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:

Dual CD4-based CAR T cells with distinct costimulatory domains mitigate HIV pathogenesis in vivo

Abstract

An effective strategy to cure HIV will likely require a potent and sustained antiviral T cell response. Here we explored the utility of chimeric antigen receptor (CAR) T cells, expressing the CD4 ectodomain to confer specificity for the HIV envelope, to mitigate HIV-induced pathogenesis in bone marrow, liver, thymus (BLT) humanized mice. CAR T cells expressing the 4-1BB/CD3-ζ endodomain were insufficient to prevent viral rebound and CD4+ T cell loss after the discontinuation of antiretroviral therapy. Through iterative improvements to the CAR T cell product, we developed Dual-CAR T cells that simultaneously expressed both 4-1BB/CD3-ζ and CD28/CD3-ζ endodomains. Dual-CAR T cells exhibited expansion kinetics that exceeded 4-1BB-, CD28- and third-generation costimulated CAR T cells, elicited effector functions equivalent to CD28-costimulated CAR T cells and prevented HIV-induced CD4+ T cell loss despite persistent viremia. Moreover, when Dual-CAR T cells were protected from HIV infection through expression of the C34-CXCR4 fusion inhibitor, these cells significantly reduced acute-phase viremia, as well as accelerated HIV suppression in the presence of antiretroviral therapy and reduced tissue viral burden. Collectively, these studies demonstrate the enhanced therapeutic potency of a novel Dual-CAR T cell product with the potential to effectively treat HIV infection.

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: BLT mouse-derived HIV-specific CAR T cells are functionally indistinguishable from human-derived CAR T cells in vitro.
Fig. 2: CAR T cells expressing the 4-1BB costimulatory domain exhibit a proliferative advantage and induce B cell aplasia in vivo.
Fig. 3: HIV-specific CAR.BBζ T cells display features of T cell exhaustion after failing to control viral rebound.
Fig. 4: Dual-CAR TCP mitigates CD4+ T cell loss and exhibits superior proliferative capacity.
Fig. 5: HIV-resistant Dual-CAR T cells mediate superior virus-specific immune responses.
Fig. 6: Mitigating CAR T cell infection improves control over HIV replication.

Similar content being viewed by others

Data availability

All of the construct sequences described in this article are published (see ref. 26). All materials described in this manuscript are available via a material transfer agreement with the University of Pennsylvania or the Ragon Institute. All data are available from the corresponding authors upon request. Source data are provided with this paper.

References

  1. June, C. H. & Sadelain, M. Chimeric antigen receptor therapy. N. Engl. J. Med. 379, 64–73 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Walker, B. & McMichael, A. The T-cell response to HIV. Cold Spring Harb. Perspect. Med. 2, 1–19 (2012).

    Article  CAS  Google Scholar 

  3. Gross, G., Waks, T. & Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl Acad. Sci. USA 86, 10024–10028 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. van der Stegen, S. J., Hamieh, M. & Sadelain, M. The pharmacology of second-generation chimeric antigen receptors. Nat. Rev. Drug Disco. 14, 499–509 (2015).

    Article  CAS  Google Scholar 

  5. Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).

    PubMed  PubMed Central  Google Scholar 

  6. Porter, D. L. et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl. Med. 7, 303ra139 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Brentjens, R. J. et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin. Cancer Res. 13, 5426–5435 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Song, D. G. et al. In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4-1BB). Cancer Res. 71, 4617–4627 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Carpenito, C. et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc. Natl Acad. Sci. USA 106, 3360–3365 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Milone, M. C. et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. 17, 1453–1464 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cheng, Z. et al. In vivo expansion and antitumor activity of coinfused CD28- and 4-1BB-engineered CAR-T cells in patients with B cell leukemia. Mol. Ther. 26, 976–985 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Quintarelli, C. et al. Choice of costimulatory domains and of cytokines determines CAR T-cell activity in neuroblastoma. Oncoimmunology 7, e1433518 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity 44, 380–390 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Neelapu, S. S. et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 377, 2531–2544 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Salter, A. I. et al. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci. Signal. 11, eaat6753 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Schuster, S. J. et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N. Engl. J. Med. 380, 45–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Abate-Daga, D. & Davila, M. L. CAR models: next-generation CAR modifications for enhanced T-cell function. Mol. Ther. Oncolytics 3, 16014 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Weinkove, R., George, P., Dasyam, N. & McLellan, A. D. Selecting costimulatory domains for chimeric antigen receptors: functional and clinical considerations. Clin. Transl. Immunol. 8, e1049 (2019).

