Skip to main content

Advertisement

Log in

Evaluating the Patient with Neurotoxicity after Chimeric Antigen Receptor T-cell Therapy

  • Neuro-oncology (GJ Lesser, Section Editor)
  • Published:
Current Treatment Options in Oncology Aims and scope Submit manuscript

Opinion statement

Chimeric antigen receptor (CAR) T-cells are now a well-established treatment for hematologic malignancies. Their use in clinical practice has expanded quite rapidly and hospitals have developed CAR T-cell protocols to evaluate patients for associated toxicities, and particularly for neurotoxicity. There are many variables that influence the risk for developing this complication, many of which are not fully understood. The severity can be related to a particular product. Clinical vigilance is critical to facilitate early recognition of neurotoxicity, hence the importance of pre-CAR T-cell neurological evaluation of each patient. While details of such an evaluation may slightly differ between institutions, generally a comprehensive neurological evaluation including assessment of cognitive abilities along with magnetic resonance imaging (MRI) of the brain is a gold standard. Management of neurotoxicity requires a well-orchestrated team approach with specialists from oncology, neurology, oftentimes neurosurgery and neuro-intensive care. Diagnostic work-up frequently includes detailed neurologic evaluation with comparison to the baseline assessment, imaging of the brain, electroencephalogram, and lumbar puncture. While steroids are uniformly used for treatment, many patients also receive tocilizumab for an underlying and frequently concomitant cytokine release syndrome (CRS) in addition to symptom-driven supportive care. Novel CAR T-cell constructs and other agents allowing for potentially lower risk of toxicity are being explored. While neurotoxicity is predominantly an early, and reversible, event, a growing body of literature suggests that late neurotoxicity with variable clinical presentation can also occur.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. • Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol. 2020;17(3):147–67 This review comprehensively discusses chimeric antigen receptor design and engineering strategies being developed to overcome current challenges.

  2. Denlinger N, Bond D, Jaglowski S. CAR T-cell therapy for B-cell lymphoma. Curr Probl Cancer. 2022;46(1):100826.

    Article  Google Scholar 

  3. Benmebarek MR, et al. Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int J Mol Sci. 2019;20(6):1283.

    Article  CAS  Google Scholar 

  4. Maude SL, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–48.

    Article  CAS  Google Scholar 

  5. Schuster SJ, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380(1):45–56.

    Article  CAS  Google Scholar 

  6. Neelapu SS, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531–44.

    Article  CAS  Google Scholar 

  7. Abramson JS, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839–52.

    Article  Google Scholar 

  8. Wang M, et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2020;382(14):1331–42.

    Article  CAS  Google Scholar 

  9. Munshi NC, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med. 2021;384(8):705–16.

    Article  CAS  Google Scholar 

  10. Berdeja JG, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet. 2021;398(10297):314–24.

    Article  CAS  Google Scholar 

  11. Martin T, et al. Ciltacabtagene autoleucel, an anti-B-cell maturation antigen chimeric antigen receptor T-cell therapy, for relapsed/refractory multiple myeloma: CARTITUDE-1 2-year follow-up. J Clin Oncol. 2022. https://doi.org/10.1200/JCO.22.00842.

  12. Fischer JW, Bhattarai N. CAR-T cell therapy: mechanism, management, and mitigation of inflammatory toxicities. Front Immunol. 2021;12:693016.

    Article  CAS  Google Scholar 

  13. van der Stegen SJ, Hamieh M, Sadelain M. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov. 2015;14(7):499–509.

    Article  Google Scholar 

  14. Neelapu SS, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47–62.

    Article  CAS  Google Scholar 

  15. Park JH, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):449–59.

    Article  CAS  Google Scholar 

  16. Schuster SJ, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017;377(26):2545–54.

    Article  CAS  Google Scholar 

  17. Weinkove R, et al. Selecting costimulatory domains for chimeric antigen receptors: functional and clinical considerations. Clin Transl Immunology. 2019;8(5):e1049.

    Article  Google Scholar 

  18. Grigor EJM, et al. Risks and benefits of chimeric antigen receptor T-cell (CAR-T) therapy in cancer: a systematic review and meta-analysis. Transfus Med Rev. 2019;33(2):98–110.

