Ueda, N. et al. Immunotherapy views within the new period of B-cell modifying. Blood Adv. 5, 1770–1779 (2021).
Edelstein, J., Fritz, M. & Lai, S. Ok. Challenges and alternatives in gene modifying of B cells. Biochem. Pharmacol. 206, 115285 (2022).
Jeske, A. M., Boucher, P., Curiel, D. & Voss, J. Vector methods to actualize B cell-based gene therapies. J. Immunol. 207, 755–764 (2021).
Rogers, G. L. et al. Optimization of AAV6 transduction enhances site-specific genome modifying of main human lymphocytes. Mol. Ther. Strategies Clin. Dev. 23, 198–209 (2021).
Web page, A., Hubert, J., Fusil, F. & Cosset, F.-L. Exploiting B cell switch for most cancers remedy: engineered B cells to eradicate tumors. Int. J. Mol. Sci. 22, 9991 (2021).
Radbruch, A. et al. Competence and competitors: the problem of changing into a long-lived plasma cell. Nat. Rev. Immunol. 6, 741–750 (2006).
Hibi, T. & Dosch, H. M. Limiting dilution evaluation of the B cell compartment in human bone marrow. Eur. J. Immunol. 16, 139–145 (1986).
Amanna, I. J., Carlson, N. E. & Slifka, M. Ok. Length of humoral immunity to frequent viral and vaccine antigens. N. Engl. J. Med. 357, 1903–1915 (2007).
Slifka, M. Ok., Antia, R., Whitmire, J. Ok. & Ahmed, R. Humoral immunity on account of long-lived plasma cells. Immunity 8, 363–372 (1998).
Gorzelany, J. A. & de Souza, M. P. Protein alternative therapies for uncommon illnesses: a breeze for regulatory approval? Sci. Transl. Med. 5, 178fs10 (2013).
Samelson-Jones, B. J. & George, L. A. Adeno-associated virus gene remedy for hemophilia. Annu. Rev. Med. 74, 231–247 (2023).
Srivastava, A. et al. Lentiviral gene remedy with CD34+ hematopoietic cells for hemophilia A. N. Engl. J. Med. 392, 450–457 (2025).
Tiede, A. Half-life prolonged issue VIII for the remedy of hemophilia A. J. Thromb. Haemost. 13, S176–S179 (2015).
Chen, H. H. et al. Enzyme alternative remedy for mucopolysaccharidoses; previous, current, and future. J. Hum. Genet. 64, 1153–1171 (2019).
Chu, W. et al. Standing and frontiers of Fabre illness. Orphanet J. Uncommon Dis. 20, 123 (2025).
Salabarria, S. M. et al. Developments in AAV-mediated gene remedy for Pompe illness. J. Neuromuscul. Dis. 7, 15–31 (2020).
Placci, M., Giannotti, M. I. & Muro, S. Polymer-based drug supply techniques beneath investigation for enzyme alternative and different therapies of lysosomal storage problems. Adv. Drug Deliv. Rev. 197, 114683 (2023).
Ozelo, M. C. et al. Valoctocogene roxaparvovec gene remedy for hemophilia A. N. Engl. J. Med. 386, 1013–1025 (2022).
Pipe, S. W. et al. Gene remedy with etranacogene dezaparvovec for hemophilia B. N. Engl. J. Med. 388, 706–718 (2023).
Mingozzi, F. & Excessive, Ok. A. Overcoming the host immune response to adeno-associated virus gene supply vectors: the race between clearance, tolerance, neutralization, and escape. Annu. Rev. Virol. 4, 511–534 (2017).
Cunningham, S. C., Dane, A. P., Spinoulas, A. & Alexander, I. E. Gene supply to the juvenile mouse liver utilizing AAV2/8 vectors. Mol. Ther. 16, 1081–1088 (2008).
Hill, T. F. et al. Human plasma cells engineered to secrete bispecifics drive efficient in vivo leukemia killing. Mol. Ther. https://doi.org/10.1016/j.ymthe.2024.06.004 (2024).
