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:

Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results

This article has been updated

Abstract

Recurrent glioblastoma (rGBM) remains a major unmet medical need, with a median overall survival of less than 1 year. Here we report the first six patients with rGBM treated in a phase 1 trial of intrathecally delivered bivalent chimeric antigen receptor (CAR) T cells targeting epidermal growth factor receptor (EGFR) and interleukin-13 receptor alpha 2 (IL13Rα2). The study’s primary endpoints were safety and determination of the maximum tolerated dose. Secondary endpoints reported in this interim analysis include the frequency of manufacturing failures and objective radiographic response (ORR) according to modified Response Assessment in Neuro-Oncology criteria. All six patients had progressive, multifocal disease at the time of treatment. In both dose level 1 (1 ×107 cells; n = 3) and dose level 2 (2.5 × 107 cells; n = 3), administration of CART-EGFR-IL13Rα2 cells was associated with early-onset neurotoxicity, most consistent with immune effector cell-associated neurotoxicity syndrome (ICANS), and managed with high-dose dexamethasone and anakinra (anti-IL1R). One patient in dose level 2 experienced a dose-limiting toxicity (grade 3 anorexia, generalized muscle weakness and fatigue). Reductions in enhancement and tumor size at early magnetic resonance imaging timepoints were observed in all six patients; however, none met criteria for ORR. In exploratory endpoint analyses, substantial CAR T cell abundance and cytokine release in the cerebrospinal fluid were detected in all six patients. Taken together, these first-in-human data demonstrate the preliminary safety and bioactivity of CART-EGFR-IL13Rα2 cells in rGBM. An encouraging early efficacy signal was also detected and requires confirmation with additional patients and longer follow-up time. ClinicalTrials.gov identifier: NCT05168423.

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: Regression of multifocal rGBM after intraventricular delivery of CART-EGFR-IL13Rα2 cells (dose level 1).
Fig. 2: Regression of multifocal rGBM after intraventricular delivery of CART-EGFR-IL13Rα2 cells (dose level 2).
Fig. 3: CAR T engraftment kinetics and CSF cytokine levels.

Similar content being viewed by others

Data availability

The data that support the findings of this study are included in the paper or may be available from the corresponding author, recognizing that certain patient-related data not included in the paper were generated as part of the clinical trial and may be subject to patient confidentiality. It is estimated that the corresponding author will respond to external data requests within 2 weeks of receipt of request. Further information on research design is available in the Nature Research Reporting Summary linked to this article. Source data are provided with this paper.

Change history

  • 22 March 2024

    In the version of the article initially published an incorrect version of the Supplementary Information was included. This has now been updated in the HTML version of the article.

References

  1. Wen, P. Y. et al. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 22, 1073–1113 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Goff, S. L. et al. Pilot trial of adoptive transfer of chimeric antigen receptor-transduced T cells targeting EGFRvIII in patients with glioblastoma. J. Immunother. 42, 126–135 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ahmed, N. et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 3, 1094–1101 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Brown, C. E. et al. Bioactivity and safety of IL13Rα2-redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clin. Cancer Res. 21, 4062–4072 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lin, Q. et al. First-in-human trial of EphA2-redirected CAR T-cells in patients with recurrent glioblastoma: a preliminary report of three cases at the starting dose. Front. Oncol. 11, 694941 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Liu, Z. et al. Safety and antitumor activity of GD2-specific 4SCAR-T cells in patients with glioblastoma. Mol. Cancer 22, 3 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Durgin, J. S. et al. Case Report: Prolonged survival following EGFRvIII CAR T cell treatment for recurrent glioblastoma. Front. Oncol. 11, 669071 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Alizadeh, D. et al. IFNγ is critical for CAR T cell-mediated myeloid activation and induction of endogenous immunity. Cancer Discov. 11, 2248–2265 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Choe, J. H. et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci. Transl. Med. 13, eabe7378 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bielamowicz, K. et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro Oncol. 20, 506–518 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Yin, Y. et al. Locally secreted BiTEs complement CAR T cells by enhancing killing of antigen heterogeneous solid tumors. Mol. Ther. 30, 2537–2553 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Thokala, R. et al. High-affinity chimeric antigen receptor with cross-reactive scFv to clinically relevant EGFR oncogenic isoforms. Front. Oncol. 11, 664236 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gan, H. K., Burgess, A. W., Clayton, A. H. & Scott, A. M. Targeting of a conformationally exposed, tumor-specific epitope of EGFR as a strategy for cancer therapy. Cancer Res. 72, 2924–2930 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Jungbluth, A. A. et al. A monoclonal antibody recognizing human cancers with amplification/overexpression of the human epidermal growth factor receptor. Proc. Natl Acad. Sci. USA 100, 639–644 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Reilly, E. B. et al. Characterization of ABT-806, a humanized tumor-specific anti-EGFR monoclonal antibody. Mol. Cancer Ther. 14, 1141–1151 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Yin, Y. et al. Checkpoint blockade reverses anergy in IL13Rα2 humanized scFv based CAR T cells to treat murine and canine gliomas. Mol. Ther. Oncolytics 11, 20–38 (2018).

