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:

Single-cell transcriptomic atlas-guided development of CAR-T cells for the treatment of acute myeloid leukemia

Abstract

Chimeric antigen receptor T cells (CAR-T cells) have emerged as a powerful treatment option for individuals with B cell malignancies but have yet to achieve success in treating acute myeloid leukemia (AML) due to a lack of safe targets. Here we leveraged an atlas of publicly available RNA-sequencing data of over 500,000 single cells from 15 individuals with AML and tissue from 9 healthy individuals for prediction of target antigens that are expressed on malignant cells but lacking on healthy cells, including T cells. Aided by this high-resolution, single-cell expression approach, we computationally identify colony-stimulating factor 1 receptor and cluster of differentiation 86 as targets for CAR-T cell therapy in AML. Functional validation of these established CAR-T cells shows robust in vitro and in vivo efficacy in cell line- and human-derived AML models with minimal off-target toxicity toward relevant healthy human tissues. This provides a strong rationale for further clinical development.

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: A scRNA-seq-based screening approach identifies CSF1R and CD86 as potential CAR targets in AML.
Fig. 2: CSF1R and CD86 are preferentially expressed on malignant HSPC-like cells compared to healthy HSPCs, and off-tumor expression is restricted to infiltrating or tissue-resident immune cells.
Fig. 3: mCSF1R CART do not cause toxicity in mice.
Fig. 4: Anti-target CAR-T cells are functional and efficiently lyse AML cell lines in vitro and in vivo.
Fig. 5: CSF1R and CD86 are readily detected on primary AML samples, and hCSF1R CART show efficient lysis of primary AML samples in vitro and in vivo.
Fig. 6: hCSF1R CART show better discriminatory capacity toward healthy human hematopoietic cells than CD33 CART.

Similar content being viewed by others

Data availability

Data from publicly available scRNA-seq studies can be found via the following accession numbers or the provided links: GSE116256 (ref. 21), https://zenodo.org/record/3345981 (ref. 39), https://www.gtexportal.org/home/datasets (ref. 27), GSE134355 (ref. 37), https://www.gutcellatlas.org/ (ref. 36), GSE131907 (ref. 28), GSE115469 (ref. 33), https://www.tissuestabilitycellatlas.org/ (ref. 31), GSE136103 (ref. 34), https://nupulmonary.org/resources/ (ref. 32), https://www.kidneycellatlas.org/ (ref. 29), https://www.synapse.org/#!Synapse:syn21041850/files/ (ref. 30) and EGAS00001002927 (ref. 35). Data from publicly available bulk sequencing studies can be found via the accession numbers E-MTAB-5214 (ref. 82) and E-MTAB-2801 (ref. 83) via the Expression Atlas (https://www.ebi.ac.uk/gxa/home). The surface gene library was obtained by integrating publicly available data23,24,25,26 using OmniPath22 (https://omnipathdb.org/). Targets of FDA-approved drugs were obtained using DrugBank (https://go.drugbank.com/). All reagents and biological material will be made available upon reasonable request to the authors given the agreement by the providing institution.

Code availability

Python scripts for replicating the figures from the scRNA-seq data as jupyter notebooks can be found in the GitHub repository at https://github.com/marrlab/CAR_T_TargetIdentification ref. 96. Count matrices of processed scRNA-seq data will be made available upon reasonable request.

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439–448 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. Raje, N. et al. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N. Engl. J. Med. 380, 1726–1737 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lesch, S. et al. Determinants of response and resistance to CAR T cell therapy. Semin. Cancer Biol. 65, 80–90 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Lamers, C. H. et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol. Ther. 21, 904–912 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cummins, K. D. & Gill, S. Will CAR T cell therapy have a role in AML? Promises and pitfalls. Semin. Hematol. 56, 155–163 (2019).