Luo, B. et al. Engineering of α-PD-1 antibody-expressing long-lived plasma cells by CRISPR/Cas9-mediated focused gene integration. Cell Dying Dis. 11, 973 (2020).
Moffett, H. F. et al. B cells engineered to specific pathogen-specific antibodies defend in opposition to an infection. Sci. Immunol. 4, eaax0644 (2019).
Silacci, P., Mottet, A., Steimle, V., Reith, W. & Mach, B. Developmental extinction of main histocompatibility advanced class II gene expression in plasmocytes is mediated by silencing of the transactivator gene CIITA. J. Exp. Med. 180, 1329–1336 (1994).
Duan, M. et al. Understanding heterogeneity of human bone marrow plasma cell maturation and survival pathways by single-cell analyses. Cell Rep. 42, 112682 (2023).
Younger, D. J. et al. In vivo monitoring of ex-vivo-generated 89Zr-oxine-labeled plasma cells by PET in a non-human primate mannequin. Mol. Ther. https://doi.org/10.1016/j.ymthe.2024.12.042 (2024).
Hung, Ok. L. et al. Engineering protein-secreting plasma cells by homology-directed restore in main human B cells. Mol. Ther. 26, 456–467 (2018).
David, M. et al. Manufacturing of therapeutic ranges of human FIX-R338L by engineered B cells utilizing GMP-compatible medium. Mol. Ther. Strategies Clin. Dev. 31, 101111 (2023).
Liu, H. et al. A precision gene engineered B cell drugs producing sustained ranges of energetic issue IX for hemophilia B remedy. Preprint at bioRxiv https://doi.org/10.1101/2025.04.06.647090 (2025).
Philippidis, A. Immusoft stories promising early information for lead candidate in MPS I. GEN—Genetic Engineering and Biotechnology Information. www.genengnews.com/topics/drug-discovery/immusoft-reports-promising-early-data-for-lead-candidate-in-mps-i/ (2024).
Pipe, S. W. et al. Change into-9: a section 1/2 dose escalation and enlargement research of be-101 for the remedy of adults with reasonably extreme or extreme hemophilia B. Blood 144, 2593.1 (2024).
Rastogi, I. et al. Position of B cells as antigen presenting cells. Entrance. Immunol. 13, 954936 (2022).
Okada, T. et al. Antigen-engaged B cells endure chemotaxis towards the T zone and kind motile conjugates with helper T cells. PLoS Biol. 3, e150 (2005).
Track, W. & Craft, J. T follicular helper cell heterogeneity: time, house, and performance. Immunol. Rev. 288, 85–96 (2019).
Elsner, R. A. & Shlomchik, M. J. Germinal heart and extrafollicular B cell responses in vaccination, immunity, and autoimmunity. Immunity 53, 1136–1150 (2020).
Hartweger, H. et al. HIV-specific humoral immune responses by CRISPR/Cas9-edited B cells. J. Exp. Med. 216, 1301–1310 (2019).
Huang, D. et al. B cells expressing genuine naive human VRC01-class BCRs may be recruited to germinal facilities and affinity mature in a number of impartial mouse fashions. Proc. Natl Acad. Sci. USA 117, 22920–22931 (2020).
Huang, D. et al. Vaccine elicitation of HIV broadly neutralizing antibodies from engineered B cells. Nat. Commun. 11, 5850 (2020).
Nahmad, A. D. et al. Engineered B cells expressing an anti-HIV antibody allow reminiscence retention, isotype switching and clonal enlargement. Nat. Commun. 11, 5851 (2020).
Greiner, V. et al. CRISPR-mediated modifying of the B cell receptor in main human B cells. iScience 12, 369–378 (2019).
Voss, J. E. et al. Reprogramming the antigen specificity of B cells utilizing genome-editing applied sciences. eLife 8, e42995 (2019).
Rogers, G. L. et al. Reprogramming human B cells with customized heavy-chain antibodies. Nat. Biomed. Eng. https://doi.org/10.1038/s41551-024-01240-4 (2024).
