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:

In vivo CRISPR screening in CD8 T cells with AAV–Sleeping Beauty hybrid vectors identifies membrane targets for improving immunotherapy for glioblastoma

Abstract

Targeting membrane proteins could improve the efficacy of T cell–based immunotherapies. To facilitate the identification of T cell targets, we developed a hybrid genetic screening system where the Sleeping Beauty (SB) transposon and single guide RNA cassette are nested in an adeno-associated virus (AAV). SB-mediated genomic integration of the single guide RNA cassette enables efficient gene editing in primary murine T cells as well as a screen readout. We performed in vivo AAV–SB-CRISPR screens for membrane protein targets in CD8+ T cells in mouse models of glioblastoma (GBM). We validated screen hits by demonstrating that adoptive transfer of CD8+ T cells with Pdia3, Mgat5, Emp1 or Lag3 gene editing enhances the survival of GBM-bearing mice in both syngeneic and T-cell receptor transgenic models. Transcriptome profiling, single cell sequencing, cytokine assays and T cell signaling analysis showed that Pdia3 editing in T cells enhances effector functions. Engineered PDIA3 mutant EGFRvIII chimeric antigen T cells are more potent in antigen-specific killing of human GBM cells.

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: In vivo AAV–SB-CRISPR CD8+ T-cell screen of membrane proteome knockouts in GBM.
Fig. 2: In vivo validation of top candidates by adoptive transfer of mutant CD8+ T cells in mouse models of GBM.
Fig. 3: Single-cell RNA-seq and bulk mRNA-seq analysis of Pdia3 knockout in CD8+ T cells.
Fig. 4: Mechanistic analysis and preclinical efficacy testing of Pdia3 knockout in CD8+ T cells.
Fig. 5: Human CD8+ T-cell PDIA3 knockout and effector function analysis.
Fig. 6: Human PDIA3−/−-EGFRvIII CAR-T-cell establishment and GBM cell killing.

Similar content being viewed by others

Data availability

Source data and statistics for non-NGS experiments such as tumor studies, flow cytometry, T7E1, qPCR, protein experiments and coculture assays are provided in an Excel table as Source Data. Genome sequencing data are deposited to the Sequence Read Archive with accession number PRJNA553676. Other data, reagents, methods, computational code and materials that support the findings of this research are available from the corresponding author upon reasonable request.

Code availability

Custom code used to support the findings of this research is available from the corresponding author upon reasonable request.

References

  1. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).

    PubMed  Google Scholar 

  2. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kvistborg, P. et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8(+) T cell response. Sci. Transl. Med. 6, 254ra128 (2014).

    PubMed  Google Scholar 

  4. Ribas, A. Tumor immunotherapy directed at PD-1. New Engl. J. Med. 366, 2517–2519 (2012).

    CAS  PubMed  Google Scholar 

  5. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Omuro, A. et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro Oncol. 20, 674–686 (2018).

    CAS  PubMed  Google Scholar 

  7. O’Rourke, D. M. et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 9, eaaa0984 (2017).

    PubMed  PubMed Central  Google Scholar 

  8. Chow, R. D. et al. AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma. Nat. Neurosci. 20, 1329–1341 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, G. et al. Mapping a functional cancer genome atlas of tumor suppressors in mouse liver using AAV-CRISPR-mediated direct in vivo screening. Sci. Adv. 4, eaao5508 (2018).

    PubMed  PubMed Central  Google Scholar 

  10. Mates, L. et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat. Genet. 41, 753–761 (2009).

    CAS  PubMed  Google Scholar 

  11. Colella, P., Ronzitti, G. & Mingozzi, F. Emerging issues in AAV-mediated in vivo gene therapy. Mol. Ther. Methods Clin. Dev. 8, 87–104 (2018).

    CAS  PubMed  Google Scholar 

  12. Wang, D., Tai, P. W. L. & Gao, G. P. Adeno-associated virus vector as a platform for gene therapy delivery. Nat. Rev. Drug Discov. 18, 358–378 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Platt, R. J. et al. CRISPR–Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Hogquist, K. A. et al. T-cell receptor antagonist peptides induce positive selection. Cell 76, 17–27 (1994).

    CAS  PubMed  Google Scholar 

  15. Workman, C. J., Dugger, K. J. & Vignali, D. A. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J. Immunol. 169, 5392–5395 (2002).

