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-targeting CAR-T cells with optimal co-stimulation and metabolic fitness enhance antitumor activity and prevent escape in solid tumors

Abstract

Chimeric antigen receptor (CAR)-T cells showed great activity in hematologic malignancies. However, heterogeneous antigen expression in tumor cells and suboptimal CAR-T-cell persistence remain critical aspects to achieve clinical responses in patients with solid tumors. Here we show that CAR-T cells targeting simultaneously two tumor-associated antigens and providing trans-acting CD28 and 4-1BB co-stimulation, while sharing the same CD3ζ-chain cause rapid antitumor effects in in vivo stress conditions, protection from tumor re-challenge and prevention of tumor escape due to low antigen density. Molecular and signaling studies indicate that T cells engineered with the proposed CAR design demonstrate sustained phosphorylation of T-cell-receptor-associated signaling molecules and a molecular signature supporting CAR-T-cell proliferation and long-term survival. Furthermore, metabolic profiling of CAR-T cells displayed induction of glycolysis that sustains rapid effector T-cell function, but also preservation of oxidative functions, which are critical for T-cell long-term persistence.

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: Single- or dual-antigen targeting and single or dual CD28 or 4-1BB co-stimulation do not eradicate tumors in stress conditions.
Fig. 2: One single shared CD3ζ chain is sufficient for transducing the activation signal in dual specific CAR-T cells.
Fig. 3: CD3ζ sharing in dual CAR relies on CD8α-mediated dimerization.
Fig. 4: Dual targeting with split co-stimulation and shared single CD3ζ promotes sustained antitumor activity.
Fig. 5: MSLN and CSPG4 dual-targeting CAR-T cells with split co-stimulation and shared CD3ζ show sustained T-cell activation and proliferation in vitro and in vivo.
Fig. 6: Dual targeting with split co-stimulation and shared D3ζ promote TCR tonic signaling.
Fig. 7: Dual targeting with split co-stimulation and shared CD3ζ promote CAR-T-cell proliferation and glycolytic and oxidative metabolism.
Fig. 8: Dual targeting, split signaling and one single CD3ζ endodomain prevent tumor escape due to antigen loss.

Similar content being viewed by others

Data availability

Source data for this study have been provided as source data files. RNA-seq datasets generated and analyzed during the current study are not publicly available (the genetic information from primary human T cells in this study was not consented to be published in the public domain) and will be available from corresponding authors upon request. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Fucà, G., Reppel, L., Landoni, E., Savoldo, B. & Dotti, G. Enhancing chimeric antigen receptor T-cell efficacy in solid tumors. Clin. Cancer Res. 26, 2444–2451 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Grada, Z. et al. TanCAR: a novel bispecific chimeric antigen receptor for cancer immunotherapy. Mol. Ther. Nucleic Acids 2, e105 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Ahn, S. et al. Cancer immunotherapy with T cells carrying bispecific receptors that mimic antibodies. Cancer Immunol. Res. 7, 773–783 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zah, E., Lin, M. Y., Silva-Benedict, A., Jensen, M. C. & Chen, Y. Y. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol. Res. 4, 498–508 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Qin, H. et al. Preclinical development of bivalent chimeric antigen receptors targeting both CD19 and CD22. Mol. Ther. Oncolytics 11, 127–137 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wang, G. et al. Fully human antibody VH domains to generate mono and bispecific CAR to target solid tumors. J. Immunother. Cancer https://doi.org/10.1136/jitc-2020-002173 (2021).

  8. Miller, B. R. et al. Stability engineering of scFvs for the development of bispecific and multivalent antibodies. Protein Eng. Des. Sel. 23, 549–557 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Maude, S. L., Teachey, D. T., Porter, D. L. & Grupp, S. A. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 125, 4017–4023 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Brentjens, R. J. et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl. Med. 5, 177ra138 (2013).

