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.

  • Review Article
  • Published:

T cell-engaging therapies — BiTEs and beyond

Abstract

Immuno-oncology approaches have entered clinical practice, with tremendous progress particularly in the field of T cell-engaging therapies over the past decade. Herein, we provide an overview of the current status of bispecific T cell engager (BiTE) therapy, considering the unprecedented new indication for such therapy in combating minimal (or measurable) residual disease in patients with acute lymphoblastic leukaemia, and the development of novel approaches based on this concept. Key aspects that we discuss include the current clinical data, challenges relating to treatment administration and patient monitoring, toxicities and resistance to treatment, and novel strategies to overcome these hurdles as well as to broaden the indications for BiTE therapy, particularly to common solid cancers. Elucidation of mechanisms of resistance and immune escape and new technologies used in drug development pave the way for new and more-effective therapies and rational combinatorial approaches. In particular, we highlight novel therapeutic agents, such as bifunctional checkpoint-inhibitory T cell engagers (CiTEs), simultaneous multiple interaction T cell engagers (SMITEs), trispecific killer engagers (TriKEs) and BiTE-expressing chimeric antigen receptor (CAR) T cells (CART.BiTE cells), designed to integrate various immune functions into one molecule or a single cellular vector and thereby enhance efficacy without compromising safety. We also discuss the targeting of intracellular tumour-associated epitopes using bispecific constructs with T cell receptor (TCR)-derived, rather than an antibody-based, antigen-recognition domains, termed immune-mobilizing monoclonal TCRs against cancer (ImmTACs), which might broaden the armamentarium of T cell-engaging therapies.

Key points

  • The first-in-class bispecific T cell engager (BiTE), blinatumomab, has been approved for the treatment of patients with relapsed and/or refractory B cell-precursor acute lymphoblastic leukaemia (B-ALL).

  • Blinatumomab is also the first targeted immunotherapy approved specifically for the treatment of minimal residual disease in patients with B-ALL who are in haematological complete remission.

  • BiTE therapy might be effective not only in a small group of B cell malignancies, but also in a broader range of malignancies, including common types of solid cancer.

  • An understanding of the mechanisms of resistance and immune escape might pave the way for innovative drug development with the intention to mitigate toxicity and to enhance efficacy.

  • New constructs, such as bifunctional checkpoint-inhibitory T cell engagers (CiTEs), simultaneous multiple interaction T cell engagers (SMITEs), trispecific killer engagers (TriKEs) and BiTE-expressing chimeric antigen receptor (CAR) T cells (CART.BiTE cells), have been developed to integrate various immune functions into one therapeutic approach or cellular vector, thereby enhancing anticancer activity without substantially increasing the risk of immune-related adverse effects.

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: BiTE antibodies — mechanisms of action and resistance, and strategies to restore T cell function.
Fig. 2: Schematic overview of novel killer cell-engaging constructs for antibody-based or TCR-based tumour cell targeting.

Similar content being viewed by others

References

  1. McLaughlin, P. et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J. Clin. Oncol. 16, 2825–2833 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Vokes, E. E. et al. Nivolumab versus docetaxel in previously treated advanced non-small-cell lung cancer (CheckMate 017 and CheckMate 057): 3-year update and outcomes in patients with liver metastases. Ann. Oncol. 29, 959–965 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Reck, M. et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 375, 1823–1833 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Garon, E. B. et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 372, 2018–2028 (2015).

    Article  PubMed  Google Scholar 

  7. Sharma, P. et al. Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): a multicentre, open-label, two-stage, multi-arm, phase 1/2 trial. Lancet Oncol. 17, 1590–1598 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kao, H.-F. & Lou, P.-J. Immune checkpoint inhibitors for head and neck squamous cell carcinoma: current landscape and future directions. Head Neck 41, 4–18 (2019).

    Article  PubMed  Google Scholar 

  9. Hizal, M. et al. A historical turning point for the treatment of advanced renal cell carcinoma: inhibition of immune checkpoint. Curr. Med. Res. Opin. https://doi.org/10.1080/03007995.2020.1716705 (2020).

    Article  PubMed  Google Scholar 

  10. Zhang, X. et al. Safety and efficacy in relapsed or refractory classic Hodgkin’s lymphoma treated with PD-1 inhibitors: a meta-analysis of 9 prospective clinical trials. BioMed. Res. Int. 2019, 9283860 (2019).

