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:

Small-molecule-mediated control of the anti-tumour activity and off-tumour toxicity of a supramolecular bispecific T cell engager

An Author Correction to this article was published on 15 March 2024

This article has been updated

Abstract

The broader clinical use of bispecific T cell engagers for inducing anti-tumour toxicity is hindered by their on-target off-tumour toxicity and the associated neurotoxicity and cytokine-release syndrome. Here we show that the off-tumour toxicity of a supramolecular bispecific T cell engager binding to the T cell co-receptor CD3 and to the human epidermal growth factor receptor 2 on breast tumour cells can be halted by disengaging the T cells from the tumour cells via the infusion of the small-molecule drug amantadine, which disassembles the supramolecular aggregate. In mice bearing human epidermal growth factor receptor 2-expressing tumours and with a human immune system, high intravenous doses of such a ‘switchable T cell nanoengager’ elicited strong tumour-specific adaptive immune responses that prevented tumour relapse, while the infusion of amantadine restricted off-tumour toxicity, cytokine-release syndrome and neurotoxicity. Supramolecular chemistry may be further leveraged to control the anti-tumour activity and off-tumour toxicity of bispecific antibodies.

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: SiTE for cancer immunotherapy.
Fig. 2: Synthesis and characterization of SiTE.
Fig. 3: Small-molecule AMD controls SiTE activity in vitro.
Fig. 4: AMD reduces the on-target off-tumour toxicity of SiTE in vivo.
Fig. 5: High doses of SiTE induce a strong tumour-specific T cell immune response.
Fig. 6: AMD reduces SiTE-induced CRS in vivo.

Similar content being viewed by others

Data availability

The data supporting the results in this study are available within the paper and its Supplementary Information. The raw and analysed datasets generated during the study are available for research purposes from the corresponding authors on reasonable request. Source data are provided with this paper.

Change history

References

  1. Huehls, A. M., Coupet, T. A. & Sentman, C. L. Bispecific T‐cell engagers for cancer immunotherapy. Immunol. Cell Biol. 93, 290–296 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Rader, C. Bispecific antibodies in cancer immunotherapy. Curr. Opin. Biotechnol. 65, 9–16 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Weidanz, J. Targeting cancer with bispecific antibodies. Science 371, 996–997 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  4. de Miguel, M., Umana, P., de Morais, A. L. G., Moreno, V. & Calvo, E. T-cell-engaging therapy for solid tumors. Clin. Cancer Res. 27, 1595–1603 (2021).

    Article  PubMed  Google Scholar 

  5. Tian, Z., Liu, M., Zhang, Y. & Wang, X. Bispecific T cell engagers: an emerging therapy for management of hematologic malignancies. J. Hematol. Oncol. 14, 1–18 (2021).

    Article  Google Scholar 

  6. Smits, N. C. & Sentman, C. L. Bispecific T-cell engagers (BiTEs) as treatment of B-cell lymphoma. J. Clin. Oncol. 34, 1131 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Krishnamurthy, A. & Jimeno, A. Bispecific antibodies for cancer therapy: a review. Pharmacol. Ther. 185, 122–134 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Duell, J. et al. Bispecific antibodies in the treatment of hematologic malignancies. Clin. Pharmacol. Ther. 106, 781–791 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Przepiorka, D. et al. FDA approval: blinatumomab. Clin. Cancer Res. 21, 4035–4039 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. Labrijn, A. F., Janmaat, M. L., Reichert, J. M. & Parren, P. W. Bispecific antibodies: a mechanistic review of the pipeline. Nat. Rev. Drug Discov. 18, 585–608 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. 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 

  13. 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 

  14. Fadul, C. et al. A phase I study targeting newly diagnosed glioblastoma with anti-CD3× anti-EGFR bispecific antibody armed T cells (EGFR BATs) in combination with radiation and temozolomide. Brain Tumor Res. Treat. 10, S193 (2022).

    Google Scholar 

  15. Fiedler, W. et al. Phase I safety and pharmacology study of the EpCAM/CD3-bispecific BiTE antibody MT110 in patients with metastatic colorectal, gastric, or lung cancer. J. Clin. Oncol. 28, 2573–2573 (2010).