    Article  PubMed  Google Scholar 

  8. Cancer Genome Atlas Research Network et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

    Article  Google Scholar 

  9. Thol, F. & Ganser, A. Treatment of relapsed acute myeloid leukemia. Curr. Treat. Options Oncol. 21, 66 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Cummins, K. D. & Gill, S. Chimeric antigen receptor T-cell therapy for acute myeloid leukemia: how close to reality? Haematologica 104, 1302–1308 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. MacKay, M. et al. The therapeutic landscape for cells engineered with chimeric antigen receptors. Nat. Biotechnol. 38, 233–244 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Tambaro, F. P. et al. Autologous CD33-CAR-T cells for treatment of relapsed/refractory acute myelogenous leukemia. Leukemia 35, 3282–3286 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sauer, T. et al. CD70-specific CAR T-cells have potent activity against acute myeloid leukemia (AML) without HSC toxicity. Blood 138, 318–330 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jetani, H. et al. Siglec-6 is a novel target for CAR T-cell therapy in acute myeloid leukemia (AML). Blood 138, 1830–1842 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Myburgh, R. et al. Anti-human CD117 CAR T-cells efficiently eliminate healthy and malignant CD117-expressing hematopoietic cells. Leukemia 34, 2688–2703 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Tashiro, H. et al. Treatment of acute myeloid leukemia with T cells expressing chimeric antigen receptors directed to C-type lectin-like molecule 1. Mol. Ther. 25, 2202–2213 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Casucci, M. et al. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood 122, 3461–3472 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Perna, F. et al. Integrating proteomics and transcriptomics for systematic combinatorial chimeric antigen receptor therapy of AML. Cancer Cell 32, 506–519 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Suva, M. L. & Tirosh, I. Single-cell RNA sequencing in cancer: lessons learned and emerging challenges. Mol. Cell 75, 7–12 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Jing, Y. et al. Expression of chimeric antigen receptor therapy targets detected by single-cell sequencing of normal cells may contribute to off-tumor toxicity. Cancer Cell 39, 1558–1559 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. van Galen, P. et al. Single-cell RNA-seq reveals AML hierarchies relevant to disease progression and immunity. Cell 176, 1265–1281 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Turei, D. et al. Integrated intra- and intercellular signaling knowledge for multicellular omics analysis. Mol. Syst. Biol. 17, e9923 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Bausch-Fluck, D. et al. A mass spectrometric-derived cell surface protein atlas. PLoS ONE 10, e0121314 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Bausch-Fluck, D. et al. The in silico human surfaceome. Proc. Natl Acad. Sci. USA 115, E10988–E10997 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Thul, P. J. et al. A subcellular map of the human proteome. Science 356, eaal3321 (2017).