    PubMed  PubMed Central  Google Scholar 

  11. Zolkind, P. & Uppaluri, R. Checkpoint immunotherapy in head and neck cancers. Cancer Metastasis Rev. 36, 475–489 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Stühler, V., Maas, J. M., Rausch, S., Stenzl, A. & Bedke, J. Immune checkpoint inhibition for the treatment of renal cell carcinoma. Expert Opin. Biol. Ther. 20, 83–94 (2020).

    Article  PubMed  CAS  Google Scholar 

  13. Eggermont, A. M. et al. Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy. N. Engl. J. Med. 375, 1845–1855 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hamilton, G. & Rath, B. Immunotherapy for small cell lung cancer: mechanisms of resistance. Expert Opin. Biol. Ther. 19, 423–432 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Veldman, J., Visser, L., Berg, A.v.d. & Diepstra, A. Primary and acquired resistance mechanisms to immune checkpoint inhibition in Hodgkin lymphoma. Cancer Treat. Rev. 82, 101931 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Havel, J. J., Chowell, D. & Chan, T. A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer 19, 133–150 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kobold, S. Innate and adaptive immunity combined for cancer treatment. Proc. Natl Acad. Sci. USA 116, 1087–1088 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zappasodi, R., Wolchok, J. D. & Merghoub, T. Strategies for predicting response to checkpoint inhibitors. Curr. Hematol. Malig. Rep. 13, 383–395 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Dreier, T. et al. T cell costimulus-independent and very efficacious inhibition of tumor growth in mice bearing subcutaneous or leukemic human B cell lymphoma xenografts by a CD19-/CD3- bispecific single-chain antibody construct. J. Immunol. 170, 4397–4402 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Staerz, U. D., Kanagawa, O. & Bevan, M. J. Hybrid antibodies can target sites for attack by T cells. Nature 314, 628–631 (1985).

    Article  CAS  PubMed  Google Scholar 

  22. Brinkmann, U. & Kontermann, R. E. The making of bispecific antibodies. mAbs 9, 182–212 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Spiess, C., Zhai, Q. & Carter, P. J. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol. Immunol. 67, 95–106 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Felices, M., Lenvik, T. R., Davis, Z. B., Miller, J. S. & Vallera, D. A. Generation of BiKEs and TriKEs to improve NK cell-mediated targeting of tumor cells. Methods Mol. Biol. 1441, 333–346 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chames, P., Van Regenmortel, M., Weiss, E. & Baty, D. Therapeutic antibodies: successes, limitations and hopes for the future. Br. J. Pharmacol. 157, 220–233 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ruf, P. et al. Pharmacokinetics, immunogenicity and bioactivity of the therapeutic antibody catumaxomab intraperitoneally administered to cancer patients. Br. J. Clin. Pharmacol. 69, 617–625 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chames, P. & Baty, D. Bispecific antibodies for cancer therapy: the light at the end of the tunnel? mAbs 1, 539–547 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Mack, M., Riethmuller, G. & Kufer, P. A small bispecific antibody construct expressed as a functional single-chain molecule with high tumor cell cytotoxicity. Proc. Natl Acad. Sci. USA 92, 7021–7025 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Loffler, A. et al. A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood 95, 2098–2103 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Dreier, T. et al. Extremely potent, rapid and costimulation-independent cytotoxic T-cell response against lymphoma cells catalyzed by a single-chain bispecific antibody. Int. J. Cancer 100, 690–697 (2002).

    Article  CAS  PubMed  Google Scholar 

  31. Loffler, A. et al. Efficient elimination of chronic lymphocytic leukaemia B cells by autologous T cells with a bispecific anti-CD19/anti-CD3 single-chain antibody construct. Leukemia 17, 900–909 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Hoffmann, P. et al. Serial killing of tumor cells by cytotoxic T cells redirected with a CD19-/CD3-bispecific single-chain antibody construct. Int. J. Cancer 115, 98–104 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Nagorsen, D., Kufer, P., Baeuerle, P. A. & Bargou, R. Blinatumomab: a historical perspective. Pharmacol. Ther. 136, 334–342 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Gross, G., Waks, T. & Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl Acad. Sci. USA 86, 10024–10028 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. June, C. H., O’Connor, R. S., Kawalekar, O. U., Ghassemi, S. & Milone, M. C. CAR T cell immunotherapy for human cancer. Science 359, 1361–1365 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Grupp, S. A. et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 368, 1509–1518 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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 