    Article  CAS  Google Scholar 

  11. 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 

  12. Sun, C. et al. THEMIS-SHP1 recruitment by 4-1BB tunes LCK-mediated priming of chimeric antigen receptor-redirected T cells. Cancer Cell 37, 216–225 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. 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 

  15. Ramos, C. A. et al. In vivo fate and activity of second- versus third-generation CD19-specific CAR-T cells in B cell non-Hodgkin’s lymphomas. Mol. Ther. 26, 2727–2737 (2018).

  16. Stephan, M. T. et al. T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection. Nat. Med. 13, 1440–1449 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Heczey, A. et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol. Ther. 25, 2214–2224 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Enblad, G. et al. A phase I/IIa trial using CD19-targeted third-generation CAR T cells for lymphoma and leukemia. Clin. Cancer Res. 24, 6185–6194 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Pule, M. A. et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat. Med. 14, 1264–1270 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yu, A. L. et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N. Engl. J. Med. 363, 1324–1334 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Du, H. et al. Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells. Cancer Cell 35, 221–237 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Castelletti, L., Yeo, D., van Zandwijk, N. & Rasko, J. E. J. Anti-mesothelin CAR T cell therapy for malignant mesothelioma. Biomark Res. 9, 11 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Landoni, E. et al. Modifications to the framework regions eliminate chimeric antigen receptor tonic signaling. Cancer Immunol. Res. https://doi.org/10.1158/2326-6066.Cir-20-0451 (2021).

  24. Beatty, G. L. et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol. Res. 2, 112–120 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Fujiwara, K. et al. Hinge and transmembrane domains of chimeric antigen receptor regulate receptor expression and signaling threshold. Cells https://doi.org/10.3390/cells9051182 (2020).

  26. Heczey, A. et al. Anti-GD2 CAR-NKT cells in patients with relapsed or refractory neuroblastoma: an interim analysis. Nat. Med. 26, 1686–1690 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Kramer, K. et al. Disialoganglioside G(D2) loss following monoclonal antibody therapy is rare in neuroblastoma. Clin. Cancer Res. 4, 2135–2139 (1998).

    CAS  PubMed  Google Scholar 

  28. Chen, Y. et al. Eradication of neuroblastoma by T cells redirected with an optimized GD2-specific chimeric antigen receptor and interleukin-15. Clin. Cancer Res. 25, 2915–2924 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Louis, C. U. et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118, 6050–6056 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Schumacher-Kuckelkorn, R. et al. Lack of immunocytological GD2 expression on neuroblastoma cells in bone marrow at diagnosis, during treatment, and at recurrence. Pediatr. Blood Cancer 64, 46–56 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Valitutti, S., Müller, S., Cella, M., Padovan, E. & Lanzavecchia, A. Serial triggering of many T-cell receptors by a few peptide-MHC complexes. Nature 375, 148–151 (1995).

    Article  CAS  PubMed  Google Scholar 

  32. Kofler, D. M. et al. CD28 costimulation Impairs the efficacy of a redirected T-cell antitumor attack in the presence of regulatory T cells which can be overcome by preventing Lck activation. Mol. Ther. 19, 760–767 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Morello, A., Sadelain, M. & Adusumilli, P. S. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 6, 133–146 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Pellegatta, S. et al. Constitutive and TNF-α-inducible expression of chondroitin sulfate proteoglycan 4 in glioblastoma and neurospheres: implications for CAR-T cell therapy. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aao2731 (2018).

  35. Geldres, C. et al. T lymphocytes redirected against the chondroitin sulfate proteoglycan-4 control the growth of multiple solid tumors both in vitro and in vivo. Clin. Cancer Res. 20, 962–971 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Salter, A. I. et al. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci. Signal https://doi.org/10.1126/scisignal.aat6753 (2018).

  37. 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 

  38. Sohara, Y. et al. Lytic bone lesions in human neuroblastoma xenograft involve osteoclast recruitment and are inhibited by bisphosphonate. Cancer Res. 63, 3026–3031 (2003).