    Article  PubMed  Google Scholar 

  27. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kim, N. et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat. Commun. 11, 2285 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stewart, B. J. et al. Spatiotemporal immune zonation of the human kidney. Science 365, 1461–1466 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Travaglini, K. J. et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature 587, 619–625 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Madissoon, E. et al. scRNA-seq assessment of the human lung, spleen, and esophagus tissue stability after cold preservation. Genome Biol. 21, 1 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Reyfman, P. A. et al. Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 199, 1517–1536 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. MacParland, S. A. et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat. Commun. 9, 4383 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Ramachandran, P. et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cheng, J. B. et al. Transcriptional programming of normal and inflamed human epidermis at single-cell resolution. Cell Rep. 25, 871–883 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. James, K. R. et al. Distinct microbial and immune niches of the human colon. Nat. Immunol. 21, 343–353 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Han, X. et al. Construction of a human cell landscape at single-cell level. Nature 581, 303–309 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Wishart, D. S. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 46, D1074–D1082 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Petti, A. A. et al. A general approach for detecting expressed mutations in AML cells using single cell RNA-sequencing. Nat. Commun. 10, 3660 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Xu, C. et al. Probabilistic harmonization and annotation of single-cell transcriptomics data with deep generative models. Mol. Syst. Biol. 17, e9620 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Jurga, A. M., Paleczna, M. & Kuter, K. Z. Overview of general and discriminating markers of differential microglia phenotypes. Front. Cell Neurosci. 14, 198 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Erblich, B., Zhu, L., Etgen, A. M., Dobrenis, K. & Pollard, J. W. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS ONE 6, e26317 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chihara, T. et al. IL-34 and M-CSF share the receptor FMS but are not identical in biological activity and signal activation. Cell Death Differ. 17, 1917–1927 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Qin, D. et al. Potential lung attack and lethality generated by EpCAM-specific CAR-T cells in immunocompetent mouse models. Oncoimmunology 9, 1806009 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Nayak, D., Roth, T. L. & McGavern, D. B. Microglia development and function. Annu. Rev. Immunol. 32, 367–402 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Edwards, D. K. T. et al. CSF1R inhibitors exhibit antitumor activity in acute myeloid leukemia by blocking paracrine signals from support cells. Blood 133, 588–599 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pabst, C. et al. Identification of small molecules that support human leukemia stem cell activity ex vivo. Nat. Methods 11, 436–442 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Vick, B. et al. An advanced preclinical mouse model for acute myeloid leukemia using patients’ cells of various genetic subgroups and in vivo bioluminescence imaging. PLoS ONE 10, e0120925 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  49. McQuade, A. et al. Development and validation of a simplified method to generate human microglia from pluripotent stem cells. Mol. Neurodegener. 13, 67 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Reifschneider, A. et al. Loss of TREM2 rescues hyperactivation of microglia, but not lysosomal deficits and neurotoxicity in models of progranulin deficiency. EMBO J. 41, e109108 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sletta, K. Y., Castells, O. & Gjertsen, B. T. Colony stimulating factor 1 receptor in acute myeloid leukemia. Front. Oncol. 11, 654817 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Haque, A., Engel, J., Teichmann, S. A. & Lonnberg, T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 9, 75 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Tamura, H. et al. Expression of functional B7-H2 and B7.2 costimulatory molecules and their prognostic implications in de novo acute myeloid leukemia. Clin. Cancer Res. 11, 5708–5717 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Re, F. et al. Expression of CD86 in acute myelogenous leukemia is a marker of dendritic/monocytic lineage. Exp. Hematol. 30, 126–134 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Zheng, Z. et al. Expression patterns of costimulatory molecules on cells derived from human hematological malignancies. J. Exp. Clin. Cancer Res. 17, 251–258 (1998).

    CAS  PubMed  Google Scholar 

  56. Gavile, C. M. et al. CD86 regulates myeloma cell survival. Blood Adv. 1, 2307–2319 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sedek, L. et al. Differential expression of CD73, CD86 and CD304 in normal vs. leukemic B-cell precursors and their utility as stable minimal residual disease markers in childhood B-cell precursor acute lymphoblastic leukemia. J. Immunol. Methods 475, 112429 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Guinan, E. C., Gribben, J. G., Boussiotis, V. A., Freeman, G. J. & Nadler, L. M. Pivotal role of the B7:CD28 pathway in transplantation tolerance and tumor immunity. Blood 84, 3261–3282 (1994).

    Article  CAS  PubMed  Google Scholar 

  59. Zhou, L. J. & Tedder, T. F. CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells. Proc. Natl Acad. Sci. USA 93, 2588–2592 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Smyth, C. et al. Identification of a dynamic intracellular reservoir of CD86 protein in peripheral blood monocytes that is not associated with the Golgi complex. J. Immunol. 160, 5390–5396 (1998).

    Article  CAS  PubMed  Google Scholar 

  61. Blair, H. A. & Deeks, E. D. Abatacept: a review in rheumatoid arthritis. Drugs 77, 1221–1233 (2017).

    Article  CAS  PubMed  Google Scholar 

  62. Adusumilli, P. S. et al. A phase I trial of regional mesothelin-targeted CAR T-cell therapy in patients with malignant pleural disease, in combination with the anti-PD-1 agent pembrolizumab. Cancer Discov. 11, 2748–2763 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Parker, K. R. et al. Single-cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies. Cell 183, 126–142 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Majzner, R. G. & Mackall, C. L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 25, 1341–1355 (2019).

    Article  CAS  Google Scholar 

  65. Jetani, H. et al. CAR T-cells targeting FLT3 have potent activity against FLT3ITD+ AML and act synergistically with the FLT3-inhibitor crenolanib. Leukemia 32, 1168–1179 (2018).

