Granulocyte–macrophage colony-stimulating factor inactivation in CAR T-cells prevents monocyte-dependent release of key cytokine release syndrome mediators

Chimeric antigen receptor T-cell (CAR T-cell) therapy has been shown to be clinically effective for managing a variety of hematological cancers. However, CAR T-cell therapy is associated with multiple adverse effects, including neurotoxicity and cytokine release syndrome (CRS). CRS arises from massive cytokine secretion and can be life-threatening, but it is typically managed with an anti-IL-6Ra mAb or glucocorticoid administration. However, these treatments add to a patient's medication burden and address only the CRS symptoms. Therefore, alternative strategies that can prevent CRS and neurotoxicity associated with CAR T-cell treatment are urgently needed. Here, we explored a therapeutic route aimed at preventing CRS rather than limiting its consequences. Using a cytokine-profiling assay, we show that granulocyte–macrophage colony-stimulating factor (GMCSF) is a key CRS-promoting protein. Through a combination of in vitro experiments and gene-editing technology, we further demonstrate that antibody-mediated neutralization or TALEN-mediated genetic inactivation of GMCSF in CAR T-cells drastically decreases available GMCSF and abolishes macrophage-dependent secretion of CRS biomarkers, including monocyte chemoattractant protein 1 (MCP-1), interleukin (IL) 6, and IL-8. Of note, we also found that the genetic inactivation of GMCSF does not impair the antitumor function or proliferative capacity of CAR T-cells in vitro. We conclude that it is possible to prevent CRS by using “all-in-one” GMCSF-knockout CAR T-cells. This approach may eliminate the need for anti-CRS treatment and may improve the overall safety of CAR T-cell therapies for cancer patients.

[1]  Aaron J Johnson,et al.  GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. , 2019, Blood.

[2]  M. Gooderham,et al.  Granulocyte–macrophage colony‐stimulating factor (GM‐CSF) as a therapeutic target in psoriasis: randomized, controlled investigation using namilumab, a specific human anti‐GM‐CSF monoclonal antibody† , 2018, The British journal of dermatology.

[3]  Valérie Guyot,et al.  A Versatile Safeguard for Chimeric Antigen Receptor T-Cell Immunotherapies , 2018, Scientific Reports.

[4]  C. Doglioni,et al.  Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells , 2018, Nature Medicine.

[5]  M. Sadelain,et al.  CAR T cell–induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade , 2018, Nature Medicine.

[6]  K. Davis,et al.  Tisagenlecleucel in Children and Young Adults with B‐Cell Lymphoblastic Leukemia , 2018, The New England journal of medicine.

[7]  Stephen J. Schuster,et al.  Chimeric Antigen Receptor T Cells in Refractory B‐Cell Lymphomas , 2017, The New England journal of medicine.

[8]  R. Levy,et al.  Axicabtagene Ciloleucel CAR T‐Cell Therapy in Refractory Large B‐Cell Lymphoma , 2017, The New England journal of medicine.

[9]  M. Wurfel,et al.  Endothelial Activation and Blood-Brain Barrier Disruption in Neurotoxicity after Adoptive Immunotherapy with CD19 CAR-T Cells. , 2017, Cancer discovery.

[10]  N. Frey Cytokine release syndrome: Who is at risk and how to treat. , 2017, Best practice & research. Clinical haematology.

[11]  D. Teachey,et al.  Monocyte lineage-derived IL-6 does not affect chimeric antigen receptor T-cell function. , 2017, Cytotherapy.

[12]  T. Huizinga,et al.  Phase 1b randomized, double-blind study of namilumab, an anti-granulocyte macrophage colony-stimulating factor monoclonal antibody, in mild-to-moderate rheumatoid arthritis , 2017, Arthritis Research & Therapy.

[13]  Mithat Gönen,et al.  Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection , 2017, Nature.

[14]  D. Teachey,et al.  Cytokine Release Syndrome After Chimeric Antigen Receptor T Cell Therapy for Acute Lymphoblastic Leukemia , 2017, Critical care medicine.