    CAS  PubMed  Google Scholar 

  39. Song, L. et al. Vα24-invariant NKT cells mediate antitumor activity via killing of tumor-associated macrophages. J. Clin. Invest 119, 1524–1536 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Song, L. et al. Oncogene MYCN regulates localization of NKT cells to the site of disease in neuroblastoma. J. Clin. Invest. 117, 2702–2712 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hirschmann-Jax, C. et al. A distinct ‘side population’ of cells with high drug efflux capacity in human tumor cells. Proc. Natl Acad. Sci. USA 101, 14228–14233 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Di Paolo, D. et al. Neuroblastoma-targeted nanoparticles entrapping siRNA specifically knockdown ALK. Mol. Ther. 19, 1131–1140 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Vera, J. et al. T lymphocytes redirected against the kappa light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells. Blood 108, 3890–3897 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Diaconu, I. et al. Inducible caspase-9 selectively modulates the toxicities of CD19-specific chimeric antigen receptor-modified T cells. Mol. Ther. 25, 580–592 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, G. et al. Fully human antibody V(H) domains to generate mono and bispecific CAR to target solid tumors. J. Immunother. Cancer https://doi.org/10.1136/jitc-2020-002173 (2021).

  46. Xu, Y. et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 123, 3750–3759 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Imai, K., Wilson, B. S., Bigotti, A., Natali, P. G. & Ferrone, S. A 94,000-dalton glycoprotein expressed by human melanoma and carcinoma cells. J. Natl Cancer Inst. 68, 761–769 (1982).

    CAS  PubMed  Google Scholar 

  48. Hoyos, V. et al. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia 24, 1160–1170 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported in part by R01-CA193140-03 (G.D.) and R01-CA243543-01 (G.D.) from the National Cancer Institute (NCI). H.D. was supported by W81XWH-18-1-0441 from the Department of Defense (USA) and the Vicky Amidon Innovation Grant in Lung Cancer Research from the Lung Cancer Initiative of North Carolina (USA). The UNC Small Animal Imaging Facility at the Biomedical Imaging Research Center, the Microscopy Services Laboratory at Department of Pathology and Laboratory Medicine and the Flow Cytometry Core Facilities are supported in part by an NCI Cancer Center Core Support Grant to the UNC Lineberger Comprehensive Cancer Center (P30-CA016086-40) USA.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization was carried out by H.D. and G.D. Methodology was performed by K.H., H.D., Y.X., P.S., X.Z., B.S. and G.D. Investigations were conducted by K.H., H.D., Y.X., X.Z., P.S., G.F., E.L., C.S., Y.C., B.S. and G.D. Writing of the original draft was carried out by K.H., H.D., X.Y. and G.D. Review and editing was conducted by all authors. Supervision was carried by H.D. and G.D.

Corresponding authors

Correspondence to Hongwei Du or Gianpietro Dotti.

Ethics declarations

Competing interests

G. Dotti is a paid consultant for Bellicum Pharmaceuticals, Tessa Therapeutics and Catamaran and reports receiving commercial research grants from Cell Medica and Bluebird Bio; B. Savoldo is a paid consultant for Tessa Therapeutics; G. Dotti and H. Du filed a patent for the CAR targeting B7-H3. No other competing interests were disclosed by the other authors.

Additional information

Peer review information Nature Cancer thanks John Anderson and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 GD2-specific CAR-T cells and B7-H3-specific CAR-T cells target neuroblastoma in vitro.