    Article  Google Scholar 

  19. Morris EC, et al. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol. 2022;22(2):85–96.

    Article  CAS  Google Scholar 

  20. Hay KA, et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017;130(21):2295–306.

    Article  CAS  Google Scholar 

  21. Santomasso BD, et al. Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov. 2018;8(8):958–71.

    Article  CAS  Google Scholar 

  22. Sterner RM, et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood. 2019;133(7):697–709.

    Article  CAS  Google Scholar 

  23. Kang L, et al. Interleukin-6-knockdown of chimeric antigen receptor-modified T cells significantly reduces IL-6 release from monocytes. Exp Hematol Oncol. 2020;9:11.

    Article  CAS  Google Scholar 

  24. Gust J, et al. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017;7(12):1404–19.

    Article  CAS  Google Scholar 

  25. Torre M, et al. Neuropathology of a case with fatal CAR T-cell-associated cerebral edema. J Neuropathol Exp Neurol. 2018;77(10):877–82.

    Article  CAS  Google Scholar 

  26. Turtle CJ, et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of ibrutinib. J Clin Oncol. 2017;35(26):3010–20.

    Article  CAS  Google Scholar 

  27. Grant SJ, et al. Clinical presentation, risk factors, and outcomes of immune effector cell-associated neurotoxicity syndrome following chimeric antigen receptor T cell therapy: a systematic review. Transplant Cell Ther. 2022;28(6):294–302.

    Article  CAS  Google Scholar 

  28. Karschnia P, et al. Clinical presentation, management, and biomarkers of neurotoxicity after adoptive immunotherapy with CAR T cells. Blood. 2019;133(20):2212–21.

    Article  CAS  Google Scholar 

  29. Rubin DB, et al. Neurological toxicities associated with chimeric antigen receptor T-cell therapy. Brain. 2019;142(5):1334–48.

    Article  Google Scholar 

  30. Tallantyre EC, et al. Neurological updates: neurological complications of CAR-T therapy. J Neurol. 2021;268(4):1544–54.

    Article  Google Scholar 

  31. •• Maus MV, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer. 2020;8(2):e001511 The most recent Society for Immunotherapy of Cancer clinical practice guideline on immune effector cell-related adverse events comprehensively outlines an evidence- and consensus- based approach to cellular therapy-related toxicity.

  32. Hayden PJ, et al. Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol. 2022;33(3):259–75.

    Article  CAS  Google Scholar 

  33. Thompson JA, et al. Management of immunotherapy-related toxicities, Version 1.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022;20(4):387–405.

    Article  CAS  Google Scholar 

  34. Lee DW, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625–38.

    Article  CAS  Google Scholar 

  35. Chakraborty R, et al. Late effects after chimeric antigen receptor T cell therapy for lymphoid malignancies. Transplant Cell Ther. 2021;27(3):222–9.

    Article  CAS  Google Scholar 

  36. Gonzalez Castro LN, Dietrich J. Evaluation and management of chimeric antigen receptor (CAR) T-cell-associated neurotoxicity. Neurooncol Pract. 2021;8(3):259–65.

    Google Scholar 

  37. Santomasso BD, et al. Management of immune-related adverse events in patients treated with chimeric antigen receptor T-cell therapy: ASCO Guideline. J Clin Oncol. 2021;39(35):3978–92.

    Article  CAS  Google Scholar 

  38. Locke FL, et al. Preliminary results of prophylactic tocilizumab after axicabtageneciloleucel (axi-cel; KTE-C19) treatment for patients with refractory,aggressive non-Hodgkin lymphoma (NHL). Blood. 2017;130:1547.

    Article  Google Scholar 

  39. • Scholler N, et al. Tumor immune contexture is a determinant of anti-CD19 CAR T cell efficacy in large B cell lymphoma. Nat Med. 2022;28(9):1872–82 This paper examined pre-treatment and post-treatment tumor immune contexture and its influence on CAR T-cell-related outcomes among patients on the ZUMA-1 trial. They found that a pre-treatment tumor microenvironment with a low density of regulatory T-cells was associated with an increased risk of neurotoxicity in those treated with axicabtagene ciloleucel.