    Article  Google Scholar 

  20. Deeks, S. G. et al. A phase II randomized study of HIV-specific T-cell gene therapy in subjects with undetectable plasma viremia on combination antiretroviral therapy. Mol. Ther. 5, 788–797 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Colovos, C., Villena-Vargas, J. & Adusumilli, P. S. Safety and stability of retrovirally transduced chimeric antigen receptor T cells. Immunotherapy 4, 899–902 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Scholler, J. et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci. Transl. Med. 4, 132ra153 (2012).

    Article  Google Scholar 

  23. Kim, G. B., Hege, K. & Riley, J. L. CAR talk: how cancer-specific CAR T cells can instruct how to build CAR T cells to cure HIV. Front. Immunol. 10, 2310 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ali, A. et al. HIV-1-specific chimeric antigen receptors based on broadly neutralizing antibodies. J. Virol. 90, 6999–7006 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hale, M. et al. Engineering HIV-resistant, anti-HIV chimeric antigen receptor T cells. Mol. Ther. 25, 570–579 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Leibman, R. S. et al. Supraphysiologic control over HIV-1 replication mediated by CD8 T cells expressing a re-engineered CD4-based chimeric antigen receptor. PLoS Pathog. 13, e1006613 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Zhen, A. et al. Long-term persistence and function of hematopoietic stem cell-derived chimeric antigen receptor T cells in a nonhuman primate model of HIV/AIDS. PLoS Pathog. 13, e1006753 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Anthony-Gonda, K. et al. Multispecific anti-HIV duoCAR-T cells display broad in vitro antiviral activity and potent in vivo elimination of HIV-infected cells in a humanized mouse model. Sci. Transl. Med. 11, eaav5685 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Herzig, E. et al. Attacking latent HIV with convertibleCAR-T cells, a highly adaptable killing platform. Cell 179, e810 (2019).

    Article  CAS  Google Scholar 

  30. Sun, Z. et al. Intrarectal transmission, systemic infection, and CD4+ T cell depletion in humanized mice infected with HIV-1. J. Exp. Med. 204, 705–714 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Denton, P. W. et al. Antiretroviral pre-exposure prophylaxis prevents vaginal transmission of HIV-1 in humanized BLT mice. PLoS Med. 5, e16 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Brainard, D. M. et al. Induction of robust cellular and humoral virus-specific adaptive immune responses in human immunodeficiency virus-infected humanized BLT mice. J. Virol. 83, 7305–7321 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhen, A. et al. Targeting type I interferon-mediated activation restores immune function in chronic HIV infection. J. Clin. Invest. 127, 260–268 (2017).

    Article  PubMed  Google Scholar 

  34. Boritz, E. A. et al. Multiple origins of virus persistence during natural control of HIV infection. Cell 166, 1004–1015 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Estes, J. D. et al. Defining total-body AIDS-virus burden with implications for curative strategies. Nat. Med. 23, 1271–1276 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ventura, J. D. et al. Longitudinal bioluminescent imaging of HIV-1 infection during antiretroviral therapy and treatment interruption in humanized mice. PLoS Pathog. 15, e1008161 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Seo, H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410–12415 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yao, C. et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 20, 890–901 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Appay, V. et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat. Med. 8, 379–385 (2002).

    Article  CAS  PubMed  Google Scholar 

  43. Buggert, M. et al. T-bet and Eomes are differentially linked to the exhausted phenotype of CD8+ T cells in HIV infection. PLoS Pathog. 10, e1004251 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Leslie, G. J. et al. Potent and broad inhibition of HIV-1 by a peptide from the gp41 heptad repeat-2 domain conjugated to the CXCR4 amino terminus. PLoS Pathog. 12, e1005983 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Zhao, Z. et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell 28, 415–428 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Guedan, S. et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 3, e96976 (2018).