(a) Flow cytometry histogram showing the expression of GD2 and B7-H3 in two human NB cell lines, CHLA-255 and LAN-1. Representative of three independent experiments. (b) Representative flow cytometry histograms showing the expression of CARs in human T cells transduced with retroviral vectors encoding CD19.28ζ, GD2.28ζ, GD2.BBζ, B7-H3.28ζ, and B7-H3.BBζ CARs. (c-e) Representative flow cytometry plots (c) and quantification of residual CHLA-255 (d) and LAN-1 (e) cells labelled with GFP and co-cultured with CAR-T cells at the T cell to tumor cell ratio of 1 to 5. On day 5, NB cells (GFP+) and CAR-T cells (CD3+) were enumerated by flow cytometry. Data are shown as individual values and the mean + SD, n = 6 independent co-cultures using CAR-T cells generated from 6 different donors. (f, g) Summary of IFN-γ (f) and IL-2 (g) released by CAR-T cells in the culture supernatant after 24 h of co-culture with NB cells as measured by ELISA. Data are shown as individual values and the mean + SD, n = 6 independent co-cultures using CAR-T cells generated from 6 different donors. (h) Representative CFSE dilution of CSFE-labeled CAR-T cells co-cultured with NB cells for 5 days at the T cell to tumor cell ratio of 1 to 1 (red histogram). CFSE-labeled CAR-T cell alone (grey histogram) was used as negative control. Representative of three independent experiments.

Source data

Extended Data Fig. 2 The antitumor activity of GD2-specific CAR-T cells and B7-H3-specific CAR-T cells with either CD28 or 4-1BB costimulation in vivo.

(a) Schema of the CHLA-255 metastatic xenograft NB model using NSG mice inoculated via tail vein injection with 2 × 106 of FFluc-CHLA-255 cells and 14 days later received high doses of CAR-T cells (6 × 106 cells/mouse) intravenously. (b, c) Representative tumor bioluminescence (BLI) images (b) and tumor BLI kinetics (c) of FFluc-CHLA-255 tumor growth (n = 3 mice for the CD19.28ζ group, n = 5 mice for the other four groups) in the metastatic xenograft NB models shown in (a). (d) Kaplan-Meier survival curve of mice in (b, c), n = 3 mice for CD19.28ζ group, n = 5 mice for other 4 groups, comparisons of survival curves were determined by Log-rank test, **p = 0.0042 for CD19.28ζ versus other 4 groups. (e) Schema of the LAN-1 metastatic xenograft NB model using NSG mice inoculated via tail vein injection with FFLuc-LAN-1 cells and treated 21 days later with low doses CD19.28ζ, GD2.28ζ, GD2.BBζ, B7-H3.28ζ or B7-H3.BBζ CAR-T cells intravenously. (f, g) Representative tumor BLI images (f) and tumor BLI kinetics (g) of FFLuc-LAN-1 tumor growth (n = 3 mice/group). (h) Kaplan-Meier survival curve of mice in (f, g), n = 3 mice/group, comparisons of survival curves were determined by Log-rank test, *p = 0.0253 for CD19.28ζ versus GD2.28ζ, GD2.BBζ and B7-H3.BBζ groups, *p = 0.0295 for GD2.28ζ versus GD2.BBζ, *p = 0.0246 for GD2.28ζ versus B7-H3.BBζ.

Source data

Extended Data Fig. 3 Addition of 4-1BB in tandem to the GD2.28ζ CAR and co-expression of both GD2.28ζ and B7-H3.BBζ CARs do not improve antitumor activity in vitro.

(a) Representative flow cytometry plots showing the CAR expression in human T cells transduced with retroviral vectors encoding CD19.28ζ, GD2.28ζ, GD2.28.BBζ, or GD2.28ζ/B7-H3.28ζ CARs. Representative of six independent experiments. (b, c) Representative flow cytometry plots (b) and quantification of residual CHLA-255 cells (c) labelled with GFP co-cultured with CAR-T cells at the T cell to tumor cell ratio of 1 to 5. Data are shown as individual values and the mean + SD, n = 6 or 8 independent co-cultures using CAR-T cells generated from 6 or 8 different donors. (d, e) Summary of IFN-γ (d) and IL-2 (e) released by CAR-T cells in the culture supernatant after 24 h of co-culture with NB cells as measured by ELISA. Data are shown as individual values and the mean + SD, n = 6 or 8 independent co-cultures using CAR-T cells generated from 6 or 8 different donors; **p = 0.0011, ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value.