Yin, Y. et al. In vivo affinity maturation of mouse B cells reprogrammed to specific human antibodies. Nat. Biomed. Eng. 8, 361–379 (2024).
Wennhold, Ok. et al. Utilizing antigen-specific B cells to mix antibody and T cell-based most cancers immunotherapy. Most cancers Immunol. Res. 5, 730–743 (2017).
Lee-Chang, C. et al. Activation of 4-1BBL+ B cells with CD40 agonism and IFNγ elicits potent immunity in opposition to glioblastoma. J. Exp. Med. 218, e20200913 (2021).
Wang, S. et al. B cell-based remedy produces antibodies that inhibit glioblastoma development. J. Clin. Make investments. 134, e177384 (2024).
Winkler, J. et al. Adoptive switch of donor B lymphocytes: a section 1/2a research for sufferers after allogeneic stem cell transplantation. Blood Adv. 8, 2373–2383 (2024).
Winkler, J. et al. GMP-grade era of B-lymphocytes for adoptive immunotherapy in sufferers after allogeneic stem cell transplantation. Blood 120, 4352 (2012).
Winkler, J. et al. Adoptive switch of purified donor-B-lymphocytes after allogeneic stem cell transplantation: outcomes from a section I/IIa scientific trial. Blood 128, 502 (2016).
Fillatreau, S., Sweenie, C. H., McGeachy, M. J., Grey, D. & Anderton, S. M. B cells regulate autoimmunity by provision of IL-10. Nat. Immunol. 3, 944–950 (2002).
Shen, P. et al. IL-35-producing B cells are vital regulators of immunity throughout autoimmune and infectious illnesses. Nature 507, 366–370 (2014).
Parekh, V. V. et al. B cells activated by lipopolysaccharide, however not by anti-Ig and anti-CD40 antibody, induce anergy in CD8+ T cells: function of TGF-β 1. J. Immunol. 170, 5897–5911 (2003).
Knippenberg, S. et al. Discount in IL-10 producing B cells (Breg) in a number of sclerosis is accompanied by a decreased naive/reminiscence Breg ratio throughout a relapse however not in remission. J. Neuroimmunol. 239, 80–86 (2011).
Blair, P. A. et al. CD19+CD24helloCD38hello B cells exhibit regulatory capability in wholesome people however are functionally impaired in systemic lupus erythematosus sufferers. Immunity 32, 129–140 (2010).
Flores-Borja, F. et al. CD19+CD24helloCD38hello B cells keep regulatory T cells whereas limiting TH1 and TH17 differentiation. Sci. Transl. Med. 5, 173ra23 (2013).
Rosser, E. C. & Mauri, C. Regulatory B cells: origin, phenotype, and performance. Immunity 42, 607–612 (2015).
Shankar, S. et al. Ex vivo-expanded human CD19+TIM-1+ regulatory B cells suppress immune responses in vivo and are dependent upon the TIM-1/STAT3 axis. Nat. Commun. 13, 3121 (2022).
Lee, Ok. M. et al. Suppression of allograft rejection by regulatory B cells induced through TLR signaling. JCI Perception 7, e152213 (2022).
Bao, Y. et al. Ex vivo-generated human CD1c+ regulatory B cells by a chemically outlined system suppress immune responses and alleviate graft-versus-host illness. Mol. Ther. 32, 4372–4382 (2024).
Zambidis, E. T., Kurup, A. & Scott, D. W. Genetically transferred central and peripheral immune tolerance through retroviral-mediated expression of immunogenic epitopes in hematopoietic progenitors or peripheral B lymphocytes. Mol. Med. 3, 212–224 (1997).
El-Amine, M. et al. Mechanisms of tolerance induction by a gene-transferred peptide-IgG fusion protein expressed in B lineage cells. J. Immunol. 165, 5631–5636 (2000).