    Article  Google Scholar 

  16. Hutchings, M. et al. Glofitamab, a novel, bivalent CD20-targeting T-cell–engaging bispecific antibody, induces durable complete remissions in relapsed or refractory B-cell lymphoma: a phase I trial. J. Clin. Oncol. 39, 1959–1970 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Heitmann, J. S. et al. Protocol of a prospective, multicentre phase I study to evaluate the safety, tolerability and preliminary efficacy of the bispecific PSMAxCD3 antibody CC-1 in patients with castration-resistant prostate carcinoma. BMJ Open 10, e039639 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Lum, L. G. et al. Clinical and immune responses to anti-CD3 x anti-EGFR bispecific antibody armed activated T cells (EGFR BATs) in pancreatic cancer patients. Oncoimmunology 9, 1773201 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Lum, L. G. et al. Phase II clinical trial using anti-CD3× anti-HER2 bispecific antibody armed activated T cells (HER2 BATs) consolidation therapy for HER2 negative (0–2+) metastatic breast cancer. J. Immunother. Cancer 9, e002194 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Van De Vyver, A. J., Marrer-Berger, E., Wang, K., Lehr, T. & Walz, A.-C. Cytokine release syndrome by T-cell-redirecting therapies: can we predict and modulate patient risk? Clin. Cancer Res. https://doi.org/10.1158/1078-0432.CCR-21-0470 (2021).

  21. Frey, N. V. & Porter, D. L. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Hematol. Am. Soc. Hematol. Educ. Program 2016, 567–572 (2016).

    Article  Google Scholar 

  22. 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 

  23. 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. e117 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Goebeler, M.-E. & Bargou, R. C. T cell-engaging therapies—BiTEs and beyond. Nat. Rev. Clin. Oncol. 17, 418–434 (2020).

    Article  PubMed  Google Scholar 

  25. Gross, G. & Eshhar, Z. Therapeutic potential of T cell chimeric antigen receptors (CARs) in cancer treatment: counteracting off-tumor toxicities for safe CAR T cell therapy. Annu. Rev. Pharmacol. Toxicol. 56, 59–83 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Kallioniemi, O.-P. et al. ERBB2 amplification in breast cancer analyzed by fluorescence in situ hybridization. Proc. Natl Acad. Sci. USA 89, 5321–5325 (1992).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Morgan, R. A. et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843–851 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Morris, E. C., Neelapu, S. S., Giavridis, T. & Sadelain, M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat. Rev. Immunol. 22, 85–96 (2022).

    Article  CAS  PubMed  Google Scholar 

  29. Suurs, F. V., Lub-de Hooge, M. N., de Vries, E. G. & de Groot, D. J. A. A review of bispecific antibodies and antibody constructs in oncology and clinical challenges. Pharmacol. Ther. 201, 103–119 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Bai, Y., Luo, Q. & Liu, J. Protein self-assembly via supramolecular strategies. Chem. Soc. Rev. 45, 2756–2767 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Sakamoto, S. & Kudo, K. Supramolecular control of split-GFP reassembly by conjugation of β-cyclodextrin and coumarin units. J. Am. Chem. Soc. 130, 9574–9582 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Friberg, G. & Reese, D. Blinatumomab (Blincyto): lessons learned from the bispecific T-cell engager (BiTE) in acute lymphocytic leukemia (ALL). Ann. Oncol. 28, 2009–2012 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Hubsher, G., Haider, M. & Okun, M. Amantadine: the journey from fighting flu to treating Parkinson disease. Neurology 78, 1096–1099 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Schmid, D. et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 8, 1–12 (2017).

    Article  CAS  Google Scholar 

  35. McCall, M. J., Diril, H. & Meares, C. F. Simplified method for conjugating macrocyclic bifunctional chelating agents to antibodies via 2-iminothiolane. Bioconjug. Chem. 1, 222–226 (1990).

    Article  CAS  PubMed  Google Scholar 

  36. Ding, Y.-F. et al. Host–guest interactions initiated supramolecular chitosan nanogels for selective intracellular drug delivery. ACS Appl. Mater. Interfaces 11, 28665–28670 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Estrada, E., Perdomo-López, I. & Torres-Labandeira, J. J. Combination of 2D-, 3D-connectivity and quantum chemical descriptors in QSPR. Complexation of α-and β-cyclodextrin with benzene derivatives. J. Chem. Inf. Comput. Sci. 41, 1561–1568 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. Liu, L. & Guo, Q.-X. Novel prediction for the driving force and guest orientation in the complexation of α-and β-cyclodextrin with benzene derivatives. J. Phys. Chem. B 103, 3461–3467 (1999).