  39. Davenport, A. J. & Jenkins, M. R. Programming a serial killer: CAR T cells form non-classical immune synapses. Oncoscience 5, 69–70 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Offner, S., Hofmeister, R., Romaniuk, A., Kufer, P. & Baeuerle, P. A. Induction of regular cytolytic T cell synapses by bispecific single-chain antibody constructs on MHC class I-negative tumor cells. Mol. Immunol. 43, 763–771 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Brudno, J. N. et al. T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of poor-prognosis relapsed multiple myeloma. J. Clin. Oncol. 36, 2267–2280 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sadelain, M., Riviere, I. & Riddell, S. Therapeutic T cell engineering. Nature 545, 423–431 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Klinger, M. et al. Adhesion of T cells to endothelial cells facilitates blinatumomab-associated neurologic adverse events. Cancer Res. 80, 91–101 (2020).

    Article  CAS  PubMed  Google Scholar 

  44. Bargou, R. et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321, 974–977 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Schuster, S. J. et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N. Engl. J. Med. 377, 2545–2554 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Schuster, S. J. CD19-directed CAR T cells gain traction. Lancet Oncol. 20, 2–3 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. 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 

  48. Neelapu, S. S. et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 377, 2531–2544 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. D’Agostino, M. & Raje, N. Anti-BCMA CAR T-cell therapy in multiple myeloma: can we do better? Leukemia 34, 21–34 (2020).

    Article  PubMed  CAS  Google Scholar 

  50. Yan, Z. et al. A combination of humanised anti-CD19 and anti-BCMA CAR T cells in patients with relapsed or refractory multiple myeloma: a single-arm, phase 2 trial. Lancet Haematol. 6, e521–e529 (2019).

    Article  PubMed  Google Scholar 

  51. Fan, M. et al. Chimeric antigen receptors for adoptive T cell therapy in acute myeloid leukemia. J. Hematol. Oncol. 10, 151 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. 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 

  53. Schmidts, A. & Maus, M. V. Making CAR T cells a solid option for solid tumors. Front. Immunol. 9, 2593 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Metzinger, M. N. et al. Chimeric antigen receptor T-cell therapy: reach to solid tumor experience. Oncology 97, 59–74 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Rafiq, S., Hackett, C. S. & Brentjens, R. J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020).

    Article  PubMed  Google Scholar 

  56. Goebeler, M. E. et al. Bispecific T-cell engager (BiTE) antibody construct blinatumomab for the treatment of patients with relapsed/refractory non-Hodgkin lymphoma: final results from a phase I study. J. Clin. Oncol. 34, 1104–1111 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. Nagorsen, D. et al. Immunotherapy of lymphoma and leukemia with T-cell engaging BiTE antibody blinatumomab. Leuk. Lymphoma 50, 886–891 (2009).

    Article  CAS  PubMed  Google Scholar 

  58. Zhu, M. et al. Blinatumomab, a bispecific T-cell engager (BiTE®) for CD-19 targeted cancer immunotherapy: clinical pharmacology and its implications. Clin. Pharmacokinetics 55, 1271–1288 (2016).

    Article  CAS  Google Scholar 

  59. Viardot, A. et al. Phase 2 study of the bispecific T-cell engager (BiTE) antibody blinatumomab in relapsed/refractory diffuse large B-cell lymphoma. Blood 127, 1410–1416 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dufner, V. et al. Long-term outcome of patients with relapsed/refractory B-cell non-Hodgkin lymphoma treated with blinatumomab. Blood Adv. 3, 2491–2498 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Topp, M. S. et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J. Clin. Oncol. 29, 2493–2498 (2011).

    Article  CAS  PubMed  Google Scholar 

  62. Gokbuget, N. et al. Long-term relapse-free survival in a phase 2 study of blinatumomab for the treatment of patients with minimal residual disease in B-lineage acute lymphoblastic leukemia. Haematologica 102, e132–e135 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Gokbuget, N. et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 131, 1522–1531 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Topp, M. S. et al. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J. Clin. Oncol. 32, 4134–4140 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Topp, M. S. et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 16, 57–66 (2015).

    Article  CAS  PubMed  Google Scholar 

  66. Martinelli, G. et al. Complete hematologic and molecular response in adult patients with relapsed/refractory Philadelphia chromosome-positive B-precursor acute lymphoblastic leukemia following treatment with blinatumomab: results from a phase II, single-arm, multicenter study. J. Clin. Oncol. 35, 1795–1802 (2017).