[15]  Ming-Ru Wu,et al.  Mechanisms of Acute Toxicity in NKG2D Chimeric Antigen Receptor T Cell–Treated Mice , 2016, The Journal of Immunology.

[16]  A. Zhang,et al.  Third-generation CD28/4-1BB chimeric antigen receptor T cells for chemotherapy relapsed or refractory acute lymphoblastic leukaemia: a non-randomised, open-label phase I trial protocol , 2016, BMJ Open.

[17]  Burkhard Becher,et al.  GM-CSF: From Growth Factor to Central Mediator of Tissue Inflammation. , 2016, Immunity.

[18]  G. Wertheim,et al.  Identification of Predictive Biomarkers for Cytokine Release Syndrome after Chimeric Antigen Receptor T-cell Therapy for Acute Lymphoblastic Leukemia. , 2016, Cancer discovery.

[19]  K. Curran,et al.  Toxicity and management in CAR T-cell therapy , 2016, Molecular therapy oncolytics.

[20]  S. Amor,et al.  GM‐CSF promotes migration of human monocytes across the blood brain barrier , 2015, European journal of immunology.

[21]  J. Hamilton GM-CSF as a target in inflammatory/autoimmune disease: current evidence and future therapeutic potential , 2015, Expert review of clinical immunology.

[22]  S. Grupp,et al.  Refractory Cytokine Release Syndrome in Recipients of Chimeric Antigen Receptor (CAR) T Cells , 2014 .

[23]  S. Grupp,et al.  Current concepts in the diagnosis and management of cytokine release syndrome. , 2014, Blood.

[24]  M. Teixeira,et al.  The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases , 2014, Expert review of clinical immunology.

[25]  Qing He,et al.  Efficacy and Toxicity Management of 19-28z CAR T Cell Therapy in B Cell Acute Lymphoblastic Leukemia , 2014, Science Translational Medicine.

[26]  Jun S. Liu,et al.  Genetics of rheumatoid arthritis contributes to biology and drug discovery , 2013 .

[27]  Bernd Hauck,et al.  Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. , 2013, The New England journal of medicine.

[28]  A. Bagg,et al.  Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. , 2011, The New England journal of medicine.

[29]  F. Magrini,et al.  Extended Report , 2011 .

[30]  C. Ding,et al.  Tocilizumab: A Review of Its Safety and Efficacy in Rheumatoid Arthritis , 2010, Clinical medicine insights. Arthritis and musculoskeletal disorders.

[31]  S. Amini,et al.  Monocyte chemoattractant protein-1 (MCP-1): an overview. , 2009, Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research.

[32]  Irah L. King,et al.  Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. , 2009, Blood.

[33]  E. Ponomarev,et al.  GM-CSF Production by Autoreactive T Cells Is Required for the Activation of Microglial Cells and the Onset of Experimental Autoimmune Encephalomyelitis1 , 2007, The Journal of Immunology.

[34]  S. Salzano,et al.  Expression of GM-CSF receptor and "in vitro" effects of GM-CSF on human fibroblasts. , 1998, Life sciences.

[35]  M. Hüfner,et al.  [Plasma GM-CSF concentrations in rheumatoid arthritis, systemic lupus erythematosus and spondyloarthropathy]. , 1992, Zeitschrift fur Rheumatologie.

[36]  G. Firestein,et al.  Cytokines in chronic inflammatory arthritis. II. Granulocyte-macrophage colony-stimulating factor in rheumatoid synovial effusions. , 1989, The Journal of clinical investigation.

[37]  M. Vadas,et al.  The detection and initial characterization of colony-stimulating factors in synovial fluid. , 1988, Clinical and experimental immunology.

[38]  C. R. Clark,et al.  Granulocyte/macrophage colony-stimulating factor stimulates monocyte and tissue macrophage proliferation and enhances their responsiveness to macrophage colony-stimulating factor. , 1988, Blood.

[39]  A. Roberts,et al.  Targeting GM-CSF in inflammatory diseases , 2016, Nature Reviews Rheumatology.

[40]  J J Gómez-Reino Carnota,et al.  [Genetics of rheumatoid arthritis]. , 2000, Medicina clinica.