Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Jul 20;186(15):3148-3165.e20.
doi: 10.1016/j.cell.2023.06.002. Epub 2023 Jul 5.

Vaccine-boosted CAR T crosstalk with host immunity to reject tumors with antigen heterogeneity

Affiliations

Vaccine-boosted CAR T crosstalk with host immunity to reject tumors with antigen heterogeneity

Leyuan Ma et al. Cell. .

Abstract

Chimeric antigen receptor (CAR) T cell therapy effectively treats human cancer, but the loss of the antigen recognized by the CAR poses a major obstacle. We found that in vivo vaccine boosting of CAR T cells triggers the engagement of the endogenous immune system to circumvent antigen-negative tumor escape. Vaccine-boosted CAR T promoted dendritic cell (DC) recruitment to tumors, increased tumor antigen uptake by DCs, and elicited the priming of endogenous anti-tumor T cells. This process was accompanied by shifts in CAR T metabolism toward oxidative phosphorylation (OXPHOS) and was critically dependent on CAR-T-derived IFN-γ. Antigen spreading (AS) induced by vaccine-boosted CAR T enabled a proportion of complete responses even when the initial tumor was 50% CAR antigen negative, and heterogeneous tumor control was further enhanced by the genetic amplification of CAR T IFN-γ expression. Thus, CAR-T-cell-derived IFN-γ plays a critical role in promoting AS, and vaccine boosting provides a clinically translatable strategy to drive such responses against solid tumors.

Keywords: CAR T; IFN-γ; antigen loss; antigen spreading; antigenic heterogeneity; cancer; chimeric antigen receptor T cell; immunotherapy; solid tumor; tumor heterogeneity; vaccine.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests L.M. and D.J.I. are inventors on patents filed in relation to the amphiphile-vaccine technology. D.J.I. is a co-founder, shareholder, and consultant for Elicio Therapeutics, which has licensed patents related to the amphiphile-vaccine technology.