Melo, M. E. F. et al. Gene switch of Ig-fusion proteins into B cells prevents and treats autoimmune illnesses. J. Immunol. 168, 4788–4795 (2002).
Track, L. et al. Retroviral supply of GAD-IgG fusion assemble induces tolerance and modulates diabetes: a job for CD4+ regulatory T cells and TGF-β? Gene Ther. 11, 1487–1496 (2004).
Lei, T. C. & Scott, D. W. Induction of tolerance to issue VIII inhibitors by gene remedy with immunodominant A2 and C2 domains introduced by B cells as Ig fusion proteins. Blood 105, 4865–4870 (2005).
Wang, X. et al. Immune tolerance induction to issue IX via B cell gene switch: TLR9 signaling delineates between tolerogenic and immunogenic B cells. Mol. Ther. 22, 1139–1150 (2014).
Ahangarani, R. R. et al. In vivo induction of kind 1-like regulatory T cells utilizing genetically modified B cells confers long-term IL-10-dependent antigen-specific unresponsiveness. J. Immunol. 183, 8232–8243 (2009).
Calderón-Gómez, E. et al. Reprogrammed quiescent B cells present an efficient mobile remedy in opposition to continual experimental autoimmune encephalomyelitis. Eur. J. Immunol. 41, 1696–1708 (2011).
Chen, D. et al. Novel engineered B lymphocytes focusing on islet-specific T cells inhibit the event of kind 1 diabetes in non-obese diabetic Scid mice. Entrance. Immunol. 14, 1227133 (2023).
Pitner, R. A. et al. Blunting particular T-dependent antibody responses with engineered ‘decoy’ B cells. Mol. Ther. 32, 3453–3469 (2024).
Luo, X. M. et al. Engineering human hematopoietic stem/progenitor cells to provide a broadly neutralizing anti-HIV antibody after in vitro maturation to human B lymphocytes. Blood 113, 1422–1431 (2009).
Cheng, R. Y.-H. et al. Ex vivo engineered human plasma cells exhibit strong protein secretion and long-term engraftment in vivo. Nat. Commun. 13, 6110 (2022).
He, W. et al. Heavy-chain CDR3-engineered B cells facilitate in vivo analysis of HIV-1 vaccine candidates. Immunity 56, 2408–2424.e6 (2023).
Serafini, M., Naldini, L. & Introna, M. Molecular proof of inefficient transduction of proliferating human B lymphocytes by VSV-pseudotyped HIV-1-derived lentivectors. Virology 325, 413–424 (2004).
Janssens, W. et al. Effectivity of onco-retroviral and lentiviral gene switch into main mouse and human B-lymphocytes is pseudotype dependent. Hum. Gene Ther. 14, 263–276 (2003).
Frecha, C. et al. Environment friendly and steady transduction of resting B lymphocytes and first continual lymphocyte leukemia cells utilizing measles virus gp displaying lentiviral vectors. Blood 114, 3173–3180 (2009).
Vamva, E. et al. A lentiviral vector B cell gene remedy platform for the supply of the anti-HIV-1 eCD4-Ig-knob-in-hole-reversed immunoadhesin. Mol. Ther. Strategies Clin. Dev. 28, 366–384 (2023).
Levy, C. et al. Baboon envelope pseudotyped lentiviral vectors effectively transduce human B cells and permit energetic issue IX B cell secretion in vivo in NOD/SCIDγc−/− mice. J. Thromb. Haemost. 14, 2478–2492 (2016).
Bender, R. R. et al. Receptor-targeted Nipah virus glycoproteins enhance cell-type selective gene supply and reveal a choice for membrane-proximal cell attachment. PLoS Pathog. 12, e1005641 (2016).
Hamilton, J. R. et al. In vivo human T cell engineering with enveloped supply automobiles. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-02085-z (2024).
Dobson, C. S. et al. Antigen identification and high-throughput interplay mapping by reprogramming viral entry. Nat. Strategies 19, 449–460 (2022).
Yu, B. et al. Engineered cell entry hyperlinks receptor biology with single-cell genomics. Cell 185, 4904–4920.e22 (2022).
