Remote control of therapeutic T cells through a small molecule–gated chimeric receptor

Keeping a leash on cancer-killing cells Redirecting the immune system to attack tumor cells is proving to be an effective therapy against cancer. However, when patients are exposed to T cells engineered to recognize and attack cancer cells, there is a risk of runaway or excessive activity or of off-target effects, both of which can themselves be deadly. Wu et al. designed T cells expressing chimeric antigen receptors that recognize and attack cancer cells with an additional control system. This mechanism would allow a doctor administering the therapy to turn the engineered T cell “on” or “off” by administering a small molecule that is required along with cancer cell antigen to stimulate the T cells and activate their tumor cell–killing properties. Science, this issue p. 10.1126/science.aab4077 Engineering a fail-safe control mechanism in cancer-targeted T cells. INTRODUCTION Cell-based therapies have emerged as a promising treatment modality for diseases such as cancer and autoimmunity. T cells engineered with synthetic receptors known as chimeric antigen receptors (CARs) have proven effective in eliminating chemotherapy-resistant forms of B cell cancers. Such CAR T cells recognize antigens on the surface of tumor cells and eliminate them. However, CAR T cells also have adverse effects, including life-threatening inflammatory side effects associated with their potent immune activity. Risks for severe toxicity present a key challenge to the effective administration of such cell-based therapies on a routine basis. RATIONALE Concerns about the potential for severe toxicity of cellular therapeutics primarily stem from a lack of precise control over the activity of the therapeutic cells once they are infused into patients. Exogenously imposed specific regulation over the location, duration, and intensity of the therapeutic activities of engineered cells would therefore be desirable. One way to achieve the intended control is to use small molecules to gate cellular functions. Small molecules with desired pharmacologic properties could be systemically or locally administered at varying dosages to achieve refined temporal and spatial control over engineered therapeutic cells. RESULTS We developed an ON-switch CAR that enables small molecule–dependent, titratable, and reversible control over CAR T cell activity. ON-switch CAR T cells required not only a cognate antigen but also a priming small molecule to activate their therapeutic functions. Depending on the amount of small molecule present, ON-switch CAR T cells exhibited titratable therapeutic activity, from undetectable to as strong as that of conventional CAR T cells. The ON-switch CAR was constructed by splitting key signaling and recognition modules into distinct polypeptides appended to small molecule–dependent heterodimerizing domains. The ON-switch CAR design is modular; different antigen recognition domains and small-molecule dimerizing modules can be swapped in. CONCLUSION The ON-switch CAR exemplifies a simple and effective strategy to integrate cell-autonomous decision-making (e.g., detection of disease signals) with exogenous, reversible user control. The rearrangement and splitting of key modular components provides a simple strategy for achieving integrated multi-input regulation. This work also highlights the importance of developing optimized bio-inert, orthogonal control agents such as small molecules and light, together with their cellular cognate response components, in order to advance precision-controlled cellular therapeutics. Titratable control of engineered therapeutic T cells through an ON-switch chimeric antigen receptor. A conventional CAR design activates T cells upon target cell engagement but can yield severe toxicity due to excessive immune response. The ON-switch CAR design, which has a split architecture, requires a priming small molecule, in addition to the cognate antigen, to trigger therapeutic functions. The magnitude of responses such as target cell killing can be titrated by varying the dosage of small molecule to mitigate toxicity. scFv, single-chain variable fragment; ITAM, immunoreceptor tyrosine-based activation motif. There is growing interest in using engineered cells as therapeutic agents. For example, synthetic chimeric antigen receptors (CARs) can redirect T cells to recognize and eliminate tumor cells expressing specific antigens. Despite promising clinical results, these engineered T cells can exhibit excessive activity that is difficult to control and can cause severe toxicity. We designed “ON-switch” CARs that enable small-molecule control over T cell therapeutic functions while still retaining antigen specificity. In these split receptors, antigen-binding and intracellular signaling components assemble only in the presence of a heterodimerizing small molecule. This titratable pharmacologic regulation could allow physicians to precisely control the timing, location, and dosage of T cell activity, thereby mitigating toxicity. This work illustrates the potential of combining cellular engineering with orthogonal chemical tools to yield safer therapeutic cells that tightly integrate cell-autonomous recognition and user control.