Figures

Figure 1.
Figure 1.. Vaccine boosting enables CAR T-cells to elicit endogenous T-cell responses in multiple tumor models.
(A) Schematic of CAR T-vax therapy. Created with BioRender.com. (B) IFN-γ ELISPOT. Mice bearing EGFRvIII+CT-2A tumors (n=5) treated with or without CAR T + various combinations of vaccine components. (C) Priming of endogenous CD8+ and CD4+ T-cells in EGFRvIII+CT-2A tumor-bearing mice (n=5–6) following CAR-T ± vax as measured by IFN-γ ELISPOT. (D-G) Mice (n=5–6) bearing OVA+ B16F10 tumors received FITC/TA99 CAR T-vax. (D) IFN-γ ELISPOT measuring OVA-specific endogenous T-cell responses. (F) IFN-γ ELISPOT measuring endogenous T-cell responses against Trp1−/− B16F10 cells. (F-G)Tetramer-staining showing representative flow cytometry staining (F) and mean percentages of SIINFEKL tetramer+ endogenous T cells (G). (H) IFN-γ ELISPOT. Mice (n=4–5) bearing B16F10 tumors were treated with vax only, FITC-CAR T, or FITC/TA99 CAR T ± vax. Error bars show mean ± 95% CI. ***, p<0.0001; **, p<0.01; *, p<0.05; n.s., not significant by one-way ANOVA with Tukey’s post-test.
Figure 2.
Figure 2.. Endogenous tumor-infiltrating T cells show transcriptional changes associated with enhanced anti-tumor activity in response to CAR T-vax therapy.
(A) Enumeration of intratumoral host T-cells in tumor-bearing mice (n=5) post CAR T ± vax treatment. (B-G) Tumor-bearing mice were treated with CAR T ± vax, TILs were isolated for scRNA-seq. (B) Experimental setup/timeline. Created with BioRender.com. (C) UMAP of endogenous T-cells obtained from tumors. (D) Curated clusters based on signature gene expression. (E) Stacked charts showing proportions of each T-cell cluster. (F) Dot plots showing differential expression of signature genes in endogenous CD8+ CTLs or CD4+ Th cells. (G) Cytotoxicity score of endogenous p15E-specific TILs and TILs of unknown specificity. All mice bear EGFRvIII+CT-2A tumors. Error bars are mean ± 95% CI, ****p<0.0001; **, p<0.01; n.s., not significant by Student’s t-test for A, by two-sided Wilcoxon rank-sum test for G.
Figure 3.
Figure 3.. Vaccine-driven antigen spreading is required for long-tumor tumor control in immunocompetent mice.
(A-E) Treatment of tumor-bearing WT or Rag1−/− mice with WT CAR-T ± vax. (A) Tumor growth in individual mice. Untreated, n = 5; CAR-T in WT mice, n = 10; CAR T-vax, n = 15 and 10 in WT and Rag1−/− mice, respectively. (B) Percentage of mice that completely rejected tumors or experienced tumor relapse. (C) Overall survival. (D-E) Surface EGFRvIII expression (D) and mean expression normalized to untreated tumors (E) on parental or representative relapsed tumors from WT and Rag1−/− mice following CAR T-vax treatment. (F-G) Tumor-bearing WT or CD8α−/− mice (n=5–8) ± CAR T-vax treatment. (F) Tumor growth. (G) Overall survival. (H) Individual tumor growth and overall survival of WT (n=10) or Rag1−/− mice (n=5) bearing heterogeneous CT-2A tumors upon CAR T-vax treatment. EGFRvIII+:EGFRvIII cells were pre-mixed at the indicated ratios. All mice in A-G bear EGFRvIII+CT-2A tumors. Error bars are mean ± 95% CI, ***, p<0.0001; **, p<0.01; *, p<0.05 by Student’s t-test for E, by Log-rank (Mantel-Cox) test for C,G-H, by two-way ANOVA with Tukey’s post-test for F.
Figure. 4.
Figure. 4.. Vaccine boosting induces cell-intrinsic enhancements in CAR T-cell function that include metabolic reprogramming.
(A) Tumor growth (left) and overall survival (right) of tumor-bearing mice (n=8) after receiving vaccine-boosted or non-boosted CAR T cells. Created with BioRender.com. (B-C) Tumor-bearing mice received WT CAR T ± vax treatment, and CAR T-cells were isolated from spleens and tumors for RNA-seq. (B) Volcano plot showing differential gene expression in splenic CAR T-cells. (C) GSEA showing enriched pathways in intratumoral CAR T-cells. (D-E) Intracellular PGC-1α expression (D) and mitochondrial mass (E) in intratumoral CAR T cells from mice (n=5) 7 days post treatment with WT CAR T ± vax. (F) IFN-γ ELISPOT. Tumor-bearing mice (n=5) treated with WT CAR T ± vax or PGC-1α−/− CAR T-vax. All mice bear EGFRvIII+CT-2A tumors. Error bars are mean ± 95% CI, **, p<0.01; *, p<0.05 by Student’s t-test for D-E, and one-way ANOVA with Tukey’s post-test for F.
Figure. 5.
Figure. 5.. Enhanced IFN-γ production by vaccine-boosted CAR T-cells is critical for antigen spreading.
(A) IFN-γ and TNF-α expression in intratumoral CAR T-cells from mice (n=5) treated with WT CAR T ± vax. (B) IFN-γ expression in intratumoral CAR T-cells from mice (n=5) 7 days post treatment with WT or PGC-1α−/− CAR T-vax. (C) IFN-γ ELISPOT. Tumor-bearing mice (n=5) treated with WT CAR T or WT CAR T-vax + isotype control antibody (IgG), anti-TNF-α or anti-IFN-γ. (D-E) OVA+EGFRvIII+CT-2A tumor-bearing mice(n=5–10) treated by WT CAR T or WT CAR T-vax ± anti-IFN-γ. Endogenous OVA-specific T-cell responses detected by SIINFEKL-tetramer staining (D) and IFN-γ ELISPOT (E). (F) IFN-γ ELISPOT. Tumor-bearing mice (n=5) treated with WT CAR T-vax ± anti-IFN-γ at indicated time points. (G-H) Tumor growth (G) and overall survival (H) of mice left untreated (n=5) or treated (n=10) with WT CAR T-vax ± anti-IFN-γ. (I) IFN-γ ELISPOT. Tumor-bearing mice (n=5) treated with WT or IFN-γ−/− CAR T ± vax. (J) Tumor growth in mice (n=5) left untreated or treated with WT or IFN-γ−/− CAR T-vax. (K-L) Tumor growth (K) and overall survival (L) of WT or IFN-γ−/− mice (n=5–8) treated with or without WT CAR T-vax therapy. All mice bear EGFRvIII+CT-2A tumors. Error bars are mean ± 95% CI, ***, p<0.0001; **p<0.01; *, p<0.05, ns, not significant by Student’s t-test for A-B, by one-way ANOVA with Tukey’s post-test for C-F, I, by two-way ANOVA with Tukey’s post-test for J-K, and by Log-rank (Mantel-Cox) test for H and L.