  40. • Saini NY, et al. Clonal hematopoiesis is associated with increased risk of severe neurotoxicity in axicabtagene ciloleucel therapy of large B-cell lymphoma. Blood Cancer Discov. 2022;3(5):385–93 This publication examined the impact of clonal hematopoiesis prior to receiving axicabtagene ciloleucel on outcomes and found it was associated with an increased risk of severe neurotoxicity.

  41. Gaulin C, Kelemen K, Arana Yi C. Molecular pathways in clonal hematopoiesis: from the acquisition of somatic mutations to transformation into hematologic neoplasm. Life. 2022;12(8):1135.

    Article  CAS  Google Scholar 

  42. Novo M, et al. Axicabtagene ciloleucel chimeric antigen receptor T cell therapy in lymphoma with secondary central nervous system involvement. Mayo Clin Proc. 2019;94(11):2361–4.

    Article  Google Scholar 

  43. Frigault MJ, et al. Tisagenlecleucel CAR T-cell therapy in secondary CNS lymphoma. Blood. 2019;134(11):860–6.

    Article  CAS  Google Scholar 

  44. Lin Y, et al. Experience with axicabtagene ciloleucel (Axi-cel) in patients with secondary CNS involvement: results from the US Lymphoma CAR T Consortium. Blood. 2019;134(Supplement_1):763.

    Article  Google Scholar 

  45. Ghafouri S, et al. Axicabtagene Ciloleucel CAR T-cell therapy for relapsed/refractory secondary CNS non-Hodgkin lymphoma: comparable outcomes and toxicities, but shorter remissions may warrant alternative consolidative strategies? Bone Marrow Transplant. 2021;56(4):974–7.

    Article  CAS  Google Scholar 

  46. Abramson JS, et al. Anti-CD19 CAR T cells in CNS diffuse large-B-cell lymphoma. N Engl J Med. 2017;377(8):783–4.

    Article  Google Scholar 

  47. Traube C, et al. Cornell Assessment of Pediatric Delirium: a valid, rapid, observational tool for screening delirium in the PICU*. Crit Care Med. 2014;42(3):656–63.

    Article  Google Scholar 

  48. Castaneda-Puglianini O, Chavez JC. Assessing and management of neurotoxicity after CAR-T therapy in diffuse large B-cell lymphoma. J Blood Med. 2021;12:775–83.

    Article  Google Scholar 

  49. Locke FL, et al. Tumor burden, inflammation, and product attributes determine outcomes of axicabtagene ciloleucel in large B-cell lymphoma. Blood Adv. 2020;4(19):4898–911.

    Article  CAS  Google Scholar 

  50. Hunter BD, Jacobson CA. CAR T-cell associated neurotoxicity: mechanisms, clinicopathologic correlates, and future directions. J Natl Cancer Inst. 2019;111(7):646–54.

    Article  Google Scholar 

  51. Sheth VS, Gauthier J. Taming the beast: CRS and ICANS after CAR T-cell therapy for ALL. Bone Marrow Transplant. 2021;56(3):552–66.

    Article  Google Scholar 

  52. Rubin DB, et al. Clinical predictors of neurotoxicity after chimeric antigen receptor T-cell therapy. JAMA Neurol. 2020;77(12):1536–42.

    Article  Google Scholar 

  53. Nellan A, et al. Improved CNS exposure to tocilizumab after cerebrospinal fluid compared to intravenous administration in rhesus macaques. Blood. 2018;132(6):662–6.

    Article  CAS  Google Scholar 

  54. •• Oluwole OO, et al. Prophylactic corticosteroid use in patients receiving axicabtagene ciloleucel for large B-cell lymphoma. Br J Haematol. 2021;194(4):690–700 This analysis of an exploratory cohort from the ZUMA-1 trial found that prophylactic and early corticosteroids and/or tocilizumab resulted in a low rate of neurologic events in patients who received axicabtagene ciloleucel without compromising efficacy.

  55. Jatiani SS, et al. Myeloma CAR-T CRS management with IL-1R antagonist anakinra. Clin Lymphoma Myeloma Leuk. 2020;20(9):632–636 e1.

    Article  Google Scholar 

  56. Wehrli M, et al. Single-center experience using anakinra for steroid-refractory immune effector cell-associated neurotoxicity syndrome (ICANS). J Immunother Cancer. 2022;10(1):01.