    Article  CAS  PubMed  Google Scholar 

  67. von Stackelberg, A. et al. Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. J. Clin. Oncol. 34, 4381–4389 (2016).

    Article  Google Scholar 

  68. Jabbour, E. J. et al. Transplantation in adults with relapsed/refractory acute lymphoblastic leukemia who are treated with blinatumomab from a phase 3 study. Cancer 125, 4181–4192 (2019).

    Article  CAS  PubMed  Google Scholar 

  69. Kantarjian, H. et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N. Engl. J. Med. 376, 836–847 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Dombret, H. et al. Blinatumomab versus chemotherapy in first salvage or in later salvage for B-cell precursor acute lymphoblastic leukemia. Leuk. Lymphoma 60, 2214–2222 (2019).

    Article  CAS  PubMed  Google Scholar 

  71. Amgen. Amgen announces positive results from two phase 3 BLINCYTO® (blinatumomab) studies in pediatric patients with relapsed acute lymphoblastic leukemia. Amgen https://www.amgen.com/media/news-releases/2019/09/amgen-announces-positive-results-from-two-phase-3-blincyto-blinatumomab-studies-in-pediatric-patients-with-relapsed-acute-lymphoblastic-leukemia/ (2019).

  72. King, A. C., Pappacena, J. J., Tallman, M. S., Park, J. H. & Geyer, M. B. Blinatumomab administered concurrently with oral tyrosine kinase inhibitor therapy is a well-tolerated consolidation strategy and eradicates measurable residual disease in adults with Philadelphia chromosome positive acute lymphoblastic leukemia. Leukemia Res. 79, 27–33 (2019).

    Article  CAS  Google Scholar 

  73. Schwartz, M. S., Jeyakumar, D., Damon, L. E., Schiller, G. J. & Wieduwilt, M. J. A phase I/II study of blinatumomab in combination with pembrolizumab for adults with relapsed refractory B-lineage acute lymphoblastic leukemia: University of California Hematologic Malignancies Consortium Study 1504 [abstract]. J. Clin. Oncol. 37 (Suppl. 15), TPS7064 (2019).

    Article  Google Scholar 

  74. Raff, T. et al. Molecular relapse in adult standard-risk ALL patients detected by prospective MRD monitoring during and after maintenance treatment: data from the GMALL 06/99 and 07/03 trials. Blood 109, 910–915 (2007).

    Article  CAS  PubMed  Google Scholar 

  75. Bruggemann, M. et al. Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood 107, 1116–1123 (2006).

    Article  PubMed  CAS  Google Scholar 

  76. Bassan, R. et al. Improved risk classification for risk-specific therapy based on the molecular study of minimal residual disease (MRD) in adult acute lymphoblastic leukemia (ALL). Blood 113, 4153–4162 (2009).

    Article  CAS  PubMed  Google Scholar 

  77. Bader, P. et al. Prognostic value of minimal residual disease quantification before allogeneic stem-cell transplantation in relapsed childhood acute lymphoblastic leukemia: the ALL-REZ BFM Study Group. J. Clin. Oncol. 27, 377–384 (2009).

    Article  PubMed  Google Scholar 

  78. Gökbuget, N. et al. Minimal residual disease level predicts outcome in adults with Ph-negative B-precursor acute lymphoblastic leukemia. Hematology 24, 337–348 (2019).

    Article  PubMed  CAS  Google Scholar 

  79. Topp, M. S. et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood 120, 5185–5187 (2012).

    Article  CAS  PubMed  Google Scholar 

  80. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Chase, M. L. & Armand, P. Minimal residual disease in non-Hodgkin lymphoma–current applications and future directions. Br. J. Haematol. 180, 177–188 (2018).

    Article  PubMed  Google Scholar 

  82. Poulet, G., Massias, J. & Taly, V. Liquid biopsy: general concepts. Acta Cytologica 63, 449–455 (2019).

    Article  CAS  PubMed  Google Scholar 

  83. Fernandes Marques, J. et al. Circulating tumor DNA: a step into the future of cancer management. Acta Cytologica 63, 456–465 (2019).

    Article  CAS  PubMed  Google Scholar 

  84. Ravandi, F. et al. A phase 1 first-in-human study of AMG 330, an anti-CD33 bispecific T-cell engager (BiTE®) antibody construct, in relapsed/refractory acute myeloid leukemia (R/R AML) [abstract]. Blood 132 (Suppl. 1), 25 (2018).