Takano, Ok.-A. et al. Envelope protein-specific B cell receptors direct lentiviral vector tropism in vivo. Mol. Ther. 32, 1311–1327 (2024).
Ou, T. et al. Reprogramming of the heavy-chain CDR3 areas of a human antibody repertoire. Mol. Ther. 30, 184–197 (2022).
Johnson, M. J., Laoharawee, Ok., Lahr, W. S., Webber, B. R. & Moriarity, B. S. Engineering of main human B cells with CRISPR/Cas9 focused nuclease. Sci. Rep. 8, 12144 (2018).
Selvaraj, S. et al. Excessive-efficiency transgene integration by homology-directed restore in human main cells utilizing DNA-PKcs inhibition. Nat. Biotechnol. 42, 731–744 (2024).
Sheridan, C. B cells as drug factories. Nat. Biotechnol. 42, 823–826 (2024).
Hackett, P. B. & Essner, J. Integration-site directed vector techniques. US patent US7919583B2 (2005).
Laoharawee, Ok. et al. Genome engineering of main human B cells utilizing CRISPR/Cas9. J. Vis. Exp. https://doi.org/10.3791/61855 (2020).
Giguère, S. et al. Antibody manufacturing depends on the tRNA inosine wobble modification to satisfy biased codon demand. Science 383, 205–211 (2024).
Christie, S. M., Fijen, C., & Rothenberg, E. V(D)J recombination: current insights in formation of the recombinase advanced and recruitment of DNA restore equipment. Entrance. Cell Dev. Biol. 10, 886718 (2022).
Nahmad, A. D. et al. In vivo engineered B cells secrete excessive titers of broadly neutralizing anti-HIV antibodies in mice. Nat. Biotechnol. 40, 1241–1249 (2022).
Russell, D. M. et al. Peripheral deletion of self-reactive B cells. Nature 354, 308–311 (1991).
Abbott, R. Ok. et al. Precursor frequency and affinity decide B cell aggressive health in germinal facilities, examined with germline-targeting HIV vaccine immunogens. Immunity 48, 133–146.e6 (2018).
Dosenovic, P. et al. Anti–HIV-1 B cell responses are depending on B cell precursor frequency and antigen-binding affinity. Proc. Natl Acad. Sci. USA 115, 4743–4748 (2018).
Tokatlian, T. et al. Enhancing humoral responses in opposition to HIV envelope trimers through nanoparticle supply with stabilized artificial liposomes. Sci. Rep. 8, 16527 (2018).
Wagar, L. E. et al. Modeling human adaptive immune responses with tonsil organoids. Nat. Med. 27, 125–135 (2021).
Pan, A. et al. In vivo affinity maturation of the CD4 domains of an HIV-1-entry inhibitor. Nat. Biomed. Eng. 8, 1715–1729 (2024).
Buerstedde, J.-M., Alinikula, J., Arakawa, H., McDonald, J. J. & Schatz, D. G. Focusing on of somatic hypermutation by immunoglobulin enhancer and enhancer-like sequences. PLoS Biol. 12, e1001831 (2014).
O’Connor, B. P. et al. BCMA is crucial for the survival of long-lived bone marrow plasma cells. J. Exp. Med. 199, 91–98 (2004).
Wallweber, H. J. A., Compaan, D. M., Starovasnik, M. A. & Hymowitz, S. G. The crystal construction of a proliferation-inducing ligand. April. J. Mol. Biol. 343, 283–290 (2004).
Lapidot, T. Mechanism of human stem cell migration and repopulation of NOD/SCID and B2mnull NOD/SCID mice. Ann. N. Y. Acad. Sci. 938, 83–95 (2001).
Hargreaves, D. C. et al. A coordinated change in chemokine responsiveness guides plasma cell actions. J. Exp. Med. 194, 45–56 (2001).