Figure. 6.
Figure. 6.. DCs regulate CAR T-cell-induced antigen spreading through enhanced tumor antigen acquisition and IFN-γ-IL-12 crosstalk.
(A) EGFRvIII+ CT-2A cell killing by WT, IFN-γ−/−, or IFNGR1−/− CAR T-cells in vitro (n=3). (B) Enumeration of tumor-infiltrating immune cells in mice (n = 4–5) receiving WT CAR T ± vax. See supplemental methods for phenotyping details. (C) IFN-γ ELISPOT. Tumor-bearing WT or Batf3−/− mice (n=5) treated with WT CAR T ± vax. (D) Tumor-bearing mice were left untreated (n=4) or treated with WT or IFN-γ−/− CAR T-vax (n=5). Shown are chemokine expression in tumors 7 days post treatment. (E-F) Ki67 (E) and CCR7(F) expression in intratumoral CD103+ DCs and CD11b+ DCs from mice (n=5) treated with WT CAR T ± vax. (G-H) Mice bearing ZsGreen+EGFRvIII+CT-2A tumors were treated with WT CAR T, WT CAR T-vax or IFN-γ−/− CAR T-vax (n=5), shown are tumor antigen (ZsGreen) uptake by intratumoral CD103+ DCs (G) and CD11b+ DCs (H). (I-J) IFN-γ ELISPOT (I) and tumor growth (J) in mice (n = 5) treated with WT or IFNGR1−/− CAR T ± vax. (K-N) IFN-γ ELISPOT. (K) WT vs. CD11c-specific IFNGR1 KO tumor-bearing mice (n=5) following WT CAR T ± vax. (L) Tumor-bearing mice (n=5) following WT CAR T or WT CAR T-vax + anti-IFN-γ or anti-IL12(p70). (M) Tumor-bearing WT mice (n=5) following WT or IL12rb2−/− CAR T-vax therapy or in IL12p40−/− mice following WT CAR T-vax. (N) Tumor-bearing WT mice (n=7) following WT CAR T-vax or IFNGR1−/− CAR T ± vax. (O) Tumor growth in mice (n=5–7) left untreated or treated with WT or IFNGR1−/− CAR T ± vax. All mice except those in G-H bear EGFRvIII+CT-2A tumors. Error bars are mean ± 95% CI, ***, p<0.001; **, p<0.01; *, p<0.05; ns, not significant by Student’s t-test for B and E-F, by one-way ANOVA with Tukey’s post-test for A, C-D, G-I and K-N, by two-way ANOVA with Tukey’s post-test for J and O.
Figure. 7.
Figure. 7.. Engineering CAR T-cells for increased IFN-γ expression synergizes with vaccine boosting to enhance antigen spreading and rejection of solid tumors with pre-existing antigen heterogeneity.
(A-D). Heterogenous CT-2A tumors were established in C57BL/6 mice. (A)Tumor growth and (B)survival of mice (n=10) after treatment with WT CAR T-vax therapy ± anti-IFN-γ. (C)Tumor growth and (D)survival of mice (n=8) left untreated, receiving WT CAR T, or WT CAR T-vax ± anti-IL12 (p75). (E) IFN-γ secretion from WT or NFAT-IFN-γ CAR T-cells ± anti-CD3/CD28 beads (n=3). (F) IFN-γ ELISPOT. EGFRvIII+ CT-2A tumor-bearing mice (n=6) treated with WT or NFAT-IFN-γ CAR T ± vax. (G-L) Mice bearing heterogenous CT-2A tumors (n=5) treated with WT or NFAT-IFN-γ CAR T ± vax therapy. Enumeration of CAR T (G) and endogenous CD8+ T cells (J) infiltrated into tumors as well as the expression of IFN-γ (H for CAR T, K for host CD8 T) and granzyme B (I for CAR T, L for host CD8 T). (M-N) Tumor growth (M) and overall survival (N) of mice bearing heterogenous CT-2A tumors (n=10) treated with WT or NFAT-IFN-γ CAR T ± vax. (O) Schematic overview of CAR T-vax therapy triggered antigen spreading. Created with BioRender.com. Heterogenous CT-2A tumors are EGFRvIII+:EGFRvIII cells mixed at 80:20 ratio. Error bars are mean ± 95% CI. ***, p<0.001; **, p<0.01; *, p<0.05; ns not significant by one-way ANOVA with Tukey’s post-test for E-L, by two-way ANOVA with Tukey’s post-test for C, and Log-rank (Mantel-Cox) test for B, D and N.

Comment in

References

    1. Irvine DJ, Maus MV, Mooney DJ, and Wong WW (2022). The future of engineered immune cell therapies. Science 378, 853–858. 10.1126/science.abq6990. - DOI - PMC - PubMed
    1. Labanieh L, and Mackall CL (2022). CAR immune cells: design principles, resistance and the next generation. Nature 614, 635–648. 10.1038/s41586-023-05707-3. - DOI - PubMed
    1. Wang M, Munoz J, Goy A, Locke FL, Jacobson CA, Hill BT, Timmerman JM, Holmes H, Jaglowski S, Flinn IW, et al. (2020). KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. New Engl J Med 382, 1331–1342. 10.1056/nejmoa1914347. - DOI - PMC - PubMed
    1. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, et al. (2018). Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. New Engl J Medicine 378, 439–448. 10.1056/nejmoa1709866. - DOI - PMC - PubMed
    1. Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, Mehta A, Purev E, Maloney DG, Andreadis C, et al. (2020). Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet 396, 839–852. 10.1016/s0140-6736(20)31366-0. - DOI - PubMed

Publication types

Substances