    Article  Google Scholar 

  85. Topp, M. S. et al. Evaluation of AMG 420, an anti-BCMA bispecific T-cell engager (BiTE) immunotherapy, in R/R multiple myeloma (MM) patients: updated results of a first-in-human (FIH) phase I dose escalation study [abstract]. J. Clin. Oncol. 37 (Suppl. 15), 8007 (2019).

    Article  Google Scholar 

  86. Topp, M. S. et al. Anti-B-cell maturation antigen BiTE molecule AMG 420 induces responses in multiple myeloma. J. Clin. Oncol. 38, 775–783 (2020).

    Article  PubMed  Google Scholar 

  87. Hummel, H.-D. et al. Phase 1 study of pasotuxizumab (BAY 2010112), a PSMA-targeting bispecific T cell engager (BiTE) immunotherapy for metastatic castration-resistant prostate cancer (mCRPC) [abstract]. J. Clin. Oncol. 37 (Suppl. 15), 5034 (2019).

    Article  Google Scholar 

  88. Kebenko, M. et al. A multicenter phase 1 study of solitomab (MT110, AMG 110), a bispecific EpCAM/CD3 T-cell engager (BiTE(R)) antibody construct, in patients with refractory solid tumors. Oncoimmunology 7, e1450710 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Walker, A. J. et al. Tumor antigen and receptor densities regulate efficacy of a chimeric antigen receptor targeting anaplastic lymphoma kinase. Mol. Ther. 25, 2189–2201 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Aldoss, I. et al. Correlates of resistance and relapse during blinatumomab therapy for relapsed/refractory acute lymphoblastic leukemia. Am. J. Hematol. 92, 858–865 (2017).

    Article  CAS  PubMed  Google Scholar 

  91. Jiang, X. et al. Development of a target cell-biologics-effector cell (TBE) complex-based cell killing model to characterize target cell depletion by T cell redirecting bispecific agents. mAbs 10, 876–889 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Nerreter, T. et al. Super-resolution microscopy reveals ultra-low CD19 expression on myeloma cells that triggers elimination by CD19 CAR-T. Nat. Commun. 10, 3137 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Teachey, D. T. et al. Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood 121, 5154–5157 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Choudhry, J., Parson, M. & Wright, J. A retrospective review of tocilizumab for the management of blinatumomab (a bispecific T cell engager)-induced cytokine release syndrome (CRS) [abstract]. Blood 132 (Suppl. 1), 5211 (2018).

    Article  Google Scholar 

  95. Liu, D. & Zhao, J. Cytokine release syndrome: grading, modeling, and new therapy. J. Hematol. Oncol. 11, 121 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Maude, S. L., Barrett, D., Teachey, D. T. & Grupp, S. A. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 20, 119–122 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Gardner, R. A. et al. Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without attenuation of antileukemic efficacy. Blood 134, 2149–2158 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  98. Lee, D. W. et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol. Blood Marrow Transpl. 25, 625–638 (2019).

    Article  CAS  Google Scholar 

  99. Stein, A. S. et al. Neurologic adverse events in patients with relapsed/refractory acute lymphoblastic leukemia treated with blinatumomab: management and mitigating factors. Ann. Hematol. 98, 159–167 (2019).

    Article  CAS  PubMed  Google Scholar 

  100. Ribera, J. M., Ferrer, A., Ribera, J. & Genesca, E. Profile of blinatumomab and its potential in the treatment of relapsed/refractory acute lymphoblastic leukemia. Onco Targets Ther. 8, 1567–1574 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Arvedson, T. L. et al. Generation of half-life extended anti-CD33 BiTE® antibody constructs compatible with once-weekly dosing [abstract]. Cancer Res. 77 (Suppl. 13), 55 (2017).

    Google Scholar 

  102. Popplewell, L. et al. A first-in-human study of a half-life extended CD19-targeting bite in relapsed/refractory diffuse large B cell lymphoma, mantle cell lymphoma or follicular lymphoma. Hematol. Oncol. 37, 566–567 (2019).

    Article  Google Scholar 

  103. Kohnke, T., Krupka, C., Tischer, J., Knosel, T. & Subklewe, M. Increase of PD-L1 expressing B-precursor ALL cells in a patient resistant to the CD19/CD3-bispecific T cell engager antibody blinatumomab. J. Hematol. Oncol. 8, 111 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Krupka, C. et al. Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell-induced immune escape mechanism. Leukemia 30, 484–491 (2016).