  19. Lassman, A. B. et al. Comparison of biomarker assays for EGFR: implications for precision medicine in patients with glioblastoma. Clin. Cancer Res. 25, 3259–3265 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Newman, J. P. et al. Interleukin-13 receptor alpha 2 cooperates with EGFRvIII signaling to promote glioblastoma multiforme. Nat. Commun. 8, 1913 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Ellingson, B. M., Wen, P. Y. & Cloughesy, T. F. Modified criteria for radiographic response assessment in glioblastoma clinical trials. Neurotherapeutics 14, 307–320 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Mahdi, J. et al. Tumor inflammation-associated neurotoxicity. Nat. Med. 29, 803–810 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cappell, K. M. & Kochenderfer, J. N. Long-term outcomes following CAR T cell therapy: what we know so far. Nat. Rev. Clin. Oncol. 20, 359–371 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  25. Good, C. R. et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 184, 6081–6100 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Santomasso, B. D. et al. Management of immune-related adverse events in patients treated with chimeric antigen receptor T-cell therapy: ASCO guideline. J. Clin. Oncol. 39, 3978–3992 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Danylesko, I. et al. Immune imitation of tumor progression after anti-CD19 chimeric antigen receptor T cells treatment in aggressive B-cell lymphoma. Bone Marrow Transplant. 56, 1134–1143 (2021).

    Article  CAS  PubMed  Google Scholar 

  28. Majzner, R. G. et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature 603, 934–941 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Vitanza, N. A. et al. Intraventricular B7-H3 CAR T cells for diffuse intrinsic pontine glioma: preliminary first-in-human bioactivity and safety. Cancer Discov. 13, 114–131 (2023).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the patients who participated in this study and their families for their dedication to furthering GBM treatment. The authors also thank the Neurosurgery Clinical Research Division, the Translational and Correlative Sciences Laboratory and the Clinical Cell and Vaccine Production Facility at the University of Pennsylvania Perelman School of Medicine for all of their clinical trial contributions and support. This work was funded by Kite Pharma (a Gilead company), the Abramson Cancer Center Glioblastoma Translational Center of Excellence to D.M.O., the Templeton Family Initiative in Neuro-Oncology to D.M.O., the Maria and Gabriele Troiano Brain Cancer Immunotherapy Fund to D.M.O., National Institutes of Health grants R35NS116843 to H.S. and R35NS097370 to G.-L.M. and the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation to G.-L.M. Kite Pharma had an advisory role in the design of the study and review of the final manuscript but had no role in data collection, analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Study design: S.J.B., A.S.D., E.M., L.L., A.M., R.L, J.K.J., S.C, B.S.O., G.P., A.B., W.G., D.B., W.-T.H., E.O.H., Z.A.B. and D.M.O. Patient recruitment and treatment: S.J.B., A.S.D., R.M. and E.M. Data generation, curation and analyses: S.J.B., M.L., J.A.F., X.W., A.S.D., L.J.B., A.N., D.J., R.M., C.S., R.L., J.K.J., S.C., V.G., F.C., Y.S., M.P.N., W.-T.H., G.-L.M., H.S., D.L.S., E.O.H., Z.A.B. and D.M.O. Writing—original draft: S.J.B. and Z.A.B. Writing—review and editing: S.J.B., C.H.J., E.O.H., Z.A.B. and D.M.O. Supervision: S.J.B., E.M., L.L., C.S., G.P., A.B., H.S., D.L.S., C.H.J., E.O.H., Z.A.B. and D.M.O. Funding support: W.G. and D.B.