    Article  CAS  Google Scholar 

  39. Jiang, W., Kim, B., Rutka, J. T. & Chan, W. C. Nanoparticle-mediated cellular response is size-dependent. Nat. Nanotechnol. 3, 145–150 (2008).

    Article  ADS  CAS  PubMed  Google Scholar 

  40. Yuan, H. et al. Multivalent bi-specific nanobioconjugate engager for targeted cancer immunotherapy. Nat. Nanotechnol. 12, 763–769 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Selvin, P. R. The renaissance of fluorescence resonance energy transfer. Nat. Struct. Biol. 7, 730–734 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Zhang, B. et al. Site-specific PEGylation of interleukin-2 enhances immunosuppression via the sustained activation of regulatory T cells. Nat. Biomed. Eng. 5, 1288–1305 (2021).

  43. Harris, J. M. & Chess, R. B. Effect of pegylation on pharmaceuticals. Nat. Rev. Drug Discov. 2, 214–221 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. 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 

  45. Aoki, F. Y. & Sitar, D. S. Clinical pharmacokinetics of amantadine hydrochloride. Clin. Pharmacokinet. 14, 35–51 (1988).

    Article  CAS  PubMed  Google Scholar 

  46. Kim, C., Agasti, S. S., Zhu, Z., Isaacs, L. & Rotello, V. M. Recognition-mediated activation of therapeutic gold nanoparticles inside living cells. Nat. Chem. 2, 962–966 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tonga, G. Y. et al. Supramolecular regulation of bioorthogonal catalysis in cells using nanoparticle-embedded transition metal catalysts. Nat. Chem. 7, 597–603 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tokumura, T. et al. Enhancement of bioavailability of cinnarizine from its β‐cyclodextrin complex on oral administration with dl‐phenylalanine as a competing agent. J. Pharm. Sci. 75, 391–394 (1986).

    Article  CAS  PubMed  Google Scholar 

  49. Braegelman, A. S. & Webber, M. J. Integrating stimuli-responsive properties in host–guest supramolecular drug delivery systems. Theranostics 9, 3017 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Castellarin, M. et al. A rational mouse model to detect on-target, off-tumor CAR T cell toxicity. JCI Insight 5, e136012 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Kim, M. J. & Ahituv, N. in Pharmacogenomics (Innocenti, F. & van Schaik, R. H. N. eds) 279–289 (Springer, 2013).

  52. Arteta, M. Y. et al. Successful reprogramming of cellular protein production through mRNA delivered by functionalized lipid nanoparticles. Proc. Natl Acad. Sci. USA 115, E3351–E3360 (2018).

    CAS  Google Scholar 

  53. Zhang, Y., Sun, C., Wang, C., Jankovic, K. E. & Dong, Y. Lipids and lipid derivatives for RNA delivery. Chem. Rev. https://doi.org/10.1021/acs.chemrev.1c00244 (2021).

  54. Min, Y. et al. Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy. Nat. Nanotechnol. 12, 877–882 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhou, Q. et al. Mannose-derived carbon dots amplify microwave ablation-induced antitumor immune responses by capturing and transferring ‘danger signals’ to dendritic cells. ACS Nano 15, 2920–2932 (2021).

    Article  CAS  PubMed  Google Scholar 

  56. Norelli, M. et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat. Med. 24, 739–748 (2018).

    Article  CAS  PubMed  Google Scholar 

  57. Chen, F. et al. Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CAR T cell therapy. J. Immunol. Methods 434, 1–8 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Melero, I., Castanon, E., Alvarez, M., Champiat, S. & Marabelle, A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat. Rev. Clin. Oncol. 18, 558–576 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Di, S. et al. Combined adjuvant of poly I: C improves antitumor effects of CAR-T cells. Front. Oncol. 9, 241 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

M.J.M. acknowledges support from a United States National Institutes of Health Director’s New Innovator Award (DP2 TR002776), a Burroughs Wellcome Fund Career Award at the Scientific Interface, an NSF CAREER Award (CBET-2145491) and the American Cancer Society (RSG-22-122-01-ET).

Author information

Authors and Affiliations

Authors

Contributions

N.G. and M.J.M. conceived and designed the experiments. N.G., L.X., J.Q. and X.H. performed the experiments. N.G., L.X., J.Q., X.H. and R.E. analysed the data. N.G., M.J.M. and M.M.B. wrote and edited the manuscript. N.C.S. and C.H.J. provided materials and were involved in discussions of the work. M.J.M. supervised the project. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Michael J. Mitchell.