    Article  CAS  PubMed  Google Scholar 

  105. Feucht, J. et al. T-cell responses against CD19+ pediatric acute lymphoblastic leukemia mediated by bispecific T-cell engager (BiTE) are regulated contrarily by PD-L1 and CD80/CD86 on leukemic blasts. Oncotarget 7, 76902–76919 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  106. 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 

  107. Tabernero, J. et al. Phase Ia and Ib studies of the novel carcinoembryonic antigen (CEA) T-cell bispecific (CEA CD3 TCB) antibody as a single agent and in combination with atezolizumab: preliminary efficacy and safety in patients with metastatic colorectal cancer (mCRC) [abstract]. J. Clin. Oncol. 35 (Suppl. 15), 3002 (2017).

    Article  Google Scholar 

  108. Topp, M. S. et al. Safety and preliminary antitumor activity of the anti-PD-1 monoclonal antibody REGN2810 alone or in combination with REGN1979, an anti-CD20 × anti-CD3 bispecific antibody, in patients with B-lymphoid malignancies [abstract]. Blood 130 (Suppl. 1), 1495 (2017).

    Google Scholar 

  109. Duell, J. et al. Frequency of regulatory T cells determines the outcome of the T-cell-engaging antibody blinatumomab in patients with B-precursor ALL. Leukemia 31, 2181–2190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Annacker, O. et al. CD25+CD4+ T cells regulate the expansion of peripheral CD4 T cells through the production of IL-10. J. Immunol. 166, 3008–3018 (2001).

    Article  CAS  PubMed  Google Scholar 

  111. Schmidt, A., Oberle, N. & Krammer, P. Molecular mechanisms of Treg-mediated T cell suppression. Front. Immunol. 3, 51 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Katz, J. B., Muller, A. J. & Prendergast, G. C. Indoleamine 2,3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunol. Rev. 222, 206–221 (2008).

    Article  CAS  PubMed  Google Scholar 

  113. Zappasodi, R., Merghoub, T. & Wolchok, J. D. Emerging concepts for immune checkpoint blockade-based combination therapies. Cancer Cell 33, 581–598 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Braig, F. et al. Resistance to anti-CD19/CD3 BiTE in acute lymphoblastic leukemia may be mediated by disrupted CD19 membrane trafficking. Blood 129, 100–104 (2017).

    Article  CAS  PubMed  Google Scholar 

  115. Brudno, J. N. & Kochenderfer, J. N. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev. 34, 45–55 (2019).

    Article  CAS  PubMed  Google Scholar 

  116. Jabbour, E. et al. Outcome of patients with relapsed/refractory acute lymphoblastic leukemia after blinatumomab failure: no change in the level of CD19 expression. Am. J. Hematol. 93, 371–374 (2018).

    Article  CAS  PubMed  Google Scholar 

  117. Ruella, M. & Maus, M. V. Catch me if you can: leukemia escape after CD19-directed T cell immunotherapies. Comput. Struct. Biotechnol. J. 14, 357–362 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ruella, M. et al. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J. Clin. Invest. 126, 3814–3826 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Horn, L. A. et al. CD3xPDL1 bi-specific T cell engager (BiTE) simultaneously activates T cells and NKT cells, kills PDL1(+) tumor cells, and extends the survival of tumor-bearing humanized mice. Oncotarget 8, 57964–57980 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Correnti, C. E. et al. Simultaneous multiple interaction T-cell engaging (SMITE) bispecific antibodies overcome bispecific T-cell engager (BiTE) resistance via CD28 co-stimulation. Leukemia 32, 1239–1243 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Vallera, D. A. et al. IL15 trispecific killer engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo expansion, and enhanced function. Clin. Cancer Res. 22, 3440–3450 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Kobold, S., Pantelyushin, S., Rataj, F. & Vom Berg, J. Rationale for combining bispecific T cell activating antibodies with checkpoint blockade for cancer therapy. Front. Oncol. 8, 285 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Memarnejadian, A. et al. PD-1 blockade promotes epitope spreading in anticancer CD8+T cell responses by preventing fratricidal death of subdominant clones to relieve immunodomination. J. Immunol. 199, 3348–3359 (2017).