Chatterjee, S., Behnam Azad, B. & Nimmagadda, S. The intricate function of CXCR4 in most cancers. Adv. Most cancers Res. 124, 31–82 (2014).
Vonderheide, R. H., Tedder, T. F., Springer, T. A. & Staunton, D. E. Residues inside a conserved amino acid motif of domains 1 and 4 of VCAM-1 are required for binding to VLA-4. J. Cell Biol. 125, 215–222 (1994).
Newham, P. et al. Α4 integrin binding interfaces on VCAM-1 and MAdCAM-1. J. Biol. Chem. 272, 19429–19440 (1997).
Benet, Z., Jing, Z. & Fooksman, D. R. Plasma cell dynamics within the bone marrow area of interest. Cell Rep. 34, 108733 (2021).
Nguyen, D. C. et al. Writer correction: components of the bone marrow microniche that assist human plasma cell survival and immunoglobulin secretion. Nat. Commun. 10, 372 (2019).
Roldán, E., García-Pardo, A. & Brieva, J. A. VLA-4-fibronectin interplay is required for the terminal differentiation of human bone marrow cells able to spontaneous and excessive fee immunoglobulin secretion. J. Exp. Med. 175, 1739–1747 (1992).
Fiorillo, M. T., Cabibbo, A., Iacopetti, P., Fattori, E. & Ciliberto, G. Evaluation of human/mouse interleukin-6 hybrid proteins: each amino and carboxy termini of human interleukin-6 are required for in vitro receptor binding. Eur. J. Immunol. 22, 2609–2615 (1992).
Neuber, T. et al. Characterization and screening of IgG binding to the neonatal Fc receptor. MAbs 6, 928–942 (2014).
Ober, R. J., Radu, C. G., Ghetie, V. & Ward, E. S. Variations in promiscuity for antibody-FcRn interactions throughout species: implications for therapeutic antibodies. Int. Immunol. 13, 1551–1559 (2001).
Andersen, J. T., Daba, M. B., Berntzen, G., Michaelsen, T. E. & Sandlie, I. Cross-species binding analyses of mouse and human neonatal Fc receptor present dramatic variations in immunoglobulin G and albumin binding. J. Biol. Chem. 285, 4826–4836 (2010).
Li, F. et al. Mouse strains affect clearance and efficacy of antibody and antibody-drug conjugate through Fc-FcγR interplay. Mol. Most cancers Ther. 18, 780–787 (2019).
Oldham, R. J. et al. FcγRII (CD32) modulates antibody clearance in NOD SCID mice resulting in impaired antibody-mediated tumor cell deletion. J. Immunother. Most cancers 8, e000619 (2020).
Yu, H. et al. A novel humanized mouse mannequin with important enchancment of class-switched, antigen-specific antibody manufacturing. Blood 129, 959–969 (2017).
Li, Y. et al. A human immune system mouse mannequin with strong lymph node improvement. Nat. Strategies 15, 623–630 (2018).
Chupp, D. P. et al. A humanized mouse that mounts mature class-switched, hypermutated and neutralizing antibody responses. Nat. Immunol. https://doi.org/10.1038/s41590-024-01880-3 (2024).
Solar, Ok. & Liao, M. Z. Medical pharmacology concerns on recombinant adeno-associated virus-based gene remedy. J. Clin. Pharmacol. 62, S79–S94 (2022).
Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. H. Chimeric antigen receptor-modified T cells in continual lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).
Tsuchida, C. A. et al. Mitigation of chromosome loss in scientific CRISPR–Cas9-engineered T cells. Cell 186, 4567–4582.e20 (2023).
Lazar, N. H. et al. Excessive-resolution genome-wide mapping of chromosome-arm-scale truncations induced by CRISPR-Cas9 modifying. Nat. Genet. 56, 1482–1493 (2024).
Nahmad, A. D. et al. Frequent aneuploidy in main human T cells after CRISPR–Cas9 cleavage. Nat. Biotechnol. 40, 1807–1813 (2022).
