Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia (original) (raw)

Change history

A Correction to this paper has been published: https://doi.org/10.1038/s41591-021-01248-2

References

  1. Makkouk, A. & Weiner, G. J. Cancer immunotherapy and breaking immune tolerance: new approaches to an old challenge. Cancer Res. 75, 5–10 (2015).
    Article CAS PubMed Google Scholar
  2. Vesely, M. D., Kershaw, M. H., Schreiber, R. D. & Smyth, M. J. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 29, 235–271 (2011).
    Article CAS PubMed Google Scholar
  3. Brown, J. R. et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110delta, for relapsed/refractory chronic lymphocytic leukemia. Blood 123, 3390–3397 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  4. Jain, P. et al. Outcomes of patients with chronic lymphocytic leukemia after discontinuing ibrutinib. Blood 125, 2062–2067 (2015).
    Article CAS PubMed PubMed Central Google Scholar
  5. Kalos, M. et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 3, 95ra73 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  6. Porter, D. L. et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl. Med. 7, 303ra139 (2015).
    Article PubMed PubMed Central Google Scholar
  7. 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 CAS Google Scholar
  8. Turtle, C. J. et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl. Med. 8, 355ra116 (2016).
    Article PubMed PubMed Central CAS Google Scholar
  9. Lee, D. W. et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385, 517–528 (2015).
    Article CAS PubMed Google Scholar
  10. Turtle, C. J. et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 126, 2123–2138 (2016).
    Article PubMed PubMed Central Google Scholar
  11. Kochenderfer, J. N. et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J. Clin. Oncol. 33, 540–549 (2015).
    Article CAS PubMed Google Scholar
  12. Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity 44, 712 (2016).
    Article CAS PubMed Google Scholar
  13. Gattinoni, L. et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J. Clin. Invest. 115, 1616–1626 (2005).
    Article CAS PubMed PubMed Central Google Scholar
  14. Hinrichs, C. S. et al. Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc. Natl. Acad. Sci. USA 106, 17469–17474 (2009).
    Article CAS PubMed PubMed Central Google Scholar
  15. Gattinoni, L. et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  16. Radvanyi, L. G. et al. Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin. Cancer Res. 18, 6758–6770 (2012).
    Article CAS PubMed PubMed Central Google Scholar
  17. Rosenberg, S. A. et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 17, 4550–4557 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  18. Wirth, T. C. et al. Repetitive antigen stimulation induces stepwise transcriptome diversification but preserves a core signature of memory CD8+ T cell differentiation. Immunity 33, 128–140 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  19. Betz, U. A. & Müller, W. Regulated expression of gp130 and IL-6 receptor alpha chain in T cell maturation and activation. Int. Immunol. 10, 1175–1184 (1998).
    Article CAS PubMed Google Scholar
  20. Siegel, A. M. et al. A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory. Immunity 35, 806–818 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  21. Lugli, E. et al. Identification, isolation and in vitro expansion of human and nonhuman primate T stem cell memory cells. Nat. Protoc. 8, 33–42 (2013).
    Article CAS PubMed Google Scholar
  22. Bruggner, R. V., Bodenmiller, B., Dill, D. L., Tibshirani, R. J. & Nolan, G. P. Automated identification of stratifying signatures in cellular subpopulations. Proc. Natl. Acad. Sci. USA 111, E2770–E2777 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  23. Aghaeepour, N. et al. Early immunologic correlates of HIV protection can be identified from computational analysis of complex multivariate T-cell flow cytometry assays. Bioinformatics 28, 1009–1016 (2012).
    Article CAS PubMed PubMed Central Google Scholar
  24. Lécuroux, C. et al. Identification of a particular HIV-specific CD8+ T-cell subset with a CD27+ CD45RO–/RA+ phenotype and memory characteristics after initiation of HAART during acute primary HIV infection. Blood 113, 3209–3217 (2009).
    Article PubMed CAS Google Scholar
  25. Dunne, P. J. et al. Epstein-Barr virus-specific CD8+ T cells that re-express CD45RA are apoptosis-resistant memory cells that retain replicative potential. Blood 100, 933–940 (2002).
    Article CAS PubMed Google Scholar
  26. Rufer, N. et al. Ex vivo characterization of human CD8+ T subsets with distinct replicative history and partial effector functions. Blood 102, 1779–1787 (2003).
    Article CAS PubMed Google Scholar
  27. Precopio, M. L. et al. Immunization with vaccinia virus induces polyfunctional and phenotypically distinctive CD8+ T cell responses. J. Exp. Med. 204, 1405–1416 (2007).