[1]  Wendell A. Lim,et al.  Counting molecules in single organelles with superresolution microscopy allows tracking of the endosome maturation trajectory , 2013, Proceedings of the National Academy of Sciences.

[2]  J. Lippincott-Schwartz,et al.  Imaging Intracellular Fluorescent Proteins at Nanometer Resolution , 2006, Science.

[3]  L. Banaszynski,et al.  Conditional control of protein function , 2006, Chemistry & biology.

[4]  C. Mason,et al.  Cell therapy industry: billion dollar global business with unlimited potential. , 2011, Regenerative medicine.

[5]  J. Lippincott-Schwartz,et al.  High-density mapping of single-molecule trajectories with photoactivated localization microscopy , 2008, Nature Methods.

[6]  David L. Porter,et al.  T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia , 2011, Science Translational Medicine.

[7]  Michel Sadelain,et al.  PD-1– and CTLA-4–Based Inhibitory Chimeric Antigen Receptors (iCARs) Divert Off-Target Immunotherapy Responses , 2013, Science Translational Medicine.

[8]  S. Kim,et al.  Human 4-1BB (CD137) signals are mediated by TRAF2 and activate nuclear factor-kappa B. , 1998, Biochemical and biophysical research communications.

[9]  Robert DeRose,et al.  Rapid and orthogonal logic gating with a gibberellin-induced dimerization system. , 2012, Nature chemical biology.

[10]  A. Weiss,et al.  Functional characterization of a signal transducing motif present in the T cell antigen receptor zeta chain , 1993, The Journal of experimental medicine.

[11]  R. Henry,et al.  Neural Stem Cell Engraftment and Myelination in the Human Brain , 2012, Science Translational Medicine.

[12]  B. Kuhlman,et al.  A genetically-encoded photoactivatable Rac controls the motility of living cells , 2009, Nature.

[13]  A. Weiss,et al.  The tyrosine phosphatase CD148 is excluded from the immunologic synapse and down-regulates prolonged T cell signaling , 2003, The Journal of cell biology.

[14]  Adrian P Gee,et al.  Inducible apoptosis as a safety switch for adoptive cell therapy. , 2011, The New England journal of medicine.

[15]  T. Dull,et al.  Targeting of human immunodeficiency virus-infected cells by CD8+ T lymphocytes armed with universal T-cell receptors , 1994 .

[16]  Michel Sadelain,et al.  The basic principles of chimeric antigen receptor design. , 2013, Cancer discovery.

[17]  A. Ganser,et al.  Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): a non-randomised phase I-II study. , 2009, The Lancet. Oncology.

[18]  M. Tuszynski,et al.  Long-Distance Growth and Connectivity of Neural Stem Cells after Severe Spinal Cord Injury , 2012, Cell.

[19]  E. Kieff,et al.  The structural basis for the recognition of diverse receptor sequences by TRAF2. , 1999, Molecular cell.

[20]  Dan R. Littman,et al.  Signal transduction by lymphocyte antigen receptors , 1994, Cell.

[21]  Wendell A. Lim,et al.  Cell-Based Therapeutics: The Next Pillar of Medicine , 2013, Science Translational Medicine.

[22]  Christina D Smolke,et al.  Genetic control of mammalian T-cell proliferation with synthetic RNA regulatory systems , 2010, Proceedings of the National Academy of Sciences.

[23]  Stuart L. Schreiber,et al.  Structure of the FKBP12-Rapamycin Complex Interacting with Binding Domain of Human FRAP , 1996, Science.

[24]  Mark M Davis,et al.  Continuous T cell receptor signaling required for synapse maintenance and full effector potential , 2003, Nature Immunology.

[25]  L. Kane Faculty Opinions recommendation of Distinct TCR signaling pathways drive proliferation and cytokine production in T cells. , 2013 .

[26]  Yoshinori Hirano,et al.  Gibberellin-induced DELLA recognition by the gibberellin receptor GID1 , 2008, Nature.

[27]  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.

[28]  Martin Fussenegger,et al.  The use of light for engineered control and reprogramming of cellular functions. , 2012, Current opinion in biotechnology.