Zhang, T.-T. et al. BCR signaling is required for posttransplant lymphoproliferative illness in immunodeficient mice receiving human B cells. Sci. Transl. Med. 16, eadh8846 (2024).
Stockfelt, M., Teng, Y. Ok. O. & Important, E. M. Alternatives and limitations of B cell depletion approaches in SLE. Nat. Rev. Rheumatol. 21, 111–126 (2025).
Gargett, T. & Brown, M. P. The inducible caspase-9 suicide gene system as a “safety switch†to restrict on-target, off-tumor toxicities of chimeric antigen receptor T cells. Entrance. Pharmacol. 5, 235 (2014).
Dunkelberger, J. R. & Track, W.-C. Complement and its function in innate and adaptive immune responses. Cell Res. 20, 34–50 (2010).
Ma, A. D. & Carrizosa, D. Acquired issue VIII inhibitors: pathophysiology and remedy. Hematol. Am. Soc. Hematol. Educ. Program 2006, 432–437 (2006).
Jawa, V. et al. T-cell dependent immunogenicity of protein therapeutics pre-clinical evaluation and mitigation-updated consensus and overview 2020. Entrance. Immunol. 11, 1301 (2020).
Sabatino, D. E. et al. Efficacy and security of long-term prophylaxis in extreme hemophilia A canines following liver gene remedy utilizing AAV vectors. Mol. Ther. 19, 442–449 (2011).
Annoni, A. et al. Liver gene remedy by lentiviral vectors reverses anti-factor IX pre-existing immunity in haemophilic mice. EMBO Mol. Med. 5, 1684–1697 (2013).
Chand, D. et al. Hepatotoxicity following administration of onasemnogene abeparvovec (AVXS-101) for the remedy of spinal muscular atrophy. J. Hepatol. 74, 560–566 (2021).
Manno, C. S. et al. Profitable transduction of liver in hemophilia by AAV-Issue IX and limitations imposed by the host immune response. Nat. Med. 12, 342–347 (2006).
Gallo-Penn, A. M. et al. Systemic supply of an adenoviral vector encoding canine issue VIII leads to short-term phenotypic correction, inhibitor improvement, and biphasic liver toxicity in hemophilia A canines. Blood 97, 107–113 (2001).
Grauwet, Ok. et al. Stealth transgenes allow CAR-T cells to evade host immune responses. J. Immunother. Most cancers 12, e008417 (2024).
Wang, B. et al. Era of hypoimmunogenic T cells from genetically engineered allogeneic human induced pluripotent stem cells. Nat. Biomed. Eng. 5, 429–440 (2021).
Hu, X. et al. Hypoimmune induced pluripotent stem cells survive long run in absolutely immunocompetent, allogeneic rhesus macaques. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01784-x (2023).
Gupta, P., Alheib, O. & Shin, J.-W. In direction of single cell encapsulation for precision biology and drugs. Adv. Drug Deliv. Rev. 201, 115010 (2023).
Kershaw, M. H. et al. A section I research on adoptive immunotherapy utilizing gene-modified T cells for ovarian most cancers. Clin. Most cancers Res. 12, 6106–6115 (2006).
Hartweger, H. et al. Gene modifying of main rhesus macaque B cells. J. Vis. Exp. https://doi.org/10.3791/64858 (2023).
Vamva, E. et al. An optimized measles virus glycoprotein-pseudotyped lentiviral vector manufacturing system to advertise environment friendly transduction of human main B cells. STAR Protoc. 3, 101228 (2022).
Yu-Hong Cheng, R. et al. Era, enlargement, gene supply, and single-cell profiling in rhesus macaque plasma B cells. Cell Rep. Strategies 4, 100878 (2024).
Ishikawa, M. et al. Bone marrow plasma cells require P2RX4 to sense extracellular ATP. Nature 626, 1102–1107 (2024).
Van Dam, M. et al. Construction–perform evaluation of interleukin-6 using human/murine chimeric molecules. Involvement of two separate domains in receptor binding. J. Biol. Chem. 268, 15285–15290 (1993).