    Article  PubMed Central  Google Scholar 

  47. Pule, M. A. et al. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol. Ther. 12, 933–941 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Hombach, A. A., Heiders, J., Foppe, M., Chmielewski, M. & Abken, H. OX40 costimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirected CD4+ T cells. Oncoimmunology 1, 458–466 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Moir, S., Chun, T. W. & Fauci, A. S. Pathogenic mechanisms of HIV disease. Annu. Rev. Pathol. 6, 223–248 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Perez, E. E. et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat. Biotechnol. 26, 808–816 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Holt, N. et al. Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat. Biotechnol. 28, 839–847 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Tebas, P. et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N. Engl. J. Med. 370, 901–910 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Peterson, C. W. et al. Differential impact of transplantation on peripheral and tissue-associated viral reservoirs: implications for HIV gene therapy. PLoS Pathog. 14, e1006956 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Claiborne, D. T. et al. Replicative fitness of transmitted HIV-1 drives acute immune activation, proviral load in memory CD4+ T cells, and disease progression. Proc. Natl Acad. Sci. USA 112, E1480–E1489 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wright, J. K. et al. Influence of Gag-protease-mediated replication capacity on disease progression in individuals recently infected with HIV-1 subtype C. J. Virol. 85, 3996–4006 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Prince, J. L. et al. Role of transmitted Gag CTL polymorphisms in defining replicative capacity and early HIV-1 pathogenesis. PLoS Pathog. 8, e1003041 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Deymier, M. J. et al. Heterosexual transmission of subtype C HIV-1 selects consensus-like variants without increased replicative capacity or interferon-ɑ resistance. PLoS Pathog. 11, e1005154 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Seung, E. & Tager, A. M. Humoral immunity in humanized mice: a work in progress. J. Infect. Dis. 208(Suppl 2), S155–S159 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Martinez-Torres, F., Nochi, T., Wahl, A., Garcia, J. V. & Denton, P. W. Hypogammaglobulinemia in BLT humanized mice—an animal model of primary antibody deficiency. PLoS ONE 9, e108663 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Karpel, M. E., Boutwell, C. L. & Allen, T. M. BLT humanized mice as a small animal model of HIV infection. Curr. Opin. Virol. 13, 75–80 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Dudek, T. E. et al. Rapid evolution of HIV-1 to functional CD8+ T cell responses in humanized BLT mice. Sci. Transl. Med. 4, 143ra198 (2012).

    Article  CAS  Google Scholar 

  62. Claiborne, D. T. et al. Immunization of BLT humanized mice redirects T cell responses to Gag and reduces acute HIV-1 viremia. J. Virol. 93, e00814–e00819 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Pardi, N. et al. Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat. Commun. 8, 14630 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Boutwell, C. L., Rowley, C. F. & Essex, M. Reduced viral replication capacity of human immunodeficiency virus type 1 subtype C caused by cytotoxic-T-lymphocyte escape mutations in HLA-B57 epitopes of capsid protein. J. Virol. 83, 2460–2468 (2009).

    Article  CAS  PubMed  Google Scholar 

  65. Johnson, D. et al. Expression and structure of the human NGF receptor. Cell 47, 545–554 (1986).

    Article  CAS  PubMed  Google Scholar 

  66. Wang, X. et al. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood 118, 1255–1263 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Brelot, A., Heveker, N., Montes, M. & Alizon, M. Identification of residues of CXCR4 critical for human immunodeficiency virus coreceptor and chemokine receptor activities. J. Biol. Chem. 275, 23736–23744 (2000).

    Article  CAS  PubMed  Google Scholar 

  68. Richardson, M. W. et al. Mode of transmission affects the sensitivity of human immunodeficiency virus type 1 to restriction by rhesus TRIM5ɑ. J. Virol. 82, 11117–11128 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Clayton, K. L. et al. Resistance of HIV-infected macrophages to CD8+ T lymphocyte-mediated killing drives activation of the immune system. Nat. Immunol. 19, 475–486 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hindson, B. J. et al. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal. Chem. 83, 8604–8610 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. Ellebrecht and A. Payne (University of Pennsylvania) for the HIVYU2 Envelope-transduced K562 cell line. We thank W. Garcia-Beltran for artistic contributions. This study was supported by NIH grants P01HL129903 (T.M.A.), U19AI117950 (J.L.R.) and UM1AI126620 (J.L.R.) funded by NIAID, NIDA, NIMH and NINDS; T32 grant AI007632 (C.R.M.); and an NIAID F32 grant AI136750 (D.T.C.). We also acknowledge support from the Ragon Institute of MGH, MIT and Harvard and the MGH Scholars program (T.M.A.). We thank the Human Immune System Mouse Core at the Ragon Institute of MGH, MIT and Harvard and the Stem Cell and Xenograft Core at the University of Pennsylvania for the generation of BLT humanized mice, the Penn Center for AIDS Research (P30-AI045008) for purified human T cells, and the Allen and Riley laboratories for helpful comments.