    CAS  PubMed  Google Scholar 

  16. Workman, C. J. et al. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J. Immunol. 172, 5450–5455 (2004).

    CAS  PubMed  Google Scholar 

  17. Anderson, A. C., Joller, N. & Kuchroo, V. K. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 44, 989–1004 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Huang, C. T. et al. Role of LAG-3 in regulatory T cells. Immunity 21, 503–513 (2004).

    CAS  PubMed  Google Scholar 

  19. Smith, L. K. et al. Interleukin-10 directly inhibits CD8(+) T cell function by enhancing N-glycan branching to decrease antigen sensitivity. Immunity 48, 299–312 e295 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Shibui, A. et al. Alteration of immune responses by N-acetylglucosaminyltransferase V during allergic airway inflammation. Allergol. Int. 60, 345–354 (2011).

    CAS  PubMed  Google Scholar 

  21. Morgan, R. et al. N-acetylglucosaminyltransferase V (Mgat5)-mediated N-glycosylation negatively regulates Th1 cytokine production by T cells. J. Immunol. 173, 7200–7208 (2004).

    CAS  PubMed  Google Scholar 

  22. Sugiura, K. & Stock, C. C. Studies in a tumor spectrum. II. The effect of 2,4,6-triethylenimino-s-triazine on the growth of a variety of mouse and rat tumors. Cancer 5, 979–991 (1952).

    CAS  PubMed  Google Scholar 

  23. Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Schlager, C. et al. Effector T-cell trafficking between the leptomeninges and the cerebrospinal fluid. Nature 530, 349 (2016).

    PubMed  Google Scholar 

  25. Khalil, D. N., Smith, E. L., Brentjens, R. J. & Wolchok, J. D. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat. Rev. Clin. Oncol. 13, 273–290 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Ribas, A. et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 16, 908–918 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Preusser, M., Lim, M., Hafler, D. A., Reardon, D. A. & Sampson, J. H. Prospects of immune checkpoint modulators in the treatment of glioblastoma. Nat. Rev. Neurol. 11, 504–514 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Cloughesy, T. F. et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 25, 477–486 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Li, B. et al. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome Biol. 17, 174 (2016).

    PubMed  PubMed Central  Google Scholar 

  30. Reardona, D. A. et al. Randomized phase 3 study evaluating the efficacy and safety of nivolumab vs bevacizumab in patients with recurrent glioblastoma: Checkmate 143. Neuro Oncol. 19, 21–21 (2017).

    Google Scholar 

  31. Agarwalla, P., Barnard, Z., Fecci, P., Dranoff, G. & Curry, W. T. Sequential immunotherapy by vaccination with GM-CSF-expressing glioma cells and CTLA-4 blockade effectively treats established murine intracranial tumors. J. Immunother. 35, 385–389 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fecci, P. E. et al. Systemic CTLA-4 blockade ameliorates glioma-induced changes to the CD4(+) T cell compartment without affecting regulatory T-cell function. Clin. Cancer Res. 13, 2158–2167 (2007).

    CAS  PubMed  Google Scholar 

  33. Mathios, D. et al. Anti-PD-1 antitumor immunity is enhanced by local and abrogated by systemic chemotherapy in GBM. Sci. Transl. Med. 8, 370ra180 (2016).

    PubMed  PubMed Central  Google Scholar 

  34. Saha, D., Martuza, R. L. & Rabkin, S. D. Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade. Cancer Cell 32, 253–267.e5 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kim, J. E. et al. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin. Cancer Res. 23, 124–136 (2017).

    CAS  PubMed  Google Scholar 

  36. Dai, H., Wang, Y., Lu, X. & Han, W. Chimeric antigen receptors modified T-cells for cancer therapy. J. Natl Cancer Inst. 108, djv439 (2016).

    PubMed  PubMed Central  Google Scholar 

  37. Brown, C. E. et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 375, 2561–2569 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Pellegatta, S. et al. Constitutive and TNFɑ-inducible expression of chondroitin sulfate proteoglycan 4 in glioblastoma and neurospheres: implications for CAR-T cell therapy. Sci. Transl. Med. 10, eaao2731 (2018).

    PubMed  PubMed Central  Google Scholar 

  39. Mount, C. W. et al. Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M(+) diffuse midline gliomas. Nat. Med. 24, 572–579 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Bernards, R. Finding effective cancer therapies through loss of function genetic screens. Curr. Opin. Genet. Dev. 24, 23–29 (2014).