    Article  CAS  PubMed  Google Scholar 

  66. Griciuc, A. et al. TREM2 acts downstream of CD33 in modulating microglial pathology in Alzheimer’s disease. Neuron 103, 820–835 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Cassier, P. A. et al. CSF1R inhibition with emactuzumab in locally advanced diffuse-type tenosynovial giant cell tumours of the soft tissue: a dose-escalation and dose-expansion phase 1 study. Lancet Oncol. 16, 949–956 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. 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 

  69. Gust, J. et al. Endothelial activation and blood–brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 7, 1404–1419 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Sterner, R. M. et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood 133, 697–709 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Tan, A. H. J., Vinanica, N. & Campana, D. Chimeric antigen receptor-T cells with cytokine neutralizing capacity. Blood Adv. 4, 1419–1431 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Virshup, I., Rybakov, S., Theis, F. J., Angerer, P. & Wolf, F. A. anndata: annotated data. Preprint at bioRxiv https://doi.org/10.1101/2021.12.16.473007 (2021).

  74. Lun, A. T., Bach, K. & Marioni, J. C. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75 (2016).

    Article  PubMed  Google Scholar 

  75. Lun, A. T., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Res 5, 2122 (2016).

    PubMed  PubMed Central  Google Scholar 

  76. Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at https://ui.adsabs.harvard.edu/abs/2018arXiv180203426M (2018).

  79. Fischer, D. S. et al. Sfaira accelerates data and model reuse in single cell genomics. Genome Biol. 22, 248 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Muus, C. et al. Single-cell meta-analysis of SARS-CoV-2 entry genes across tissues and demographics. Nat. Med. 27, 546–559 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Polanski, K. et al. BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics 36, 964–965 (2020).

    Article  CAS  PubMed  Google Scholar 

  82. GTEx Consortium Human genomics. The Genotype–Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

    Article  PubMed Central  Google Scholar 

  83. Merkin, J., Russell, C., Chen, P. & Burge, C. B. Evolutionary dynamics of gene and isoform regulation in mammalian tissues. Science 338, 1593–1599 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Papatheodorou, I. et al. Expression Atlas update: from tissues to single cells. Nucleic Acids Res. 48, D77–D83 (2020).

    CAS  PubMed  Google Scholar 

  85. Sandhofer, N. et al. Dual PI3K/mTOR inhibition shows antileukemic activity in MLL-rearranged acute myeloid leukemia. Leukemia 29, 828–838 (2015).

    Article  CAS  PubMed  Google Scholar 

  86. Sudo, T. et al. Functional hierarchy of c-Kit and c-FMS in intramarrow production of CFU-M. Oncogene 11, 2469–2476 (1995).

    CAS  PubMed  Google Scholar 

  87. Benmebarek, M. R. et al. A modular and controllable T cell therapy platform for acute myeloid leukemia. Leukemia 35, 2243–2257 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. van Gosliga, D. et al. Establishing long-term cultures with self-renewing acute myeloid leukemia stem/progenitor cells. Exp. Hematol. 35, 1538–1549 (2007).

    Article  PubMed  Google Scholar 

  89. Herrmann, M. et al. Bifunctional PD-1 × αCD3 × αCD33 fusion protein reverses adaptive immune escape in acute myeloid leukemia. Blood 132, 2484–2494 (2018).

    Article  CAS  PubMed  Google Scholar 

  90. Lesch, S. et al. T cells armed with C-X-C chemokine receptor type 6 enhance adoptive cell therapy for pancreatic tumours. Nat. Biomed. Eng. 5, 1246–1260 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Dimoudis, N. et al. Antibodies binding to human CSF1R extracellular domain 4 and their use. European patent EP 2 510 010 B1 (2012).

  92. Co, M. S. et al. Humanized immunoglobulin reactive with B7-2 and methods of treatment therewith. US patent US2002/0176855A (2002).

  93. Kochenderfer, J. et al. Chimeric antigen receptors targeting CD19. Patent Cooperation Treaty WO 2015/187528 A1 (2015).