Author information

Authors and Affiliations

Authors

Contributions

C.R.M., D.T.C., K.O., C.L.B., J.L.R. and T.M.A. conceived and designed the project and contributed to the interpretation of data. C.R.M., D.T.C., K.O., T.C., D.L.D., X.S., K.A.P., R.T.T., K.K., M.P., V.D.V., S.T., T.B., G.J.L. and J.A.H. contributed to the acquisition and analysis of data. C.R.M., D.T.C., C.L.B., J.L.R. and T.M.A. drafted the manuscript.

Corresponding authors

Correspondence to James L. Riley or Todd M. Allen.

Ethics declarations

Competing interests

C.R.M. and J.L.R. have filed an institution-owned patent (20180265565: Method of Redirecting T Cells to Treat HIV Infection) describing the construction of these HIV-specific CARs specific to Figs. 1 and 2. This patent application has been published but has not yet been granted. C.R.M. and J.L.R. have also filed an institution-owned patent (Dual CAR Expressing T Cells Individually Linked to CD28 and 4-1BB) specific to Figs. 4–6. G.J.L., J.A.H. and J.L.R. have also filed an institution-owned patent (Non-Signaling HIV Fusion Inhibitors And Methods Of Use Thereof) specific to Figs. 5 and 6. J.L.R. cofounded a company called Tmunity Therapeutics that has the rights to license the technology described in this paper. J.L.R. holds an equity interest in Tmunity. C.R.M. and J.L.R. declare no other competing financial interests. No other authors declare any competing financial interests. No authors declare any nonfinancial interests.

Additional information

Peer review information Alison Farrell was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 CD28 costimulation enhances the ex vivo effector function of CAR T cells.

HIV-uninfected mice were infused with an equal mixture of CD4-based CAR T cells expressing either CD3-ζ, 4-1BB/CD3-ζ and CD28/CD3-ζ costimulatory domains linked to unique fluorescent proteins to facilitate identification in vivo as described in Fig. 2 legend. Cumulative data indicating the frequency of TNF+, IL-2+ and MIP-1β+ CAR.BBζ and CAR.28ζ T cells within the same mice after ex vivo stimulation with K.Env (+) or K.WT (-) cells. Data represents the aggregate of cytokine producing cells from liver and terminal blood (n = 8). CAR.ζ T cells were too infrequent for analysis. Data shows box and whisker plots where the middle line indicates median, bounds of the box show 25th to 75th percentiles, and bars extend to min and max values. Symbols represent biologically independent animals. Significance was calculated using two-sided Wilcoxon matched-pairs signed rank test.

Source data

Extended Data Fig. 2 CAR.BBζ T cells accumulate multiple inhibitory receptors as disease progresses.

a, Frequency of CD4+ and (b) CD8+ CAR.BBζ T cells (G1; n = 6) and CAR.BBΔζ T cells (G2; n = 6) co-expressing TIGIT and PD-1 after infusion. Shaded box indicates the window of ART. Symbols and error bars indicate mean ± SEM. c, Frequency of CD4+ and (d) CD8+ CAR.BBζ T cells (G1) and CAR.BBΔζ T cells (G2) co-expressing TIGIT, PD-1 and 2B4 in tissues 12 weeks post-infusion. e, Cumulative data indicating the frequency of 2B4+, PD-1+ and TIGIT+ CD4+ CAR.BBζ T cells (G1) compared to CAR- CD4+ T cells (G1) within the spleens of the same mice, and (f) CD8+ CAR.BBζ T cells (G1) compared to CAR- CD8+ T cells (G1) within the spleens of the same mice. c–f, Bars indicate mean, error bars show ± SEM and symbols represent individual mice. Significance was calculated using two-sided Wilcoxon rank-sum test. Sample sizes in these studies represent biologically independent animals.

Source data

Extended Data Fig. 3 EomeshiT-betdim CAR.BBζ T cells accumulate from acute to chronic phases of infection.