    CAS  PubMed  Google Scholar 

  41. Okada, M. et al. Blockage of core fucosylation reduces cell-surface expression of PD-1 and promotes anti-tumor immune responses of T cells. Cell Rep. 20, 1017–1028 (2017).

    CAS  PubMed  Google Scholar 

  42. Shifrut, E. et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell 175, 1958–1971.e15 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Ting, P. Y. et al. Guide Swap enables genome-scale pooled CRISPR–Cas9 screening in human primary cells. Nat. Methods 15, 941–946 (2018).

    CAS  PubMed  Google Scholar 

  44. Zhou, P. et al. In vivo discovery of immunotherapy targets in the tumour microenvironment. Nature 506, 52–57 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Chen, R. et al. In vivo RNA interference screens identify regulators of antiviral CD4(+) and CD8(+) T cell differentiation. Immunity 41, 325–338 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Shalem, O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    CAS  PubMed  Google Scholar 

  47. Demetriou, M., Granovsky, M., Quaggin, S. & Dennis, J. W. Negative regulation of T-cell activation and autoimmunity by Mgat5 N-glycosylation. Nature 409, 733–739 (2001).

    CAS  PubMed  Google Scholar 

  48. Granovsky, M. et al. Suppression of tumor growth and metastasis in Mgat5-deficient mice. Nat. Med. 6, 306–312 (2000).

    CAS  PubMed  Google Scholar 

  49. Doench, J. G. et al. Rational design of highly active sgRNAs for CRISPR–Cas9-mediated gene inactivation. Nat. Biotechnol. 32, 1262–1267 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Uren, A. G. et al. A high-throughput splinkerette-PCR method for the isolation and sequencing of retroviral insertion sites. Nat. Protoc. 4, 789–798 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10–12 (2011).

    Google Scholar 

  52. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    PubMed  PubMed Central  Google Scholar 

  53. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  55. Koboldt, D. C. et al. VarScan: variant detection in massively parallel sequencing of individual and pooled samples. Bioinformatics 25, 2283–2285 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    CAS  PubMed  Google Scholar 

  57. Pimentel, H., Bray, N. L., Puente, S., Melsted, P. & Pachter, L. Differential analysis of RNA-seq incorporating quantification uncertainty. Nat. Methods 14, 687–690 (2017).

    CAS  PubMed  Google Scholar 

  58. Huang da, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    PubMed  Google Scholar 

  59. Jiang, P. et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 24, 1550–1558 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. Fuchs, D. Hafler, R. Montgomery, D. Rimm and M. Gunel for discussions. We thank S. Lam, L. Shen, Z. Bai, H. Ye, R. Kim and other members of the Chen laboratory, as well as various colleagues in the Department of Genetics, Systems Biology Institute, Cancer Systems Biology Center, MCGD Program, Immunobiology Program, BBS Program, Cancer Center and Stem Cell Center at Yale for assistance and/or discussion. We thank the Center for Genome Analysis, the Center for Molecular Discovery, Pathology Tissue Services, Histology Services, the High Performance Computing Center, West Campus Analytical Chemistry Core and West Campus Imaging Core, the Mass Cytometry Core and Keck Biotechnology Resource Laboratory at Yale for technical support. S.C. is supported by Yale SBI/Genetics Startup Fund, the NIH/NCI (grant nos. DP2CA238295, R01CA231112, U54CA209992-8697, R33CA225498, RF1048811, P50CA196530-A10805, P50CA121974-A08306), the Damon Runyon Dale Frey Award (grant no. DFS-13-15), the Melanoma Research Alliance (grant nos. 412806, 16-003524), St-Baldrick’s Foundation (grant no. 426685), the Breast Cancer Alliance, the Cancer Research Institute (CLIP), AACR (grant nos. 499395, 17-20-01-CHEN), The Mary Kay Foundation (grant no. 017-81), The V Foundation (grant no. V2017-022), the Ludwig Family Foundation, DoD (grant no. W81XWH-17-1-0235), the Sontag Foundation and the Chenevert Family Foundation. G.W. is supported by CRI Irvington and RJ Anderson Fellowships. X.D. is supported by a Revson Fellowship. M.B.D., R.D.C. and J.J.P. are supported by the Yale MSTP training grant from the NIH (grant no. T32GM007205).