  94. Ghani, K. et al. Efficient human hematopoietic cell transduction using RD114- and GALV-pseudotyped retroviral vectors produced in suspension and serum-free media. Hum. Gene Ther. 20, 966–974 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Mulazzani, M. et al. Long-term in vivo microscopy of CAR T cell dynamics during eradication of CNS lymphoma in mice. Proc. Natl Acad. Sci. USA 116, 24275–24284 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Thomas, M. Computational identification of targets for CAR-T cell therapy in AML. GitHub https://github.com/marrlab/CAR_T_TargetIdentification (2023).

Download references

Acknowledgements

This study was supported by the Förderprogramm für Forschung und Lehre der Medizinischen Fakultät der LMU (grant number 1138 to A.G.), the Bavarian Cancer Research Center (A.G.), the Deutsche Forschungsgemeinschaft (DFG; grant number GO 3823/1-1 to A.G.; grant number KO5055/3-1 and KO5055-2-1 to S.K.), the international doctoral program ‘i-Target: immunotargeting of cancer’ (funded by the Elite Network of Bavaria; to S.K. and S.E.), the Melanoma Research Alliance (grant number 409510 to S.K.), Marie Sklodowska-Curie Training Network for the Immunotherapy of Cancer (IMMUTRAIN; funded by the Horizon 2020 program of the European Union; to S.E. and S.K.), Marie Sklodowska-Curie Training Network for Optimizing Adoptive T Cell Therapy of Cancer (funded by the Horizon 2020 program of the European Union; grant 955575 to S.K.), Else Kröner-Fresenius-Stiftung (to S.K.), German Cancer Aid (to S.K.), the Wilhelm-Sander-Stiftung (to S.K.), Ernst Jung Stiftung (to S.K.), Institutional Strategy LMUexcellent of LMU Munich (within the framework of the German Excellence Initiative; to S.E. and S.K.), the Go-Bio-Initiative (to S.K.), the m4-Award of the Bavarian Ministry for Economical Affairs (to S.E. and S.K.), Bundesministerium für Bildung und Forschung (to S.E. and S.K.), European Research Council (Starting Grant 756017 and PoC Grant 101100460 to S.K.), DFG (to S.K.), by the SFB-TRR 338/1 2021–452881907 (to S.K.), Fritz-Bender Foundation (to S.K.), Deutsche José Carreras Leukämie Stiftung (to S.K.), the Bayerische Forschungsstiftung (BAYCELLator to S.K.) and Hector Foundation (to S.K.), DFG (German Research Foundation) under Germany’s Excellence Strategy within the framework of the Munich Cluster for Systems Neurology (EXC 2145 SyNergy, ID 390857198, to E.B. and D.P.) and Alzheimer’s Association (to D.P.). M.T. is funded by the Volkswagen Foundation (project OntoTime). S.S. was supported by the Else Kröner-Fresenius Clinician Scientist Program Cancer Immunotherapy, the Munich Clinician Scientist Program and the DKTK School of Oncology. C.M. has received funding from the European Research Council under the European Union’s Horizon 2020 research and innovation program (grant agreement number 866411). Cytometry data were obtained in the Flow Cytometry Core Facility of the University Hospital, LMU Munich, using a BD LSRFortessa II or Beckman Coulter CytoFLEX. The in vivo imaging device was funded by the DFG (German Research Foundation; INST 409/231-1). We acknowledge Life Science Editors for their editing services. Figure illustrations were created with Biorender.com under a paid subscription.

Author information

Authors and Affiliations

Authors

Contributions

A.G., M.T., R.G., S.L., L.R., V.I., D.B., M.-R.B., S.D., K.M., T.X., D.D., F.M., S.R., A.S., H.S., Ö.U., V.K., C.A.T., E.C., S.N., T.S., T.L., S.S., P.J.M., J.D., M.S., B.L.C., R.B., N.R., F.R. and M.N. performed or assisted with the experiments. A.G., M.T., R.G., S.L., L.R., S.D., K.M., T.X., E.B. and T.H. analyzed the data and supported the project. S.R., B.V., J.W., F.M., W.F., C.K., D.P., I.J. and L.v.B. provided critical reagents, S.K., C.M. and S.E. supervised the project and acquired the funding. A.G., M.T., R.G., S.L., T.H., D.P., L.v.B, S.E., M.S., C.M. and S.K. designed the experiments. A.G., M.T., C.M. and S.K. wrote the paper. All authors critically read and approved the final paper.