Source data

Extended Data Fig. 4 Cytotoxic activity of the double CAR-T cells with shared CD3ζ is antigen dependent.

(a) Flow cytometry plots showing the expression of B7-H3 and GD2 in Raji cells wild type and B7-H3 expression in Raji cells transduced with a retroviral vector encoding B7-H3 (Raji-B7-H3). Representative of three independent experiments. (b–d) CAR-T cells (B7-H3.BB, B7-H3.BBζ, GD2.28ζ, GD2.28ζ/B7-H3.BB, dNGFR.28ζ/B7-H3.BB and 28ζ/B7-H3.BB) were co-cultured with Raji-B7-H3 cell at 1 to 1 ratio, and 5 days later tumor cells (CD19+) and T cells (CD3+) were collected and enumerated by flow cytometry (b). Supernatants of the co-cultures were collected 24 h later, and IFN-γ (c) and IL-2 (d) released by CAR-T cells were measured by ELISA. Data are shown as individual values and the mean + SD, n = 3 independent co-cultures using CAR-T cells generated from 3 different donors for dNGFR.28ζ/B7-H3.BB group, and n = 5 independent co-cultures using CAR-T cells generated from 5 different donors for all the other groups; *p <0.05 (0.0228 in c, 0.0141 in d), **p <0.01 (0.0025 in c, 0.0015 in d), ***p = 0.0005, ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value. (e-g) CAR-T cells (CD19.28ζ, GD2.28ζ/B7-H3.BB, dNGFR.28ζ/B7-H3.BB and 28ζ/B7-H3.BB) were co-cultured with Raji cell wild type at 1 to 1 ratio, and 5 days later tumor cells (CD19+) and T cells (CD3+) were collected and enumerated by flow cytometry (e). Supernatants of the co-cultures were collected 24 h later, and IFN-γ (f) and IL-2 (g) released by CAR-T cells were measured by ELISA. Data are shown as individual values and the mean + SD, n = 4 independent co-cultures using CAR-T cells generated from 4 different donors for each group; ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value.

Source data

Extended Data Fig. 5 CAR clustering and aggregation in CAR-T cells after CAR engagement.

Representative confocal microscopy imaging showing CAR molecule clustering in T cells expressing GFP-tagged GD2.28ζ (green) and B7-H3.BB (red) with and without engagement of the CARs using either the anti-14g2a idiotype antibody (1A7) or the B7-H3.Fc protein. Blue staining indicates the DAPI. Shown are representative cells of a single field (Magnification 63X). Data are representative of three independent validations. Shown in white are the scale bars that correspond to 20 µm.

Extended Data Fig. 6 Phenotypic analysis of CAR-T cells in vitro and in vivo.

(a, b) Frequency of CD45RA+CCR7+, CD45RA-CCR7+, CCR7-CD28+CD27+, CCR7-CD28+CD27-, CCR7-CD28-CD27+, and CCR7-CD28-CD27- in CD4+ (a) and CD8+ (b) T cells on day 13 after retroviral vector transduction and expansion in vitro. Data are shown as individual values and the mean + SD, n = 4 independent experiments using CAR-T cells generated from 4 different donors; *p = 0.0299, two-tailed p value determined by unpaired t test. (c–f) Tumor-baring mice infused with CAR-T cells were bled at day 14 and CAR-T cells in the peripheral blood were analyzed by flow cytometry. (c, d) Frequency of CD45RA+CCR7+, CD45RA-CCR7+, CCR7-CD28+CD27+, CCR7-CD28+CD27-, CCR7-CD28-CD27+, and CCR7-CD28-CD27- in CD4+ (c) and CD8+ (d) T cells. Data are shown as individual values and the mean + SD, n = 5 samples from 5 mice, *p < 0.05, two-tailed p value determined by unpaired t test. (e, f) Mean Fluorescence Intensity (MFI) of PD-1 (e) and TIM-3 (f) in T cells. Data are shown as individual values and the mean + SD, n = 5 samples from 5 mice; *p = 0.0109, ***p = 0.0008, two-tailed p value determined by unpaired t test.