    Article  Google Scholar 

  57. Strati P, et al. Clinical efficacy of anakinra to mitigate CAR T-cell therapy-associated toxicity in large B-cell lymphoma. Blood Adv. 2020;4(13):3123–7.

    Article  CAS  Google Scholar 

  58. Shah NN, et al. Intrathecal chemotherapy for management of steroid-refractory CAR T-cell-associated neurotoxicity syndrome. Blood Adv. 2020;4(10):2119–22.

    Article  Google Scholar 

  59. Garfall AL, et al. Posterior reversible encephalopathy syndrome (PRES) after infusion of anti-Bcma CAR T cells (CART-BCMA) for multiple myeloma: successful treatment with cyclophosphamide. Blood. 2016;128(22):5702.

    Article  Google Scholar 

  60. Kenderian SS, et al. ZUMA-19: a phase 1/2 multicenter study of lenzilumab use with axicabtagene ciloleucel (axi-cel) in patients (pts) with relapsed or refractory large B cell lymphoma (R/R LBCL). Blood. 2020;136(Supplement 1):6–7.

    Article  Google Scholar 

  61. Zhang L, et al. Etanercept as a new therapeutic option for cytokine release syndrome following chimeric antigen receptor T cell therapy. Exp Hematol Oncol. 2021;10(1):16.

    Article  CAS  Google Scholar 

  62. Kenderian SS, et al. Ruxolitinib prevents cytokine release syndrome after CART cell therapy without impairing the anti-tumor effect in a xenograft model. Blood. 2016;128(22):652.

    Article  Google Scholar 

  63. Pan J, et al. Ruxolitinib mitigates steroid-refractory CRS during CAR T therapy. J Cell Mol Med. 2021;25(2):1089–99.

    Article  CAS  Google Scholar 

  64. Weber EW, et al. Pharmacologic control of CAR-T cell function using dasatinib. Blood Adv. 2019;3(5):711–7.

    Article  CAS  Google Scholar 

  65. 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. 2016;22(11):2684–96.

    Article  CAS  Google Scholar 

  66. Ruella M, et al. Kinase inhibitor ibrutinib to prevent cytokine-release syndrome after anti-CD19 chimeric antigen receptor T cells for B-cell neoplasms. Leukemia. 2017;31(1):246–8.

    Article  CAS  Google Scholar 

  67. Siegler EL, Kenderian SS. Neurotoxicity and cytokine release syndrome after chimeric antigen receptor T cell therapy: insights into mechanisms and novel therapies. Front Immunol. 2020;11:1973.

    Article  CAS  Google Scholar 

  68. Maillet D, et al. Evaluation of mid-term (6-12 months) neurotoxicity in B-cell lymphoma patients treated with CAR T cells: a prospective cohort study. Neuro Oncol. 2021;23(9):1569–75.

    Article  CAS  Google Scholar 

  69. Ursu R, et al. Long-term neurological safety in B-cell lymphoma patients treated with anti-CD19 CAR T-cell therapy. Neurology 2022.

  70. Cordeiro A, et al. Late events after treatment with CD19-targeted chimeric antigen receptor modified T cells. Biol Blood Marrow Transplant. 2020;26(1):26–33.

    Article  CAS  Google Scholar 

  71. Ruark J, et al. Patient-reported neuropsychiatric outcomes of long-term survivors after chimeric antigen receptor T cell therapy. Biol Blood Marrow Transplant. 2020;26(1):34–43.

    Article  Google Scholar 

  72. Hoogland AI, et al. Change in neurocognitive performance among patients with non-Hodgkin lymphoma in the first year after chimeric antigen receptor T cell therapy. Transplant Cell Ther. 2022;28(6):305 e1–9.

    Article  Google Scholar 

  73. Van Oekelen O, et al. Neurocognitive and hypokinetic movement disorder with features of parkinsonism after BCMA-targeting CAR-T cell therapy. Nat Med. 2021;27(12):2099–103.