[29]  T. Dull,et al.  Targeting of human immunodeficiency virus-infected cells by CD8+ T lymphocytes armed with universal T-cell receptors. , 1994, Blood.

[30]  S. Rosenberg,et al.  Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

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

[32]  P. Simms,et al.  Utility of flow cytometric detection of CD69 expression as a rapid method for determining poly- and oligoclonal lymphocyte activation , 1996, Clinical and Diagnostic Laboratory Immunology.

[33]  J. Bluestone,et al.  Current and future immunomodulation strategies to restore tolerance in autoimmune diseases. , 2012, Cold Spring Harbor perspectives in biology.

[34]  W. Wilson,et al.  B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. , 2012, Blood.

[35]  S. Schreiber,et al.  Proximity and orientation underlie signaling by the non‐receptor tyrosine kinase ZAP70 , 1997, The EMBO journal.

[36]  M. Kalos,et al.  Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. , 2013, Immunity.

[37]  S. Rosenberg,et al.  Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors , 2013, Nature Reviews Clinical Oncology.

[38]  Christopher A. Voigt,et al.  Spatiotemporal Control of Cell Signalling Using A Light-Switchable Protein Interaction , 2009, Nature.

[39]  Avijit Ray,et al.  Isolation of Mouse Peritoneal Cavity Cells , 2010, Journal of visualized experiments : JoVE.

[40]  Lloyd J. Old,et al.  The roles of IFNγ in protection against tumor development and cancer immunoediting , 2002 .

[41]  Benjamin Rhau,et al.  Synthetic control of mammalian-cell motility by engineering chemotaxis to an orthogonal bioinert chemical signal , 2014, Proceedings of the National Academy of Sciences.

[42]  Jared E. Toettcher,et al.  Light-based feedback for controlling intracellular signaling dynamics , 2011, Nature Methods.

[43]  Wendell A Lim,et al.  Rewiring cell signaling: the logic and plasticity of eukaryotic protein circuitry. , 2004, Current opinion in structural biology.

[44]  Jen Jen Yeh,et al.  Local iontophoretic administration of cytotoxic therapies to solid tumors , 2015, Science Translational Medicine.

[45]  Anthony C. Bishop,et al.  Unnatural ligands for engineered proteins: new tools for chemical genetics. , 2000, Annual review of biophysics and biomolecular structure.

[46]  S. Schreiber,et al.  Inducible gene expression and protein translocation using nontoxic ligands identified by a mammalian three-hybrid screen. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[47]  J. Sprent,et al.  The role of interleukin-2 during homeostasis and activation of the immune system , 2012, Nature Reviews Immunology.

[48]  Jun Wu,et al.  An activating immunoreceptor complex formed by NKG2D and DAP10. , 1999, Science.

[49]  K. Stankunas,et al.  Rapamycin analogs with differential binding specificity permit orthogonal control of protein activity. , 2006, Chemistry & biology.

[50]  Martin Fussenegger,et al.  Synthetic mammalian gene circuits for biomedical applications. , 2013, Current opinion in chemical biology.

[51]  Ronald D. Vale,et al.  Single-Molecule Microscopy Reveals Plasma Membrane Microdomains Created by Protein-Protein Networks that Exclude or Trap Signaling Molecules in T Cells , 2005, Cell.

[52]  W. Lim,et al.  Integration of multiple signals through cooperative regulation of the N-WASP-Arp2/3 complex. , 2000, Science.

[53]  Mark M Davis,et al.  T cell killing does not require the formation of a stable mature immunological synapse , 2004, Nature Immunology.

[54]  M. Konrad,et al.  Engineered human tmpk/AZT as a novel enzyme/prodrug axis for suicide gene therapy. , 2007, Molecular therapy : the journal of the American Society of Gene Therapy.

[55]  Marcela V Maus,et al.  Antibody-modified T cells: CARs take the front seat for hematologic malignancies. , 2014, Blood.

[56]  D. Campana,et al.  Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[57]  Jan E. Schnitzer,et al.  Role of GTP Hydrolysis in Fission of Caveolae Directly from Plasma Membranes , 1996, Science.