Source data

Extended Data Fig. 7 Inverting the orientation of the B7-H3-specifc CAR and GD2-specific CAR does not alter the beneficial effects of dual targeting CAR-T cells with split costimulation and shared CD3ζ in vitro.

(a) Schematic representation of retroviral vectors encoding B7-H3.28ζ, GD2.BB and B7-H3.28ζ/GD2.BB CARs. (b) Representative flow cytometry plots of 5 independent experiments showing the expression of CARs. (c) Summary of the transduction efficiency of the CARs. Data are shown as individual values and the mean + SD, n = 5 or 7 independent experiments using CAR-T cells generated from 5 or 7 different donors; ***p = 0.002, ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value. (d–f) CAR-T cells were co-cultured with CHLA-255-GFP at T cell to tumor cell ratio of 1 to 5. IFN-γ (e) and IL-2 (f) released by CAR-T cells were measured by ELISA. On day 5, tumor cells (GFP+) and CAR-T cells (CD3+) number were measured by flow cytometry (d). Data are shown as individual values and the mean + SD, n = 3 independent co-cultures using CAR-T cells generated from 3 different donors ; *p <0.05, **p <0.01, ***p <0.001, ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value, the full list of p values can be found in the source data. (g) Schema of the repetitive multi-round co-culture experiments. Tumor cells were seeded in 24-well plates one day prior to the addition of T cells. At day 0, CAR-T cells were added at T cell to tumor cell ratio of 1 to 5. At day 4, 7, and 10, all T cells were collected and transferred into a new well in which 5 × 105 NB cells were seeded one day before. T cells and tumor cells, and cytokine were quantified at each cycle. (h–o) Multi-round co-culture with NB cell lines CHLA-255 (h–k) and LAN-1 (l–o) cells as described in (g). Summary of percentage of residual CHLA-255 (h) and LAN-1 (l) cells and number of T cells (i, m) at the end of each round of co-culture. Summary of IFN-γ (j, n) and IL-2 (k, o) released by CAR-T cells in the culture supernatant after 24 h of co-culture with CHLA-255 (j, k) and LAN-1 (n, o) cells. Data are shown as individual values and the mean + SD, n = 6 independent co-cultures using CAR-T cells generated from 6 different donors; *p <0.05, **p <0.01, ***p <0.001, ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value, the full list of p values can be found in the source data.

Source data

Extended Data Fig. 8 Dual targeting with split co-stimulation and shared CD3ζ provide superior antitumor activity and better T cell persistence in NB model when mice are treated with inverted B7-H3-specifc CAR and GD2-specific CAR.

(a) Schema of the CHLA-255 metastatic xenograft NB model in NSG mice. Eight week old female NSG mice were inoculated with 2 x 106 FFLuc-labelled CHLA-255 cells via tail vein injection, and 14 days later mice were treated with 2 x 106 CD19.28ζ, B7-H3.28ζ or B7-H3.28ζ/GD2.BB CAR-T cells via tail vein injection. (b, c) Representative tumor bioluminescence (BLI) images (b) and tumor BLI kinetics (c) of FFLuc-CHLA-255 tumor growth in the metastatic xenograft NB model shown in (a) (n = 3 mice for the CD19.28ζ group, n = 5 mice for the other two groups). (d) Kaplan-Meier survival curve of mice in (b, c) (n = 3 for the CD19.28ζ group, n = 5 for the other two groups); **p = 0.0016 (B7-H3.28ζ vs. B7-H3.28ζ/GD2.BB) by Log-rank test. (e) Detection of circulating CAR-T cells (CD45+CD3+) in mice 14 days after CAR-T cell treatment by flow cytometry. Data are shown as individual values and the mean + SD, (n = 3 samples from 3 mice for the CD19.28ζ group, n = 5 samples from 5 mice for the other two groups); *p = 0.0144, **p = 0.0042 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value.