Corresponding authors

Correspondence to Stephen J. Bagley or Donald M. O’Rourke.

Ethics declarations

Competing interests

S.J.B. has received consulting fees from Telix, Servier, Kiyatec, Novocure and Bayer and has received research funding from Kite Pharma (a Gilead company) related to the submitted work and from Incyte, Novocure, GSK and Eli Lilly, all outside of the submitted work. J.A.F. is a member of the scientific advisory boards of Cartography Bio and Shennon Biotechnologies and has patents, royalties and other intellectual property (IP). W.G. is an employee of Kite Pharma (a Gilead company). D.B. is an employee of Kite Pharma (a Gilead company). D.L.S. holds founder’s equity and has licensed IP to Verismo Therapeutics and Vetigenics and has IP licensing to Chimeric Therapeutics. C.H.J. and the University of Pennsylvania have patents pending or issued related to the use of gene modification in T cells for adoptive T cell therapy. C.H.J. is a co-founder of Tmunity (acquired by Kite Pharma, a Gilead company); is a scientific co-founder and holds equity in Capstan Therapeutics, Dispatch Biotherapeutics and BlueWhale Bio; serves on the board of AC Immune; is a scientific advisor to BlueSphereBio, Cabaletta, Carisma, Cartography, Cellares, Cellcarta, Celldex, Danaher, Decheng, ImmuneSensor, Kite Pharma, Poseida, Verismo, Viracta, Vittoria Biotherapeutics and WIRB-Copernicus group; and is an inventor on patents and/or patent applications licensed to Novartis Institutes of Biomedical Research and Kite Pharma and may receive license revenue from such licenses. Z.A.B. has inventorship interest in IP owned by the University of Pennsylvania and has received royalties related to CAR T therapy in solid tumors. D.M.O. reports prior or active roles as consultant/scientific advisory board member for Celldex Therapeutics, Prescient Therapeutics, Century Therapeutics and Chimeric Therapeutics and has received research funding from Celldex Therapeutics, Novartis, Tmunity Therapeutics and Gilead Sciences/Kite Pharma. D.M.O is an inventor of IP (US patent numbers 7,625,558 and 6,417,168 and related families) and has received royalties related to targeted ErbB therapy in solid cancers previously licensed by the University of Pennsylvania. D.M.O is also an inventor on multiple patents related to CAR T cell therapy in solid tumors that have been licensed by the University of Pennsylvania and has received royalties from these license agreements. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Saheli Sadanand, in collaboration with the Nature Medicine team.

Additional information

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

Extended data

Extended Data Fig. 1 CART-EGFR-IL13Rα2 construct, CONSORT diagram, and study schema.

(a). The illustration depicts two parallel CARs with 4-1BBζ intracellular signaling domains. (b) CONSORT flow diagram for the patients included in this report. (c). Study schema.

Extended Data Fig. 2 Flow cytometry plots of optimized CAR detection assay.

For each patient, histograms depict EGFR CAR (left column) and IL13Rα2 CAR (center column) expression. Dual CAR expression is quantified in dot plots (right column). CAR expression values in patient infusion products based on these flow cytometry plots are displayed in Supplementary Table 1.

Extended Data Fig. 3 Immunofluorescence staining of EGFR CAR and IL13Ra2 CAR targets in patient pre-infusion tumor tissue.

Tumor tissue obtained at the time of Ommaya placement was stained for both CAR targets. All 6 patients treated demonstrated expression of one or both targets throughout their tumor. All images taken at 20x magnification. Staining was performed in duplicate. Scale bar = 100 μm.

Extended Data Table 1 Individual patient CAR T cell product information
Extended Data Table 2 Tumor measurements using mRANO criteria

Supplementary information

Supplementary Information

Full Study Protocol

Reporting Summary

Supplementary Tables 1–5

Optimized CAR T cell expression values on patient infusion product. CRS grading system. University of Pennsylvania modified CAR neurotoxicity grading system for patients with GBM. CSF and blood cytokine analyses (pg ml−1). CSF and blood cytokine fold change.

Source data

Source Data Fig. 3

Source data Fig. 3.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bagley, S.J., Logun, M., Fraietta, J.A. et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results. Nat Med (2024). https://doi.org/10.1038/s41591-024-02893-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41591-024-02893-z

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