BLT mice were infected with HIVJRCSF and infused 48 h later with either 2×107 CAR.BBζ T cells (n = 5) or inactive control CAR.BBΔζ T cells (n = 3). a, FACS plots show the change in Eomes and T-bet expression within the different CAR T cell types over time. b, Summary data indicating the longitudinal frequency of EomeshiT-betdim CD8+ (left panel) and CD4+ (right panel) CAR T cells (left y-axis), and mean log plasma HIV RNA (copies mL−1) (right y-axis). Thin dotted line denotes limit of viral load quantification. Symbols and error bars indicate mean ± SEM. c, Spearman correlation analysis of frequency of EomeshiT-betdim CD8+ CAR.BBζ T cells compared with viral burden measured as the frequency of HIVGAG+ CD8- T cells in various tissues 10 weeks post-infection. Sample sizes in these studies indicate biologically independent animals.

Source data

Extended Data Fig. 4 Dual-CAR T cell product transiently delays CD4+ T cell loss despite persistent HIVJRCSF infection.

BLT mice received Dual-CAR T cell product (TCP) (n = 6) 48 h after HIVJRCSF challenge, while control mice were untreated (Untx) (n = 5). a, Concentration of peripheral total memory CD4+ T cells (CAR). b, Concentration of peripheral central memory (CD45RACD27+CCR7+; left panel), transitional memory (CD45RA-CD27+CCR7; middle panel), and effector memory (CD45RA-CD27-CCR7-; right panel) CD4+ T cells (CAR-). c, Frequency of memory CD4+ T cell (CAR-) subsets in tissues 8 weeks post-infection. a, b, Significance was calculated using a two-sided Wilcoxon rank-sum test. Symbols and bars indicate mean, and error bars show ± SEM. Sample sizes indicate biologically independent animals.

Source data

Extended Data Fig. 5 Dual-CAR T cell product prevents CD4+ T cell loss despite persistent HIVMJ4 infection.

BLT mice were infused with Dual-CAR T cell product (TCP) (n = 6) 48 h post-HIVMJ4 challenge, while control mice were untreated (Untx) (n = 6). a, Concentration of peripheral total memory CD4+ T cells (CAR). b, Concentration of peripheral central memory (CD45RA-CD27+CCR7+; right panel), transitional memory (CD45RACD27+CCR7-; middle panel), and effector memory (CD45RA-CD27-CCR7-; left panel) CD4+ T cells (CAR-). c, Frequency of memory CD4+ T cell (CAR-) subsets in tissues 8 weeks post-infection. a, b, Significance was calculated using a two-sided Wilcoxon rank-sum test. Symbols and bars indicate mean, while error bars show ± SEM. Sample sizes indicate biologically independent animals.

Source data

Extended Data Fig. 6 Dual-CAR T cells exhibit superior in vivo expansion compared to 4-1BB-costimulated, CD28-costimulated, and 3rd-generation CAR T cells.

a, BLT mice were challenged with either HIVJRCSF (n = 6) or HIVMJ4 (n = 6) and infused with 2×107 Dual-CAR T cell product (TCP). Fold-change in CAR T cell concentration from baseline to peak levels in peripheral blood. Data is the aggregate of both infection cohorts. b, Schematic shows the structural components of the 3rd-generation (3 G) CD4-based CAR construct. c–e, Dual-CAR T cell product and 3G-CAR T cells were combined at an equal frequency prior to infusion into uninfected mice (n = 9). c, FACS plots indicate the frequency of Dual-CAR and 3G-CAR T cells present within the pre-infusion T cell product. d, Longitudinal concentration of peripheral CAR T cells following adoptive transfer into HIV-negative mice. Symbols and error bars indicate mean ± SEM. e, At 2 weeks post-infusion, mice received either 107 irradiated K.Env cells (n = 6) or 107 irradiated K.WT cells (n = 3). Fold change in the concentration of peripheral CAR T cells 1-week post-K562 infusion from baseline concentration prior to K562 infusion. a, e, Bar and error bars indicate mean ± SEM, and symbols represent individual mice. a, d, e, Two-sided Wilcoxon rank-sum test was used to calculate significance. Sample sizes in these studies indicate biologically independent animals.