Source data

Extended Data Fig. 9 MSLN and CSPG4 dual targeting CAR-T cells with split co-stimulation and shared CD3ζ show sustained T cell activation and proliferation in vitro.

(a) Representative flow cytometry plots showing the expression of CARs. (b) Summary of the transduction efficiency of the CARs (n = 7 or 9 independent experiments using CAR-T cells generated from 7 or 9 different donors). Data are shown as individual values and the mean + SD. (c-e) CAR-T cells co-cultured with GFP labeled H2052 cell at T cell to tumor cell ratio of 1 to 5. IFN-γ (d) and IL-2 (e) released by CAR-T cells. On day 5, tumor cells (GFP+) and CAR-T cells (CD3+) were measured by flow cytometry (c). Data are shown as individual values and the mean + SD, n = 3 independent co-cultures using CAR-T cells generated from 3 different donors for the CSPG4.BBζ group, n = 5 independent co-cultures using CAR-T cells generated from 5 different donors for the other groups; *p <0.05, **p <0.01, ***p <0.001, ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value, the full list of p values can be found in the source data. (f) Schema of the multi-round co-culture experiments of CAR-T cells and H2052. Tumor cells were seeded one day prior to the addition of T cells. At day 0, CAR-T cells were added at T cell to tumor cell ratio of 1 to 5. At the end of each round of co-culture, which are at days 5, 9, 13 and 17, one third of T cells were collected and transferred into a new well with 2.5 × 105 H2052 cells that were seeded one day before. T cells and tumor cells and cytokine released by CAR-T cells were quantified at each round of co-culture. (g, h) Summary of IFN-γ (g) and IL-2 (h) released by CAR-T cells in the multi-round co-culture with H2052 as described in (f). Data are shown as individual values and the mean + SD, n = 3 independent co-cultures using CAR-T cells generated from 3 different donors for the CSPG4.BBζ group, n = 4 independent co-cultures using CAR-T cells generated from 4 different donors for the other groups; *p <0.05, **p <0.01, ***p <0.001, ****p <0.0001 by one-way ANOVA with Tukey’s multiple comparison test adjusted p value, the full list of p values can be found in the source data.

Source data

Extended Data Fig. 10 Dual specific GD2 and B7-H3 CAR-T cells with split costimulation and shared CD3z have superior antitumor activity and prevent antigen escape in high tumor burden xenograft model with neuroblastoma cells showing heterogeneous GD2 expression.

(a) Schema of the high tumor burden SH-SY5Y metastatic xenograft NB model using NSG mice inoculated via tail vein injection with FFLuc-SH-SY5Y cells (1 × 106 cell/mouse) and treated 7 days later with CD19.28ζ, GD2.28ζ or GD2.28ζ/B7-H3BB CAR-T cells (1 × 107 cells/mouse) intravenously. (b,c) Representative tumor bioluminescence (BLI) images (b), and tumor BLI kinetics (c) of FFLuc-SH-SY5Y tumor growth (n = 5 mice/group) in the metastatic xenograft NB models shown in (a). (d) Kaplan-Meier survival curve of mice in (b, c), n = 5 mice/group, comparisons of survival curves were determined by Log-rank test, **p = 0.0023 for CD19.28ζ vs. GD2.28ζ, **p = 0.0027 for GD2.28ζ vs. GD2.28ζ/B7-H3.BB.

Source data

Supplementary information

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. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 8

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. 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

Hirabayashi, K., Du, H., Xu, Y. et al. Dual-targeting CAR-T cells with optimal co-stimulation and metabolic fitness enhance antitumor activity and prevent escape in solid tumors. Nat Cancer 2, 904–918 (2021). https://doi.org/10.1038/s43018-021-00244-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00244-2

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer