± SEM. Statistics were calculated by 2-way ANOVA with Tukey’s test (c) or paired 1-way ANOVA with Tukey’s test (g).

Source data

Extended Data Fig. 5 Extended analyses of engineering with AAV6 homology donors.

(a) Editing rates achieved using sg05 Cas9 RNPs and AAV6-J3 homology donors on Raji and Ramos B cell lines. Representative plots are shown, together with summary data (n = 6 control Raji and AAV6 + Cas9 Ramos, n = 4 control Ramos and AAV6 + Cas9 Raji). (b) Site-specific insertion of the J3 cassette in primary B cells treated with sg05 RNPs and AAV6-J3 donor was confirmed by in-out PCR followed by Sanger sequencing of the band. The dotted line shows the presumed sg05 cleavage site. Uncropped gel image is provided in Supplementary Fig. 3c. (c-g) Primary human B cells were edited with sg05 Cas9 RNPs and AAV6-J3 at the indicated MOIs, with BAC activation in XF media (n = 5 5e5 MOI, n = 3 1e4 and 1e5 MOI). Data from the highest MOI, (5e5), is reproduced here from Fig. 3, for comparison. (c) Editing rates, measured at day 8 by flow cytometry for surface J3-BCR (n = 6 5e5 MOI, n = 3 1e4 and 1e5 MOI). (d) Editing quantified by in-out ddPCR at day 8 and normalized per cell against a control reaction. (e) The yield of edited cells at day 8 was calculated from 5 ×105 starting B cells. (f) Fold expansion of total cells at day 8. (g) J3 HCAb secretion, measured by gp120–IgG ELISA at day 8. Error bars show mean ± SEM. Statistics were calculated by 2-way ANVOA with Šídák method (a), or 1-way ANOVA with Tukey’s test (d-h).

Source data

Extended Data Fig. 6 J3-BCR surface expression is reduced with differentiation.

(a) Edited primary B cells were differentiated in DP plus IMDM media and RNA extracted at indicated time points. RT-PCR was performed using a J3-specific forward primer and IgG reverse primers specific for the secreted (S) or membrane (M) isoforms. Uncropped gel image is provided in Supplementary Fig. 3d. (b-d) Five x 105 primary human B cells were activated with BAC from day −3 and edited at day 0 with Cas9 RNPs and an AAV6-J3 homology donor (MOI = 5 × 105 vg/cell). Edited cells (intracellular IgG+ VHH+) were identified at day 8, as described in Supplementary Fig. 11, for analysis of surface J3-BCR expression (gp120 binding). (b) Representative plot of total (surface and intracellular) VHH vs. surface J3-BCR. c,d, Representative plot (c) and summary (d) of surface J3-BCR expression in edited cells of different populations for n = 3 independent donors. Summary MFIs were normalized to the median of the total population for that donor. Error bars show mean ± SEM.

Source data

Supplementary information

Supplementary Information

Supplementary figures and tables.

Reporting Summary

Supplementary Tables

Supplementary Tables 3–8.

Supplementary Data 1

Source data for Supplementary Figs. 4, 5, 9 and 13.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rogers, G.L., Huang, C., Mathur, A. et al. Reprogramming human B cells with custom heavy-chain antibodies. Nat. Biomed. Eng (2024). https://doi.org/10.1038/s41551-024-01240-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-024-01240-4

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research