Author information

Authors and Affiliations

Authors

Contributions

L.Y. designed the AAV–SB–CRISPR vector, developed the screening system and performed the majority of experiments in this study. J.J.P. analyzed the CRISPR screen and other high-throughput data. M.B.D. generated transgenic mice and optimized the screen. R.D.C. designed the Surf library. Q.Y., L.P., Y.D., J.G., G.W. and Y.E. assisted with various experiments. Y.D. and X.D. designed and generated the EGFRvIII CAR-T system. S.C. conceived the study, secured funding and supervised the work. L.Y., J.J.P. and S.C. prepared the manuscript with input from all of the other authors.

Corresponding author

Correspondence to Sidi Chen.

Ethics declarations

Competing interests

A patent application has been filed related to the data in this manuscript.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Splinkerette PCR identify genome integration of Sleeping Beauty transposon.

(a) Nextera indel analysis for Mll3 and B2m knock-out in mouse CD8+ T cells using AAV-sgRNA vectors. Representative mutations and their frequencies were shown around predicted sgRNA target sites. (b) Schematics of splinkerette PCR procedures. The steps include genomic DNA isolation, restriction enzyme digestion, adaptor ligation, PCR, NGS library prep, and sequencing. (c) Electrophoresis of the splinkerette PCR products. The gel within red dash line was gel purified for the Nextera library preparation and sequencing. (d) Representative SB transposon integration sites in the mouse genome. (e) A bar plot of splinkerette PCR read distribution for the number of integration sites along mouse chromosomes. (f) A bar plot showed splinkerette PCR read distribution for the number of integration sites according to functional annotation of genomic regions.

Supplementary Figure 2 Single cell RT-qPCR estimation of functional MOI of AAV-SB-CRISPR screen.

Single cell RT-qPCR detection of single T cell expressing functional sgRNAs, for the estimation of functional MOI with exact transduction parameters in the AAV-Surf screen. PBS treated single T cells were used as a negative control. Numbers of wells without T cells, with sgRNA- T cells and with sgRNA+ T cells were determined to estimate MOI.

Supplementary Figure 3 Penetrance estimation of intracranial brain tumor induction using GL261 and derivatives cell lines.

(a) Bar plot of mortality rate of mouse after intracranial GBM induction. Different GBM cell lines, GL261, GL261-Luc-Ova, and GL261-Luc, were used for brain injection, and each cell line injected with different cell number. (b) Bar plot of number of mice that met euthanasia endpoint due to brain tumor.

Supplementary Figure 4 AAV-Surf library transduced mouse CD8+ T cell surface phenotypes and TCR repertoires before and after adoptive transfer.

(a) Flow cytometry analysis of surface PD-1, Lag3, and Tim-3 expression after transduced with AAV-Vector and AAV-Surf virus. Result from one experiment. (b-c) Flow cytometry analysis of proportion of PD-1+ TILs. ns, non-significant. Data are shown as mean ± s.e.m., with individual data points. The p-values and number of mice used in each group are indicated in the plots and/or in a supplemental excel table. (d) A bar plot of T cell clonal composition from TCR sequencing. Pre injection, mouse CD8+ T cells transduced with AAV-Surf virus and cultured for 5 days; Post injection (TIL), AAV-Surf transduced T cells i.v. injected into GBM-bearing mice and isolated as TILs at day 6 after T cell injection. (e) A ring plot of TCR distribution for T cells before i.v. injection. Top TCR sequences were labeled in the plot. (f) A ring plot of TCR distribution for T cells after i.v. injection (TILs). Top TCR sequences were labeled in the plot.

Source data

Supplementary Figure 5 AAV-CRISPR CD8+ T cell screens of membrane bound proteome knockouts in GBM.