Ethics declarations

Competing interests

N.G. and M.J.M. have filed a patent application related to this study. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Archana Thakur and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 AMD mediates the in vivo disassembly of SiTE.

SiTE was labelled with Cy7 dye (Cy7 was labelled on CD3 Fab) and was i.v. injected to tumour-bearing mice at 0h. Then, PBS, AMD dispersed in PBS or AMD dispersed in 5% polyoxyethylene castor oil was injected at 18 h post-SiTE-Cy7 injection. 6 h later, mice were euthanized and the Cy7 signal in different organs and tumour was measured using IVIS (a-c). d, quantification of the fluorescence in tumour tissues in a-c. Data was shown as mean ± SD (n = 3), statistical differences were analysed using two-tailed unpaired Student’s t-test. e, Half-life of AMD in mouse blood when the AMD is dispersed in PBS or in 5% polyoxyethylene castor oil. When the AMD is dispersed in PBS, the half-life is about 9 hours, which is increased to 24 h when it is dispersed in 5% polyoxyethylene castor oil. f, AMD concentrations in the liver and tumour over time were investigated. g, the distribution of AMD in mouse tumours and major organs after 24h of AMD infusion. h, In order to investigate the clearance of the disassembled SiTE and the AMD, we performed another animal experiment. SiTE-Cy7 was i.v. infused into tumour bearing mice at 0 h. 4 h later, AMD was infused. After 24 h, mice were euthanized and the Cy7 signal distribution in major organs and mouse whole body was observed. Cy7 signal was detected in the spleen, kidneys and bladder. This demonstrates the potential clearance from the kidneys and urine. We then performed a kinetics study to investigate the Cy7 signal (i) and AMD level in the urine (j).

Source data

Extended Data Fig. 2 AMD controls SiTE activity in vivo.

a, 106 E0771-HER2 cells were s.c. injected into the right flank of C57/BL6 mice. When tumour sizes reached 50 mm3 (Day 7), mice were given an i.v. injection of 100 μL of their respective treatments—PBS, SiTE, SiTE + 100 μg AMD, SiTE + 5 μg AMD, or 100 μg AMD (in 5% polyoxyethylene castor oil). One group received SiTE on day 7 followed by 100 μg AMD at day 13. All mice were euthanized at day 23. b, Images of mice from all treatment groups on day 23. c, tumour growth curves for different groups. Tumour tissue was isolated on day 23 and observed for immune cell infiltration. d, repeat of the tumour growth inhibition experiment. e, mouse body weight change during the tumour inhibition experiment. f, Percentages of CD45+, CD45+CD3+, CD45+CD3+CD4+/CD8+ cells in the tumour tissues. g, Immunofluorescence images of the tumour tissues in different groups with cell nuclei labeled with DAPI (blue) and T cells labeled with anti-CD3 antibody (green). Scale bar: 100 μm. The statistical significance of tumour volume in c and e was analysed by two-tailed unpaired Student’s t-test. **P = 0.0013 ****P<0.0001. The statistical significance displayed in f was analysed by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 3 T-cell infiltration levels in the tumour tissues before and after SiTE or AMD treatment.

The mice were s.c injected with 106 E0771-HER2 cells at day 0, SiTE was injected at day 7, 9, and day 11 and AMD was injected at day 13 a, The tumour tissues at day 6, day 12 and day 14 were collected, digested, and filtered, and the immune cell infiltration in the tumour tissue was analysed using flow (flow gating strategy is shown in b). c, d, and e are flow dot plots of CD45+, CD45+CD3+, CD45+CD3+CD4+ or CD45+CD3+CD8+ cells, respectively. f-i are quantifications of c-e, respectively. The statistical significance of tumour volume in f-i was analysed by two-tailed unpaired Student’s t-test. f,***P = 0.0007, **P = 0.0027. g, ***P = 0.0005, *P = 0.0324. h, ***P = 0.0006, *P = 0.0351. i, ***P = 0.0005, *P = 0.0089.

Source data

Extended Data Fig. 4 High dose of SiTE elicited antigen-specific immune response to tumours.

C57BL/6 mice with liver expression of HER2 were injected with PBS, SiTE low dose, SiTE high dose, or AMD at days 7, 9 and 11. AMD was administered to SiTE-treated groups once severe toxicity was observed. Mice were euthanized at day 17, and the tumour tissues were collected and analysed by flow cytometry. a, mouse weight during the treatment. b, flow gating strategy. c, CD44+CD62L+ central memory cells in the tumour tissue.