Corresponding author

Correspondence to Sebastian Kobold.

Ethics declarations

Competing interests

Parts of this work have been performed for the doctoral thesis of M.T., R.G., D.D. and S.D. at Technische Universität München and LMU München. A.G., M.T., R.G., S.L., S.E., C.M. and S.K. submitted patent applications related to this work filed by the LMU München, the University Hospital of the LMU Munich or the Helmholtz Centre Munich. S.K. has received honoraria from TCR2, Inc., Novartis, BMS and GSK. S.K. and S.E. are inventors of several patents in the field of immunooncology. S.K. and S.E. received license fees from TCR2, Inc., and Carina Biotech. A.G. received research support from Tabby Therapeutics for work unrelated to the paper. S.K. and S.E. received research support from TCR2, Inc., and Arcus Bioscience for work unrelated to the paper. F.M. received support for meeting attendance from Servier, AbbVie, Incyte, Gilead, Jazz Pharmaceuticals, Novartis, Teva, Pfizer and Amgen, received support for medical writing from Servier, received a research grant from Apis Technologies and Daiichi Sankyo and received speaker honoraria from Servier, Jazz Pharmaceuticals and AbbVie. W.F. received payment or honoraria for lectures, presentations, speakers bureaus, paper writing or educational events from Novartis, Abbvie, Pfizer and Amgen. W.F. received support for attending meetings and/or travel from Amgen, Gilead, Jazz Pharmaceuticals, Servier and Daiichi Sankyo. W.F. participates on a Data Safety Monitoring Board or Advisory Board at Amgen, ARIAD/Incyte, Pfizer, Novartis, Jazz Pharmaceuticals, Morphosys, Abbvie, Celgene, Stemline and Clinigen. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Sarah Tasian, Li Tang and Challice Bonifant for their contribution to the peer review of this work.

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 Summary of the cross-organ off-target transcriptomic atlas (COOTA).

(a) Top 100 overexpressed genes in AML HSPC (left) and healthy T cells (right) from differential expression analysis. Normalized expression values were logarithmized and scaled to unit variance. (b) Overview of 11 scRNA-seq datasets of various healthy tissues used to quantify off-target antigen expression. (c) UMAP plots of 11 scRNA-seq datasets with colors highlighting clustering into respective cell types. Cell annotations were provided by the authors of the respective studies. (d) Current CAR targets in AML were cross-referenced to filters used for the single cell-based target screening approach. OE HSC-/Prog-like: overexpressed on HSC-/Prog-like cells with log fold change > 2 and FDR-adjusted p ≤ 0.01, using a t-test with overestimated variance. Red cross: Antigen did not fulfill the respective threshold or criteria. Green check: Thresholds or criteria were passed.

Extended Data Fig. 2 CSF1R and CD86 are consistently expressed across multiple patients with differing molecular subtypes.

(a, c) Amount of malignant and normal cells per AML patient of van Galen et al. (a) or Petti et al. (c). (b, d) Percentage of malignant and normal cells expressing target genes CSF1R, CD86 and reference genes CD123, CD33 for each sequenced AML patient of van Galen et al. (b) or Petti et al. (d). (e) Data from van Galen et al.21 was used as a reference (top left) to map cells from Petti et al.39 (top right) using scANVI40. UMAP representation showing the mapped query and reference data together (bottom). (f) Computational CAR target antigen identification using the mapped dataset of Petti et al. by stepwise evaluation against a set of criteria for an ideal and effective CAR target antigen. The decreasing number of screened AML target genes are shown on the bottom. CSPA: Cell surface protein atlas; HPA: Human protein atlas. (g) Volcano plot showing CD86 and CSF1R target genes with their respective FDR-adjusted log10 p-value and log2 fold changes from differential expression analysis between malignant HSPC-like and healthy HSPC using a t-test with overestimated variance.