    Article CAS PubMed PubMed Central Google Scholar
  28. Kueberuwa, G. et al. CCR7+ selected gene-modified T cells maintain a central memory phenotype and display enhanced persistence in peripheral blood in vivo. J. Immunother. Cancer 5, 14 (2017).
    Article PubMed PubMed Central Google Scholar
  29. Jones, R. B. et al. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J. Exp. Med. 205, 2763–2779 (2008).
    Article CAS PubMed PubMed Central Google Scholar
  30. Barber, D. L. et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687 (2006).
    Article CAS PubMed Google Scholar
  31. Blackburn, S. D. et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 10, 29–37 (2009).
    Article CAS PubMed Google Scholar
  32. Ahmadzadeh, M. et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114, 1537–1544 (2009).
    Article CAS PubMed PubMed Central Google Scholar
  33. Fourcade, J. et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 207, 2175–2186 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  34. Grosso, J. F. et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J. Clin. Invest. 117, 3383–3392 (2007).
    Article CAS PubMed PubMed Central Google Scholar
  35. Grosso, J. F. et al. Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J. Immunol. 182, 6659–6669 (2009).
    Article CAS PubMed Google Scholar
  36. Milone, M. C. et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. 17, 1453–1464 (2009).
    Article CAS PubMed PubMed Central Google Scholar
  37. Laport, G. G. et al. Adoptive transfer of costimulated T cells induces lymphocytosis in patients with relapsed/refractory non-Hodgkin lymphoma following CD34+-selected hematopoietic cell transplantation. Blood 102, 2004–2013 (2003).
    Article CAS PubMed Google Scholar
  38. Ruella, M. et al. The addition of the BTK inhibitor Ibrutinib to anti-CD19 chimeric antigen receptor T cells (CART19) improves responses against mantle cell lymphoma. Clin. Cancer Res. 22, 2684–2696 (2016).
    Article CAS PubMed Google Scholar
  39. Fraietta, J. A. et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood 127, 1117–1127 (2016).
    Article CAS PubMed PubMed Central Google Scholar
  40. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
    Article CAS PubMed Google Scholar
  41. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).
    Article CAS PubMed PubMed Central Google Scholar
  42. Anders, S., Pyl, P. T. & Huber, W. HTSeq: a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).
    Article CAS PubMed Google Scholar
  43. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).
    Article CAS PubMed Google Scholar
  44. Luckey, C. J. et al. Memory T and memory B cells share a transcriptional program of self-renewal with long-term hematopoietic stem cells. Proc. Natl. Acad. Sci. USA 103, 3304–3309 (2006).
    Article CAS PubMed PubMed Central Google Scholar
  45. McKinney, E. F., Lee, J. C., Jayne, D. R., Lyons, P. A. & Smith, K. G. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015).
    Article CAS PubMed PubMed Central Google Scholar
  46. Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).
    Article CAS PubMed Google Scholar
  47. Abbas, A. R. et al. Immune response in silico (IRIS): immune-specific genes identified from a compendium of microarray expression data. Genes Immun. 6, 319–331 (2005).
    Article CAS PubMed Google Scholar
  48. Bangs, S. C. et al. Human CD4+ memory T cells are preferential targets for bystander activation and apoptosis. J. Immunol. 182, 1962–1971 (2009).
    Article CAS PubMed Google Scholar
  49. Ramirez, K. et al. Gene deregulation and chronic activation in natural killer cells deficient in the transcription factor ETS1. Immunity 36, 921–932 (2012).
    Article CAS PubMed PubMed Central Google Scholar
  50. Winter, S. C. et al. Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers. Cancer Res. 67, 3441–3449 (2007).
    Article CAS PubMed Google Scholar
  51. Campia, I. et al. An autocrine cytokine/JAK/STAT-signaling induces kynurenine synthesis in multidrug resistant human cancer cells. PLoS One 10, e0126159 (2015).
    Article PubMed PubMed Central CAS Google Scholar
  52. Jena, B. et al. Chimeric antigen receptor (CAR)-specific monoclonal antibody to detect CD19-specific T cells in clinical trials. PLoS One 8, e57838 (2013).
    Article CAS PubMed PubMed Central Google Scholar
  53. Cawthon, R. M. Telomere measurement by quantitative PCR. Nucleic Acids Res. 30, e47 (2002).
    Article PubMed PubMed Central Google Scholar
  54. Kim, N. W. et al. Specific association of human telomerase activity with immortal cells and cancer. Science 266, 2011–2015 (1994).
    Article CAS PubMed Google Scholar
  55. Herbert, B. S., Hochreiter, A. E., Wright, W. E. & Shay, J. W. Nonradioactive detection of telomerase activity using the telomeric repeat amplification protocol. Nat. Protoc. 1, 1583–1590 (2006).
    Article CAS PubMed Google Scholar