    Article  Google Scholar 

  74. •• Cohen AD, et al. Incidence and management of CAR-T neurotoxicity in patients with multiple myeloma treated with ciltacabtagene autoleucel in CARTITUDE studies. Blood Cancer J. 2022;12(2):32 This publication outlines the strategies implemented in patients receiving ciltacabtagene autoleucel in the CARTITUDE trials to monitor for and manage parkinsonism, among other movement and neurocognitive treatment-emergent adverse events.

  75. ABECMA® (idecabtagene vicleucel), suspension for intravenous infusion F.a.D. Administration, Editor. 2021, Celgene Corporation, a Bristol-Myers Squibb Company: Summit, NJ.

  76. Schmitt M, et al. Third-generation CAR T cells targeting CD19 are associated with an excellent safety profile and might improve persistence of CAR T cells in treated patients. Blood. 2019;134(Supplement_1):51.

    Article  Google Scholar 

  77. Roselli E, Faramand R, Davila ML. Insight into next-generation CAR therapeutics: designing CAR T cells to improve clinical outcomes. J Clin Invest. 2021;131(2):e142030.

    Article  CAS  Google Scholar 

  78. Spiegel JY, et al. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial. Nat Med. 2021;27(8):1419–31.

    Article  CAS  Google Scholar 

  79. Shah NN, et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial. Nat Med. 2020;26(10):1569–75.

    Article  CAS  Google Scholar 

  80. Depil S, et al. 'Off-the-shelf' allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov. 2020;19(3):185–99.

    Article  CAS  Google Scholar 

  81. Laskowski TJ, Biederstadt A, Rezvani K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat Rev Cancer. 2022;22:557–75.

    Article  CAS  Google Scholar 

  82. Bishop MR, et al. Second-line tisagenlecleucel or standard care in aggressive B-cell lymphoma. N Engl J Med. 2022;386(7):629–39.

    Article  CAS  Google Scholar 

  83. Locke FL, et al. Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma. N Engl J Med. 2022;386(7):640–54.

    Article  CAS  Google Scholar 

  84. Usmani SZ, et al. KarMMa-4: Idecabtagene vicleucel (ide-cel, bb2121), a BCMA-directed CAR T-cell therapy in high-risk newly diagnosed multiple myeloma. Journal of Clinical Oncology. 2021;39(15_suppl):TPS8053-TPS8053.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Maciej M. Mrugala MD, PhD, MPH.

Ethics declarations

Conflict of Interest

Dr. Fortin Ensign has nothing to disclose. Dr. Gaulin has nothing to disclose. Dr. Hrachova has nothing to disclose. Dr. Ruff has nothing to disclose. Dr. Harahsheh has nothing to disclose. Dr. Vicenti has nothing to disclose. Dr. Castro has nothing to disclose. Dr. Munoz reports personal fees from Pharmacyclics/Abbvie, Bayer, Gilead/Kite Pharma, Pfizer, Janssen, Juno/Celgene, BMS, Kyowa, Alexion, Fosunkite, Innovent, Seattle Genetics, Debiopharm, Karyopharm, Genmab, ADC Therapeutics, Epizyme, Beigene, Servier, Novartis, Morphosys/Incyte, Mei pharma, Zodiac, grants from Bayer, Gilead/Kite Pharma, Celgene, Merck, Portola, Incyte, Genentech, Pharmacyclics, Seattle Genetics, Janssen, Millennium, other from Targeted Oncology, OncView, Curio, Kyowa, Physicians’ Education Resource, Dava, Global clinical insights, MJH, Shanghai Youyao, and Seattle Genetics, other from Gilead/Kite Pharma, Kyowa, Bayer, Pharmacyclics/Janssen, Seattle Genetics, Acrotech/Aurobindo, Beigene, Verastem, AstraZeneca, Celgene/BMS, Genentech/Roche, during the conduct of the study. Dr. Rosenthal has nothing to disclose. Dr. Mrugala has nothing to disclose.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

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

This article is part of the Topical Collection on Neuro-oncology

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

Fortin Ensign, S.P., Gaulin, C., Hrachova, M. et al. Evaluating the Patient with Neurotoxicity after Chimeric Antigen Receptor T-cell Therapy. Curr. Treat. Options in Oncol. 23, 1845–1860 (2022). https://doi.org/10.1007/s11864-022-01035-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11864-022-01035-2

Keywords

Navigation