Extended Data Fig. 3 mCSF1R CART do not persist in immunocompetent mice.

(a) Construct design of mCSF1R or mEpCAM CART or mCherry T cells. (b) Representative histograms of mCSF1R or mEpCAM expression on J774A.1 cells. Staining was carried out twice. (c) Summary of treatment schedule for in vivo toxicity assessment of mCSF1R CART. (d) Mean weight curves of mice treated with 3 × 106 mCSF1R CART or mCherry T cells. n = 10 mice per group. Error bars indicate s.e.m. (e) Quantification of tissue-resident CD11b + cells (left) or mCherry+ T cells of parent population (right, parent population: CD3 and CD8 positive cells) in different organs by flow cytometry. Data are mean ± s.e.m. of n = 10 mice. (d, e) Statistical significance was calculated using two-way ANOVA with Šidák multiple comparison correction. (f) Scheme of treatment schedule for in vivo toxicity assessment of mCSF1R CART. WBI, whole body irradiation. Mice were treated with 3 × 106 mCSF1R CART or mCherry T cells per mouse. 6 × 106 mEpCAM CART were transferred as a positive control. (g) Serum levels of indicated markers one (d1) or seven (d7) days after ACT. Depicted is the fold change of serum levels of the indicated groups from the PBS-treated control group. CRP, C-reactive protein; AST, aspartate aminotransferase; ALT, alanine aminotransferase; GLDH, glutamate dehydrogenase; γ-GT, gamma-glutamyl-transferase; LDH, lactate dehydrogenase. n = 3 mice per group. Statistically significant increases in the serum of mEpCAM CART treated mice compared to mCSF1R CART or control-treated mice at day 7 were observed for GLDH (p < 0.0001). (h) Histopathological analysis of indicated organs after treatment with mEpCAM or mCSF1R CART or mCherry T cells. Representative images of n = 3 mice per group. Signs of organ damage are indicated in the picture: black arrowhead: thickening of alveolar epithel. (i) Representative maximum intensity projection of microglia (green) and macrophages (green) in CX3CR1-GFP mice on days 0 (baseline), 4, 7, 10, 14, 21 and 28 after intravenous injection of 107 mCSF1R CART (red). Depth from brain surface: 0–100 µm. Scale bars as depicted: 50 µm and 20 µm. mCSF1R CART: i.c. injection, n = 5 mice; i.v. injection, n = 3 mice; mCherry T cells: i.c./ i.v. injection = 2 mice.

Extended Data Fig. 4 Assessment of different hCSF1R CAR constructs in vitro.

(a) Construct design of all anti-human constructs used throughout the course of the study. (b) Representative flow cytometric images of construct expression on primary human T cells. (c) Activation of different hCSF1R CART after incubation with plate-bound hCSF1R protein quantified by flow cytometry. (d) T cells expressing different hCSF1R CAR constructs were cocultured with luc+ target antigen expressing AML tumor cell lines or antigen negative NALM-6 control cells for 48 hours at the indicated E:T ratios. Cell lysis was quantified by BLI. (e) Proliferation dye-labeled hCSF1R CART were cocultured with respective cell lines for 4 or 7 days at a E:T ratio of 0.5:1. Proliferation was subsequently assessed by trace dilution. One representative image of three different donors is shown. (f) Bead quantified T cell numbers. (g) Secretion of IFNγ by T cells transduced with different hCSF1R CAR constructs after co-culture with AML cell lines. (h) Secretion of IFNγ (left) or IL-2 (right) of hCSF1R, CD86 or control CART in co-culture with AML cell lines. (c, d, f - h) Data are mean ± s.e.m. of three independent donors. LTR, long terminal repeat; scFv, single chain fragment variable; TM, transmembrane, IC, intracellular, ED, extracellular domain, CTRL-transduced, control-transduced.

Extended Data Fig. 5 CSF1R is highly expressed on primary AML blasts.