Source data

Extended Data Fig. 5 High doses of SiTE enhance DC maturation and antibody production in vivo.

a-c, E0771-HER2 cells were s.c. injected to mice at day 0. E0771-HER2 tumour-bearing mice were treated with low doses (1 mg/kg) or high doses (5 mg/kg) of SiTE at days 7, 9, and 11. AMD was infused at day 13. Mice were euthanized at day 13 and the dendritic cell maturation levels in the tumour draining lymph nodes were evaluated. PBS or AMD-only were used as two control groups. d-f, In order to investigate the humoral immune response induced by SiTE, E0771-HER2 tumour-bearing mice were treated with low doses (1 mg/kg) or high doses (5 mg/kg) of SiTE at days 7, 9, and 11. AMD was infused at day 13. At day 27, the HER2-specific total IgG, IgG2c, and IgG1 levels in mouse blood were determined. P values were indicated in a-c, analysed by two-tailed unpaired Student’s t-test. n = 3.

Source data

Extended Data Fig. 6 Tumour-cell-rechallenging experiment.

Tumour-free mice from the high dose group were rechallenged with E0771-HER2 or E0771 cells and compared to mice pre-treated with either PBS or AMD challenged with E0771-HER2 as controls. Group R1: Healthy mice were treated with PBS at days −43, −41, and −39. The mice were i.v. injected with E0771-HER2 cells at day 0; Group R2: E0771-HER2 tumour-bearing mice were treated with a high dose of SiTE at days −43, −41, and −39, AMD was i.v. injected at day −37. The tumour-free mice were rechallenged with E0771-HER2 cells at day 0; Group R3, E0771-HER2 tumour-bearing mice were pre-treated with high dose of SiTE at days −43, −41, and −39. AMD was i.v. injected at day −37. The mice were rechallenged with E0771-HER2 cells at day 0. Group R4, healthy mice were treated with AMD at day −37 and the mice were i.v. injectied with E0771-HER2 cells at day 0. b-e are individual E0771-HER2 or E0771 tumour size curves in the different treatment groups. CR, complete regression (n = 8). The mice were euthanized, and the immune cell infiltration in the tumour tissue was analysed using flow. f-h, Treg percentages, CD44+CD62L+ central memory T cell percentages, and CD44+CD62L effector memory T cells in different treatment groups, respectively. Data were shown as mean ± SD, n = 4.

Source data

Extended Data Fig. 7 High doses of SiTE generate tumour-specific T-cell immune responses.

E0771-HER2 tumour-bearing mice were treated with high doses of SiTE. 7 days-post the last dose, T cells were collected from the mice and were co-cultured with E0771-HER2 cells (express luciferase) for 24h. E0771-HER2 cell viability was determined (a). b and c, E0771-HER2 target cells (labelled with low level of CSFE) and reference cells (B16 cell line, labelled with high level of CSFE) were i.v. infused into the high dose SiTE-treated mice mentioned above. 24 later, target cell killing was determined using flow cytometry (b). c, quantification of b. Data in a and c are shown as mean ± SD, n = 4. P values in a and c was determined using two-tailed unpaired student’s t-test.

Source data

Extended Data Fig. 8 Proteomics shows that a high dose of SiTE treatment induces damage-associated molecular patterns (DAMPs) and tumour-antigen release.

E0771-HER2 cells were incubated with mouse T cells in serum-free medium and were treated with a low dose (5 ng/mL) or a high dose (20 ng/mL) of SiTE. After 24h, the supernatant was collected and the proteins in the medium were analysed. a, relative abundance of various DAMPs released to cell culture medium in the high dose SiTE, low dose SiTE or PBS-treated group. b, and c, tumour antigen HER2 and tumour neoantigen hmmr and srrm1 abundance in the cell culture medium in different groups.