(a) Schematics of hybrid AAV-SB-CRIPSR CD8+ T cell screen in a syngeneic mouse model of GBM, showing steps of naïve CD8+ T cell isolation, AAV library transduction, GBM cell transplantation, adoptive T cell transfer, brain and tumor isolation, and sgRNA readout by deep sequencing. (b) Representative in vivo imaging illustrating the growth of GL261-FLuc tumors in the brains of C57BL/6 mice. (c) Representative photos of mice after GBM transplantation and T cell treatment. Red circles indicated macrocephaly suggestive of growing brain tumors. (d) Survival plot of mice after GBM engraftment and adoptive transfer. C57BL/6J mice were transplanted with 1.2 x 106 GL261-FLuc into the lateral ventricle (LV). 4 x 106 OT-I;Cas9β CD8+ T cells were injected after 10 days of tumor engraftment. Survival significance was assessed by a log-rank Mantel-Cox test. The p-values and number of mice used in each group was indicated in the plots. The group of mice receiving adoptive transfer of T cells has significantly enhanced survival, where the effect of AAV-Surf is slightly stronger than Vector. DPI, days post tumor implantation. (e) Representative H&E stained brain sections from PBS, AAV-Vector and AAV-Surf groups. Areas within red dashed lines indicate brain tumors. Scale bar, 2 mm for whole brain sections, and 100 μm for zoom-in sections. These are representative images at the endpoint of survival thus not quantitative for comparison in terms of tumor burden. One experiment, n = 3 individual mice for PBS, n = 5 mice for vector, n = 11 mice for AAV-Surf. (f) Scatterplot of brain vs. cell sgRNA library representation of AAV-Surf shorter term screen experiment (max survival 20 days post injection). The most enriched sgRNAs in the brain are highlighted. Purple dash line, y = x curve; blue dash line, linear regression of the distribution of the 1,000 NTCs between the brain and cell samples. (g) Scatterplot of brain vs. cell sgRNA library representation of AAV-Surf longer term screen experiment (max survival 92 days post injection). The most enriched sgRNAs in the brain are highlighted. Purple dash line, y = x curve; blue dash line, linear regression of the distribution of the 1,000 NTCs between the brain and cell samples. The p-values and number of mice used in each group are indicated in the plots and/or in a supplemental excel table.

Source data

Supplementary Figure 6 Clonal GBM cell line generation and representative mouse GBM histology with single gene knockout adoptive transfer.

(a) Flow cytometry analysis of GL261-FLuc-mCh-cOVA clones for cOVA expression level. Two independent experiments. (b) T7EI assay indicated gene editing of Mgat5 and Pdia3 with AAV-SB100x vector. Multiple, at least five independent experiments. (c) T7EI assay for mouse T cells treated with sgPdia3 on top 4 predicted off-target sites. Two experiments. (d) RT-qPCR of Mgat5 and Pdia3 after infected with AAV6 carrying specific gene-targeting sgRNAs in AAV-SB100x vector. Unpaired t test was used for the significance assessment, AAV-Vector vs. AAV-sgMgat5, p = 0.0242. * p < 0.05. (e) Schematic of the therapeutic efficacy testing strategy for top candidates identified from the AAV-Surf screens, using adoptive transfer of single gene edited CD8+ T cells in a syngeneic mouse model of GBM. (f) Survival plots of the top candidate validations in a syngeneic mouse model of GBM. C57BL/6J mice were engrafted with 2 x 105 GL261 cancer cells, and adoptive transfer treatment was performed after 10 days of tumor engraftment by intravenous injection of 6 x 105 Cas9β CD8+ T cells infected with AAV-Vector, AAV-sgLag3, AAV-sgMgat5, and AAV-sgPdia3. Survival significance was assessed by a log-rank Mantel-Cox test. (g) Representative H&E stained brain sections from AAV-Vector and AAV-sgRNA single knockout groups in C57BL/6J mice. Scale bar, 100 μm. These are representative images at the endpoint of survival thus not quantitative for comparison in terms of tumor burden. One experiment, n = 8 individual mice for each group. (h) Representative H&E stained brain sections from AAV-Vector and AAV-sgRNA single knockout groups in Rag1-/- mice. Scale bar, 100 μm. These are representative images at the endpoint of survival thus not quantitative for comparison in terms of tumor burden. One experiment, n = 8 mice for AAV-Vector, n = 10 mice for AAV-sgMgat5, n = 9 for AAV-sgPdia3, and n = 8 for AAV-sgEmp1. The p-values and number of mice used in each group are indicated in the plots and/or in a supplemental excel table.

Source data

Supplementary Figure 7 Representative luciferase imaging for tumor burden quantification.