(a) Representative histograms of CSF1R expression on AML cell lines after freeze and thaw cycles. (b, c) Schematic of culture methods used to cultivate primary AML samples throughout the course of the study. (d) Expression of CSF1R on primary AML samples after 24 to 48 hours of culture in cytokine rich medium. Left: Percentage positive cells gated to isotype. Each dot represents different primary AML samples. Right: Representative flow cytometric images from three different AML samples. Data are mean ± s.e.m. from six different donors. (e) Gating strategy to identify CD34+ CD38 ± malignant HSPC. (f, g) Expression of target antigens (CSF1R, f; CD86, g) on malignant HPC (top) and HSC (bottom). (h) Expression of CSF1R on PDX-388 sample at indicated time points after thawing. (i) IHC staining of human CSF1R in the bone marrow of control-treated PDX-388-bearing mice. Shown are two representative pictures (right, left) in two different magnifications (top 10x, bottom 20x). (j) CD33 CART used for i.v. injection into PDX-388-bearing mice (Fig. 5k–m) were cocultured ex vivo with luciferase+ Mv4-11 tumor cells for 48 hours at indicated E:T ratios. Specific lysis was quantified by BLI. Shown is mean ± s.e.m. of three biological replicates. Experiment was carried out twice. (k) Representative flow cytometric image of percentage of CD3+ T cells (left) and percentage of CAR (c-myc)+ T cells (right) in the blood of PDX-388-bearing mice. (l, m) Ex vivo CD33 expression on PDX-388 AML blasts in the bone marrow after treatment with CD33 CART or CTRL-transduced T cells (CD19 CART) measured by flow cytometry. Depicted are representative histograms (l) or the change of CD33-PE-Cy5 MFI in CD33 CART treated mice compared to CTRL-transduced treated mice (m). (m) Data are mean ± s.e.m. from n = 3 mice injected with CD33 CART compared to CTRL-transduced mouse. (km) n = 3 mice injected with CD33 CART), n = 1 mouse injected with CTRL-transuced T cells.

Extended Data Fig. 6 hCSF1R CAR T cells are effective in vivo.

(a) Treatment scheme used for PDX-372 model. (b–d) BLI images (b), BLI quantification of tumor-burden (c) and survival curves (d) of PDX-372 tumor-bearing mice injected with 2 × 106 hCSF1R, CD33 CART or control-transduced T cells (n = 5 mice per group). (b) White cross, censored mice; red cross, mice succumbed to disease. (e) IHC staining of human CSF1R in the bone marrow of control-treated PDX-372-bearing mice. Left: IHC for human CSF1R. Right: Isotype (top) and detection system control (bottom) for CSF1R IHC staining. (f) Schematic of treatment scheme used for OCI-AML3 cell line xenograft model. (g, h) BLI images (g) and survival curves (h) of OCI-AML3 tumor-bearing mice injected with 6 × 106 hCSF1R CART or control-transduced T cells (n = 3–4 mice per group). (g) Red cross, mice succumbed to disease. (ah) Statistical significance was calculated using two-way ANOVA with Šidák multiple comparison correction. (i) log2 expression of CSF1R and CD86 target antigens or CD123 and CD33 controls in bulk RNA-sequencing dataset of the Leukemia MILE study (n = 615 different patients). HBM, healthy bone marrow. Data was obtained from bloodspot.eu. Dashed line represents the median, dotted line the interquartile ranges. Statistical significance was calculated using ordinary one-way ANOVA with Šidák multiple comparison correction. (j) Simple linear regression analysis of in vitro lysis of CAR T cells and target antigen density of the indicated AML cell line measured by flow cytometry. r = spearman correlation coefficient, p = p-value. Three independent antigen density measurements were used to perform regression analysis. For Kaplan-Meier-Curves, statistical significance was calculated with log-rank test.

Extended Data Table 1 Summary of patient characteristics of all primary human patient samples used during the study

Supplementary information

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

Gottschlich, A., Thomas, M., Grünmeier, R. et al. Single-cell transcriptomic atlas-guided development of CAR-T cells for the treatment of acute myeloid leukemia. Nat Biotechnol 41, 1618–1632 (2023). https://doi.org/10.1038/s41587-023-01684-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-023-01684-0

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research