    Article  CAS  PubMed  Google Scholar 

  124. Rosenthal, M. et al. Novel anti-EGFRvIII bispecific T cell engager (BiTE) antibody construct in glioblastoma (GBM): trial in progress of AMG 596 in patients with recurrent or newly diagnosed disease [abstract]. J. Clin. Oncol. 37 (Suppl. 15), TPS2071 (2019).

    Article  Google Scholar 

  125. Choi, B. D., O’Rourke, D. M. & Maus, M. V. Engineering chimeric antigen receptor T cells to treat glioblastoma. J. Target. Ther. Cancer 6, 22–25 (2017).

    PubMed  PubMed Central  Google Scholar 

  126. 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  CAS  Google Scholar 

  127. Choi, B. D. et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat. Biotechnol. 37, 1049–1058 (2019).

    Article  CAS  PubMed  Google Scholar 

  128. Wing, A. et al. Improving CART-cell therapy of solid tumors with oncolytic virus-driven production of a bispecific T-cell engager. Cancer Immunol. Res. 6, 605–616 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Speck, T. et al. Targeted BiTE expression by an oncolytic vector augments therapeutic efficacy against solid tumors. Clin. Cancer Res. 24, 2128–2137 (2018).

    Article  CAS  PubMed  Google Scholar 

  130. Scott, E. M., Duffy, M. R., Freedman, J. D., Fisher, K. D. & Seymour, L. W. Solid tumor immunotherapy with T cell engager-armed oncolytic viruses. Macromol. Biosci. 18, 1700187 (2018).

    Article  CAS  Google Scholar 

  131. Rosewell Shaw, A. & Suzuki, M. Oncolytic viruses partner with T-cell therapy for solid tumor treatment. Front. Immunol. 9, 2103 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Fajardo, C. A. et al. Oncolytic adenoviral delivery of an EGFR-targeting T-cell engager improves antitumor efficacy. Cancer Res. 77, 2052–2063 (2017).

    Article  CAS  PubMed  Google Scholar 

  133. Johnson, S. et al. Effector cell recruitment with novel Fv-based dual-affinity re-targeting protein leads to potent tumor cytolysis and in vivo B-cell depletion. J. Mol. Biol. 399, 436–449 (2010).

    Article  CAS  PubMed  Google Scholar 

  134. Moore, P. A. et al. Application of dual affinity retargeting molecules to achieve optimal redirected T-cell killing of B-cell lymphoma. Blood 117, 4542–4551 (2011).

    Article  CAS  PubMed  Google Scholar 

  135. Liu, L. et al. MGD011, a CD19 x CD3 dual-affinity retargeting bi-specific molecule incorporating extended circulating half-life for the treatment of B-cell malignancies. Clin. Cancer Res. 23, 1506–1518 (2017).

    Article  CAS  PubMed  Google Scholar 

  136. Moore, P. A. et al. Development of MGD007, a gpA33 x CD3-bispecific DART protein for T-cell immunotherapy of metastatic colorectal cancer. Mol. Cancer Ther. 17, 1761–1772 (2018).

    Article  CAS  PubMed  Google Scholar 

  137. Al-Hussaini, M. et al. Targeting CD123 in acute myeloid leukemia using a T-cell-directed dual-affinity retargeting platform. Blood 127, 122–131 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Klein, C. et al. Engineering therapeutic bispecific antibodies using CrossMab technology. Methods 154, 21–31 (2019).

    Article  CAS  PubMed  Google Scholar 

  139. Bacac, M., Klein, C. & Umana, P. CEA TCB: a novel head-to-tail 2:1 T cell bispecific antibody for treatment of CEA-positive solid tumors. Oncoimmunology 5, e1203498 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Bacac, M. et al. A novel carcinoembryonic antigen T-cell bispecific antibody (CEA TCB) for the treatment of solid tumors. Clin. Cancer Res. 22, 3286–3297 (2016).

    Article  CAS  PubMed  Google Scholar 

  141. American Society of Hematology. Bispecific antibody REGN1979 shows promising efficacy in relapsed/refractory non-Hodgkin lymphomas. ASH Clinical News https://www.ashclinicalnews.org/on-location/other-meetings/bispecific-antibody-regn1979-shows-promising-efficacy-relapsed-refractory-non-hodgkin-lymphomas/ (2019).

  142. Oates, J., Hassan, N. J. & Jakobsen, B. K. ImmTACs for targeted cancer therapy: why, what, how, and which. Mol. Immunol. 67, 67–74 (2015).