Download references

Acknowledgements

We thank the patients for their participation in the clinical trials from which research samples were obtained. We also acknowledge A. Fesnak, A. Lamontagne, A. Malykhin, C. Corl, Y. Ohayon and other members of the Clinical Cell and Vaccine Production Facility for cell manufacturing and testing. In addition, we are grateful to V. Gonzalez, J. Finklestein, F. Nazimuddin, J.-M. Navenot, M. Bogush, Y. Tanner, N. Kengle, K. Marcucci, A. Chew, C. Pletcher, P. Hallberg and R. Schretzenmair for contributions to correlative studies and/or other research support. D. Campana, C. Imai and others at St. Jude Children’s Research Hospital designed, developed and provided, under material-transfer agreements, the CAR used in this study. B. Jena and L. Cooper (MD Anderson Cancer Center) are acknowledged for providing the CAR anti-idiotype detection reagent. The functional anti-idiotypic antibody that was used for in vitro CAR stimulation experiments was a kind gift from Novartis Pharmaceutical Corporation. This work was supported by funding from NCI T32CA009140 (J.A.F.) R01CA165206 (D.L.P. and C.H.J.), P01CA214278 (C.H.J), a Stand Up to Cancer Phillip A. Sharp Innovation in Collaboration Award (C.H.J) and Novartis.

Author information

Author notes

  1. These authors contributed equally: Simon F. Lacey and Elena J. Orlando.

Authors and Affiliations

  1. Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
    Joseph A. Fraietta, Simon F. Lacey, F. Brad Johnson, Megan M. Davis, Bruce L. Levine, Donald L. Siegel, David L. Porter, Carl H. June & J. Joseph Melenhorst
  2. Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
    Joseph A. Fraietta, Simon F. Lacey, Mercy Gohil, Stefan Lundh, Alina C. Boesteanu, Yan Wang, Roddy S. O’Connor, Edward Pequignot, David E. Ambrose, Changfeng Zhang, Nicholas Wilcox, Felipe Bedoya, Corin Dorfmeier, Fang Chen, Lifeng Tian, Harit Parakandi, Minnal Gupta, Regina M. Young, Irina Kulikovskaya, Li Liu, Jun Xu, Megan M. Davis, Bruce L. Levine, Noelle V. Frey, Donald L. Siegel, Carl H. June & J. Joseph Melenhorst
  3. Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
    Joseph A. Fraietta, Simon F. Lacey, Alexander C. Huang, E. John Wherry, Carl H. June & J. Joseph Melenhorst
  4. Novartis Institutes for BioMedical Research, Cambridge, MA, USA
    Elena J. Orlando, Iulian Pruteanu-Malinici, Sadik H. Kassim, Hans Bitter & Jennifer L. Brogdon
  5. Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, USA
    Wei-Ting Hwang
  6. Division of Hematology-Oncology, Department of Internal Medicine, University of Pennsylvania, Philadelphia, PA, USA
    Noelle V. Frey & David L. Porter
  7. Division of Transfusion Medicine and Therapeutic Pathology, University of Pennsylvania, Philadelphia, PA, USA
    Donald L. Siegel
  8. Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
    Alexander C. Huang & E. John Wherry