Source data

Extended Data Fig. 7 C34-CXCR4+ CAR T cells are selected for during chronic infection and exhibit superior ex vivo effector functions.

a, BLT mice were infected with HIVJRCSF and 48 h later infused with 107 C34-CXCR4+ Dual-CAR T cell product (TCP). FACS plots indicate the frequency of C34-CXCR4+ throughout infection. b, Mice were infected with HIVMJ4 and 48 h later were infused with 106 C34-CXCR4+ CAR.BBζ (n = 5), CAR.28ζ (n = 5), or purified Dual-CAR (n = 4) T cells. Frequency of C34-CXCR4+ CAR T cells in tissue 8 weeks post-infection. Thin dotted line indicates the frequency of C34-CXCR4+ CAR T cells in the pre-infusion TCP for the indicated CAR T cell type. Line and error bars indicate mean ± SEM. c, d, Mice were infected with HIVMJ4 and 48 h later received 106 C34-CXCR4+, purified CAR.BBζ.BBζ (n = 3), CAR.28ζ.28ζ (n = 4), or Dual-CAR (n = 3) T cells. c, FACS plots and (d) cumulative data show the frequency of each CD8+ CAR T cell population expressing MIP-1β and CD107a, and the frequency of CAR T cells with cytotoxic potential (granzyme B+ perforin+ CD107a+). CAR T cells were isolated from the spleen and bone marrow of mice 8 weeks post-infection and ex vivo stimulated. Significance was calculated using two-sided Wilcoxon matched-pairs signed rank test. For all data, symbols represent individual mice. Sample sizes in these studies indicate biologically independent animals.

Source data

Extended Data Fig. 8 CAR T cells from HIV-infected mice exhibit ex vivo cytotoxic function.

HIVJRCSF-infected mice (n = 3) treated with the Dual-CAR TCP were euthanized and the bone marrow cells were ex vivo stimulated with K.Env or K.WT cells for 24 h at the indicated E:T ratios. a, Representative FACS plots and (b) cumulative data shows the induction of active caspase-3 within the different target cell populations. Symbols and error bars indicate mean ± SEM. Sample size indicates biologically independent animals.

Source data

Extended Data Fig. 9 Dual-CAR and CAR.28ζ T cells exhibit similar ex vivo functional profiles.

BLT mice were challenged with HIVJRCSF (n = 5) and infused with 2×107 Dual-CAR T cell product (TCP) 48-hours post infection. a, Frequency of CD8+ and (b) CD4+ CAR T cell populations from tissue at necropsy (8-weeks post-infection) within the same mice expressing CD107a, MIP-1β, IL-2 and TNF after ex vivo stimulation. Bars and error bars indicate mean ± SEM, and symbols represent individual mice. Significance was calculated using two-sided Wilcoxon rank-sum test. c, Principle Components Analysis (PCA) of IL-2, TNF, MIP-1β, and CD107a expression in ex vivo stimulated CD8+ and CD4+ CAR T cells from PBMCs of HIVJRCSF-infected mice (n = 5). Sample sizes in these studies represent biologically independent animals.

Source data

Extended Data Fig. 10 HIV-resistant Dual-CAR TCP reduces virus replication in vivo.

a, Frequency of HIVGAG+ CD8- T cells (CAR-) within the bone marrow and spleen of HIVJRCSF-infected mice (n = 6) and (b) HIVMJ4-infected mice (n = 6) that were treated 48 h post-challenge with the Dual-CAR T cell product (TCP) or were untreated (Untx). c, Mean log plasma HIVMJ4 RNA (copies mL−1) after ART discontinuation of mice infused at ART initiation with 107 protected >98% C34-CXCR4+ (n = 5) or partially-protected <20% C34-CXCR4+ (n = 7) Dual-CAR TCP, or were untreated (n = 9). d, e, HIVBAL-infected mice were ART-treated and simultaneously infused with 107 HIV-resistant Dual-CAR TCP (n = 6) or were untreated (n = 6). d, Mean log plasma HIV RNA (copies mL−1). Shaded box indicates ART and arrow indicates CAR TCP infusion. e, Percent log reduction in plasma HIV RNA from pre-ART (week 3) and 0.5 and 1 week post-ART. For all data, bars and error bars indicate mean ± SEM, and symbols represent individual mice. Significance was calculated for (a–d) by two-sided Wilcoxon rank-sum test and (e) two-sided Kolmogorov-Smirnov test. Sample sizes in these studies indicate biologically independent animals.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–12.

Reporting Summary

Supplementary Data 1

Source Data for all Supplementary Figures

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 9

Statistical Source Data

Source Data Extended Data Fig. 10

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maldini, C.R., Claiborne, D.T., Okawa, K. et al. Dual CD4-based CAR T cells with distinct costimulatory domains mitigate HIV pathogenesis in vivo. Nat Med 26, 1776–1787 (2020). https://doi.org/10.1038/s41591-020-1039-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-1039-5

This article is cited by

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