    Article  CAS  PubMed  Google Scholar 

  143. Liddy, N. et al. Monoclonal TCR-redirected tumor cell killing. Nat. Med. 18, 980–987 (2012).

    Article  CAS  PubMed  Google Scholar 

  144. Lowe, K. L. et al. Novel TCR-based biologics: mobilising T cells to warm ‘cold’ tumours. Cancer Treat. Rev. 77, 35–43 (2019).

    Article  CAS  PubMed  Google Scholar 

  145. Sato, T. et al. Redirected T cell lysis in patients with metastatic uveal melanoma with gp100-directed TCR IMCgp100: overall survival findings [abstract]. J. Clin. Oncol. 36 (Suppl. 15), 9521 (2018).

    Article  Google Scholar 

  146. Jain, T. & Litzow, M. R. No free rides: management of toxicities of novel immunotherapies in ALL, including financial. Hematology 2018, 25–34 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Delea, T. E. et al. Cost-effectiveness of blinatumomab versus salvage chemotherapy in relapsed or refractory Philadelphia-chromosome-negative B-precursor acute lymphoblastic leukemia from a US payer perspective. J. Med. Econ. 20, 911–922 (2017).

    Article  PubMed  Google Scholar 

  148. Delea, T. E. et al. Cost effectiveness of blinatumomab versus inotuzumab ozogamicin in adult patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia in the United States. Pharmacoeconomics 37, 1177–1193 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  149. Stein, A. S. et al. Exposure-adjusted adverse events comparing blinatumomab with chemotherapy in advanced acute lymphoblastic leukemia. Blood Adv. 2, 1522–1531 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Slaney, C. Y., Wang, P., Darcy, P. K. & Kershaw, M. H. CARs versus BiTEs: a comparison between T cell-redirection strategies for cancer treatment. Cancer Discov. 8, 924–934 (2018).

    Article  CAS  PubMed  Google Scholar 

  151. Ajina, A. & Maher, J. Strategies to address chimeric antigen receptor tonic signaling. Mol. Cancer Ther. 17, 1795–1815 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Davenport, A. J. et al. CAR-T cells inflict sequential killing of multiple tumor target cells. Cancer Immunol. Res. 3, 483–494 (2015).

    Article  CAS  PubMed  Google Scholar 

  153. Darcy, P. K. et al. Redirected perforin-dependent lysis of colon carcinoma by ex vivo genetically engineered CTL. J. Immunol. 164, 3705–3712 (2000).

    Article  CAS  PubMed  Google Scholar 

  154. Yu, J., Wang, W. & Huang, H. Efficacy and safety of bispecific T-cell engager (BiTE) antibody blinatumomab for the treatment of relapsed/refractory acute lymphoblastic leukemia and non-Hodgkin’s lymphoma: a systemic review and meta-analysis. Hematology 24, 199–207 (2019).

    Article  CAS  PubMed  Google Scholar 

  155. Velasquez, M. P., Bonifant, C. L. & Gottschalk, S. Redirecting T cells to hematological malignancies with bispecific antibodies. Blood 131, 30–38 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. 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 

Download references

Acknowledgements

The authors particularly thank A. Wenzl for graphic design support and M. Goebeler for his thorough reading and editing of the manuscript.

Reviewer information

Nature Reviews Clinical Oncology thanks D. Vallera, E. Jabbour and the other, anonymous, reviewer for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

Both authors made a substantial contribution to all aspects of the preparation of this manuscript.

Corresponding author

Correspondence to Ralf C. Bargou.

Ethics declarations

Competing interests

M.-E.G. has consulted for and received honoraria from Gemoab. R.C.B. has consulted for and received honoraria from Amgen, Cellex and Gemoab. R.C.B. is patent holder for blinatumomab, from which he receives royalty payments. M.-E.G and R.C.B. have both received honoraria for serving on the advisory boards of Amgen, Bristol-Myers Squibb, Gemoab, Novartis and Roche.

Additional information

Publisher’s note

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

Related links

Blinatumomab prescribing information: https://www.drugs.com/cdi/blinatumomab.html

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Goebeler, ME., Bargou, R.C. T cell-engaging therapies — BiTEs and beyond. Nat Rev Clin Oncol 17, 418–434 (2020). https://doi.org/10.1038/s41571-020-0347-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-020-0347-5

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