Authors

  1. Joseph A. Fraietta
    You can also search for this author inPubMed Google Scholar
  2. Simon F. Lacey
    You can also search for this author inPubMed Google Scholar
  3. Elena J. Orlando
    You can also search for this author inPubMed Google Scholar
  4. Iulian Pruteanu-Malinici
    You can also search for this author inPubMed Google Scholar
  5. Mercy Gohil
    You can also search for this author inPubMed Google Scholar
  6. Stefan Lundh
    You can also search for this author inPubMed Google Scholar
  7. Alina C. Boesteanu
    You can also search for this author inPubMed Google Scholar
  8. Yan Wang
    You can also search for this author inPubMed Google Scholar
  9. Roddy S. O’Connor
    You can also search for this author inPubMed Google Scholar
  10. Wei-Ting Hwang
    You can also search for this author inPubMed Google Scholar
  11. Edward Pequignot
    You can also search for this author inPubMed Google Scholar
  12. David E. Ambrose
    You can also search for this author inPubMed Google Scholar
  13. Changfeng Zhang
    You can also search for this author inPubMed Google Scholar
  14. Nicholas Wilcox
    You can also search for this author inPubMed Google Scholar
  15. Felipe Bedoya
    You can also search for this author inPubMed Google Scholar
  16. Corin Dorfmeier
    You can also search for this author inPubMed Google Scholar
  17. Fang Chen
    You can also search for this author inPubMed Google Scholar
  18. Lifeng Tian
    You can also search for this author inPubMed Google Scholar
  19. Harit Parakandi
    You can also search for this author inPubMed Google Scholar
  20. Minnal Gupta
    You can also search for this author inPubMed Google Scholar
  21. Regina M. Young
    You can also search for this author inPubMed Google Scholar
  22. F. Brad Johnson
    You can also search for this author inPubMed Google Scholar
  23. Irina Kulikovskaya
    You can also search for this author inPubMed Google Scholar
  24. Li Liu
    You can also search for this author inPubMed Google Scholar
  25. Jun Xu
    You can also search for this author inPubMed Google Scholar
  26. Sadik H. Kassim
    You can also search for this author inPubMed Google Scholar
  27. Megan M. Davis
    You can also search for this author inPubMed Google Scholar
  28. Bruce L. Levine
    You can also search for this author inPubMed Google Scholar
  29. Noelle V. Frey
    You can also search for this author inPubMed Google Scholar
  30. Donald L. Siegel
    You can also search for this author inPubMed Google Scholar
  31. Alexander C. Huang
    You can also search for this author inPubMed Google Scholar
  32. E. John Wherry
    You can also search for this author inPubMed Google Scholar
  33. Hans Bitter
    You can also search for this author inPubMed Google Scholar
  34. Jennifer L. Brogdon
    You can also search for this author inPubMed Google Scholar
  35. David L. Porter
    You can also search for this author inPubMed Google Scholar
  36. Carl H. June
    You can also search for this author inPubMed Google Scholar
  37. J. Joseph Melenhorst
    You can also search for this author inPubMed Google Scholar

Contributions

J.A.F., S.F.L., F.B.J., R.M.Y., N.V.F., B.L.L., D.L.S., E.J.W., J.L.B., D.L.P., C.H.J. and J.J.M. designed the experiments and/or performed analysis. J.A.F., M.Gohil, S.L., A.C.B., Y.W., R.S.O., D.E.A., C.Z., N.W., F.B., C.D., F.C., L.T., H.P., M. Gupta, I.K., L.L., J.X., S.H.K., M.M.D. and A.C.H. performed experiments. E.J.O. and H.B. analyzed RNA-seq. data. I.P.-M. carried out the computational analyses of flow cytometric data. W.-T.H. and E.P. performed statistical analyses. J.A.F., C.H.J. and J.J.M. wrote the paper, and all authors contributed to writing and providing feedback.

Corresponding author

Correspondence toJ. Joseph Melenhorst.

Ethics declarations

Competing interests

J.A.F., S.F.L., F.B., R.M.Y., B.L.L., J.L.B., D.L.P., C.H.J. and J.J.M. hold patents related to CTL019 cell therapy. These authors declare no additional interests. The remaining authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 and Supplementary Tables 1, 7, 9, 10

Reporting Summary

Supplementary Table 2

Treatment and clinical characteristics of responding patients.

Supplementary Table 3

Genes differentially expressed in pre-infusion CAR T cells between CR/PRTD and PR/NR patients.

Supplementary Table 4

List of leading edge genes associated with the gene sets of Figure 2c.

Supplementary Table 5

Transcriptomic profiling of mock-stimulated (control) and CAR-stimulated CTL019 infusion products as well as ex vivo CD3+ T cells (leukapheresis).

Supplementary Table 6

Phenotypes of leukapheresed CD8+ and CD4+ T cells identified by the flowType analysis that segregate CR from NR patients.

Supplementary Table 8

Phenotypes of CD8+ T cells in the CAR T cell infusion product identified by the flowType analysis that segregate CR from NR patients.

Supplementary Table 11

Holm-Sidak adjusted P values and summary statistics for data sets involving multiple comparisons.

Rights and permissions

About this article

Cite this article

Fraietta, J.A., Lacey, S.F., Orlando, E.J. et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia.Nat Med 24, 563–571 (2018). https://doi.org/10.1038/s41591-018-0010-1

Download citation