Source data

Extended Data Fig. 9 AMD reduces the on-target off-tumour toxicity of SiTE in vivo.

a, A humanized immune system mouse model was constructed by treating the mice with an i.p. injection of Busulfan at day −50 followed by an i.v. injection of 105 human CD34+ foetal liver cells at day −49. Human CD19 antigen was expressed in the livers of the mice using a piggyBac transposon system delivered via lipid nanoparticles (LNPs) encapsulating two plasmids, pCMV-hyPBase and pPB CMV-hCD19:T2A:EGFP, at day −35. At day −14, 106 Raji-Luc-GFP tumour cells were i.v. injected. Before using Blinatumomab or SiTE for cancer treatment, Tocilizumab (10 mg/kg) was administered to mice to prevent CRS-related symptoms. PBS, Blinatumomab in vivo bio-similar antibody (5 mg/kg), or SiTE (5 mg/kg) were i.v. injected every two days for three total doses. When an approximate 15% decrease in body weight (indicative of toxicity) was observed on day 7, half of the mice that had received SiTE were given an i.v. injection of AMD (in 5% polyoxyethylene castor oil). Throughout the study, mice were euthanized when body weight decreased more than 20%. IVIS was used to monitor tumour burden in vivo. b, IVIS images of the mice at days 0, 7, 10, and 30. c, and d, Mouse body weight and survival curves, respectively. n = 10 mice. The red stars in b and c indicate that mice were euthanized. e-h, Measurements for markers of toxicity and inflammation including AST, ALT, TNF-α, IFN-γ levels in mouse blood at different time points. n = 10 mice. i, In order to evaluate liver toxicity induced by various treatments, an additional animal experiment was performed and mouse livers were harvested at day 9. H&E staining was conducted to detect liver damage in different groups. Scale bar: 100 μm. j, Immunofluorescence imaging of CD3+ T cells in liver tissue. Blue: DAPI; Red, CD3+ T cells. k, tumour-free mice from the SiTE+AMD group were rechallenged with 106 Raji-Luc-GFP cells and the tumour burden post Raji-Luc-GFP cell rechallenging was monitored with IVIS. l-m, T cells in the tumour-free mice from the SiTE+AMD group were sorted and were co-cultured with Raji-Luc-GFP cells for 24 h and tumour cell viability was determined using a luciferase assay kit (l). T cells from normal humanized mice co-cultured with Raji-Luc-GFP cells were used as a control group. m, IFN-γ concentrations in the cell culture medium were determined using an ELISA kit. The data in e-h were plotted as mean ± s.d. (n = 10) from three independent experiments. P values indicated in e-h were analysed by two-tailed unpaired Student’s t-test. Pink, Blinatumomab vs SiTE + AMD at day 9; blue, SiTE vs SiTE + AMD at day 9; ***P = 0.0003, ****P<0.0001. P value in d was determined using Log-rank (Mantel-Cox) test, ****P<0.0001. P values in l and m were determined by two-tailed unpaired Student’s t-test, ****P<0.0001, ***P = 0.0006.

Source data

Extended Data Fig. 10 AMD reduces SiTE-induced neurotoxicity in vivo.

a, NSG-SGM3 mice were treated with Busulfan at day −50 to remove mouse bone marrow. At day −49, 105 human CD34+ foetal liver cells were i.v. injected to allow the mouse develop a human immune system. 106 Raji-Luc-GFP tumour cells were i.v. injected at day −14. At days 1, 3, and 5, PBS or SiTE was i.v. injected (in the PBS group, only PBS was injected into tumour-bearing mice). When an approximate 15% decrease in body weight (indicative of toxicity) was observed on day 7, half of the mice that had received SiTE were given an i.v. injection of AMD (10 mg/kg) and half of them were given Tocilizumab (10 mg/kg) for CRS treatment. b, c, d, and e, Mouse body weight, temperature, IL-6 levels, and mouse survival curves, respectively. n = 10 mice. The red stars in b indicate that mice were euthanized. f, Measurements of IL-1 in mouse blood at different time points. n = 10 mice. At around day 33 post-SiTE injection, humanized NSG-SGM3 mice that received either PBS or Tocilizumab treatment developed paralysis (g) or experienced a seizure as indicated by movement along the red arrows (h), which are signs of lethal neurological syndrome. However, mice treated with AMD did not develop paralysis and were not observed to experience seizures indicative of lethal neurological syndrome. Brain H&E staining (i) and human CD68 immunohistochemistry (j) images of mice at day 35. The data in b-f were plotted as mean ± s.d. (n = 10) from three independent experiments. P value in f was determined using Log-rank (Mantel-Cox) test, ****P < 0.0001.

Source data

Supplementary information

Source data

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

Gong, N., Han, X., Xue, L. et al. Small-molecule-mediated control of the anti-tumour activity and off-tumour toxicity of a supramolecular bispecific T cell engager. Nat. Biomed. Eng (2024). https://doi.org/10.1038/s41551-023-01147-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-023-01147-6

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