(a) Quantification of tumor burden as total luciferase flux at days 12, 16, and 18. Day 20 and 22 data points were not used for statistics because most mice in the AAV-Vector group had already reached endpoint. Mice being imaged, n = 8 for Vector, n = 8 for sgMgat5, n = 5 for sgPdia3. Data are shown as mean ± s.e.m. plus individual data points on bar graphs. (b) Quantification of tumor burden as total luciferase flux at days 14, 15, 17, 19, and 22 after tumor induction. Mice being imaged, n = 7 for Vector, n = 7 for sgMgat5, n = 8 for sgPdia3, n = 8 for sgMgat5+sgPdia3. Data are shown as mean ± s.e.m. plus individual data points on bar graphs. The numbers of mice that had reached endpoint, euthanized, and therefore removed from the imaging group are indicated on the top table.

Source data

Supplementary Figure 8 Single cell RNA-seq profiling of Pdia3 KO CD8+ T cells.

(a) t-SNE plot of hierarchical clustering of scRNA-seq results. (b) Volcano plot of scRNA-seq of mouse CD8+ T cells after Pdia3 knockout. Pdia3 is a significantly downregulated gene after infection with AAV-sgPdia3. A total 9,193 single cells were captured and their transcriptomes were sequenced for the AAV-sgPdia3 (n = 4,549 single cells) and AAV-Vector (n = 4,644 single cells) treated CD8 T cells. Two-sided Wilcoxon signed-rank test by gene, with p-values adjusted by Benjamini & Hochberg.

Supplementary Figure 9 Immune marker analysis of PDIA3 KO human CD8+ T cells by CyTOF.

(a) t-SNE plot clustered by samples, showing that the 3 PDIA3 KO samples grouped with each other, the 3 WT samples also grouped with each other, and that PDIA3 KO samples and WT samples formed distinct groups. (b) t-SNE plot of CyTOF data with k-means clustering revealing 10 major clusters. (c) t-SNE and violin plots of CyTOF data of CD127 / IL7R, FAS / CD95, 4-1BB / CD137 and TIM-3 / HAVCR2 at the surface protein level, for both PDIA3 KO and wildtype single human CD8+ T cells (n = 3 replicates each, sampled 7,000 cells per replicate for comparison). Violins show kernel probability density on side, and boxplot is standard, i.e. middle band is median, hinges/ends of box are interquartile range (25% and 75% quantiles), lower whisker = smallest observation greater than or equal to lower hinge - 1.5 * IQR, upper whisker = largest observation less than or equal to upper hinge + 1.5 * IQR. Wilcoxon test, two-sided, p value adjusted by Benjamini & Hochberg method. KO vs WT: p = 1.91e-81 for CD127 / IL7R, p = 0.1147 for FAS / CD95, p = 6.75e-83 for 4-1BB / CD137, and p = 0 for TIM-3 / HAVCR2.

Supplementary Figure 10 TIDE analysis of PDIA3-related T cell dysfunction in human cancer patients.

(a–c) Analyses of PDIA3 expression signatures linked to in cytotoxic T lymphocyte (CTL) – associated survival benefits in patients, where high-level of PDIA3 abolishes or weakens the overall survival benefit of CTL-high patients with GBM (a), TNBC (b), or LUAD (c). X-axis is overall survival in months. Y-axis is survival fraction. Specific statistical tests using TIDE (Methods). (d) Analyses of PDIA3 expression signatures linked to survival benefits of patients treated with an immune-checkpoint antibody (Ipilimumab, anti-CTLA4), in melanoma. X-axis is overall survival in days. Y-axis is survival fraction. Specific statistical tests were performed using TIDE (Methods).

Supplementary Figure 11 Representative flow cytometry gating.

(a) Gating of mouse Ifnγ+ CD8+ T cells. (b) Gating of human IFNγ+ CD8+ T cells.

Supplementary information

Supplementary Information

Supplementary Figs. 1–11 and Methods.

Reporting Summary

Supplementary Datasets

Supplementary Datasets 1–9.

Supplementary Tables

Supplementary Tables 1–11.

Source Data for Figs. 1–6 and Supplementary Figs. 4–7

Source Data for Figs. 1–6 and Supplementary Figs. 4–7

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ye, L., Park, J.J., Dong, M.B. et al. In vivo CRISPR screening in CD8 T cells with AAV–Sleeping Beauty hybrid vectors identifies membrane targets for improving immunotherapy for glioblastoma. Nat Biotechnol 37, 1302–1313 (2019). https://doi.org/10.1038/s41587-019-0246-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0246-4

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