Team:SYSU-CHINA/Description







Description


Abstract

CAR-T therapy is one of the most promising treatment for cancer, with multiple ongoing clinical trials worldwide and 2 therapies approved by the FDA. However, without proper control after administration of CAR-T cells, severe adverse effects may bring fatal risks to the patients, especially during the clinical trial stages. While suicide switches serve as common methods for controlling adverse effects, they completely halt the expensive treatment, and repeating the treatment process could be a burden for the patients, both physically and financially. To provide a safer yet affordable CAR-T therapy, we developed a reversible safe switch controlled by small molecules called CAR BRAKE. By expressing U24 protein of the human herpesvirus 6A under the control of tet-ON promoter, we can downregulate CAR molecules on the cell surface through endosomal recycling inhibition. We demonstrated in Jurkat cells that U24 can downregulate surface T cell receptors, which can be subsequently restored by removal of doxycycline. Modest effect of U24 on surface level of CARs was seen in Jurkat cells. In addition, we determined the optimal concentration for induction, expression time course and degradation half-life of U24 in HEK293T cells. No cytotoxicity was observed in cells transfected with U24. Collectively, our results indicates our design could potentially be used as a universal add-on for all CAR-Ts and TCR-Ts to ensure safety.


Background
CAR-T Therapy

CAR-T (chimeric antigen receptor T cell) therapy is one of the most promising treatment for cancer, with multiple ongoing clinical trials worldwide and 2 therapies already approved by the FDA(June et al., 2018) .

The therapy engineers endogenous T cells from the patients with synthetic receptors for tumor antigens, enabling them to target tumor cells. The synthetic receptor, or chimeric antigen receptor (CAR), consists of an extracellular antibody-derived single chain variable fragment (scFv) domain that can bind specific antigen with high affinity, a transmembrane domain, and an intracellular signaling domain. The 1st generation CAR molecules only include a CD3zeta signaling domain, the minimal domain for T cell activation, while 2nd and 3rd generation CARs also include one or more co-stimulating domains from CD28, CD137, 4-1BB, or others. The addition signals enhance cytokines production and facilitate expansion of CAR T cells population upon repeated exposure to target
antigens(June et al., 2018) .


Similar to other types of adoptive cell transfer (ACT) therapy, CAR T therapy requires drawing peripheral blood from the patients, Sorting out T cells, introducing new receptors by transducing T cells with lentiviral vectors, and expanding the population in vitro before re-infusion into patients.


CAR T therapy has achieved unprecedented success in hematological cancer. In several clinical trials using CAR T to treat leukemia and lymphoma, high percentage of patients experienced partial or complete response(June and Sadelain, 2018) .

The Adverse Effects

However, without proper control after administration of CAR-T cells, severe adverse effects associated with CAR-T therapy may bring fatal risks to the patients, especially during the clinical trial stages. The most common side effects include cytokine release syndrome, on-target off-tumor toxicity and off-target toxicity.

cytokine release syndrome (CRS)

CRS is considered the most common acute toxicity in the CAR T therapy, with patients experiencing fever, tachycardia, hypotension, and other symptoms after infusion of CAR T cells. The cytokines can be produced by CAR T cells themselves or by other immune cells like macrophages. Likewise, a wide range of cytokines participate in these pathological conditions, including interleukin-6 (IL-6), IL-2 and interferon-γ(Maude et al., 2014) . The CRS development correlates with high level of CAR T cells number and peak level of several cytokines and is also related to tumor burden(Brentjens et al., 2013) .


Neurotoxicity

Neurotoxicity of CAR T therapy has been reported by several research groups. The symptoms include confusion, delirium, expressive aphasia, obtundation, myoclonus, and seizure. Similar neurological toxicity has been reported with blinatumomab, a bispecific anti- CD19 anti-CD3 antibody(Brudno and Kochenderfer, 2016) . However, the underlying mechanism remains unknown(June et al., 2018) .

On-target off-tumor toxicity

On-target off-tumor toxicity is induced when the target antigen is also expressed on non-tumor tissues, despite its lower expression level, and CAR T cells can inflict normal tissue damage by target antigen recognition. Such toxicity has been documented in the literature and has even lead to fatality. In one study 3 patients with metastatic renal cell carcinoma receiving CAR T cells targeting carboxyanhydrase-IX (CAIX) developed liver damage, due to unexpected expression of CAIX on the bile ducts (Lamers et al., 2013) . The risk is further exemplified by another study in which a patient with metastatic colorectal cancer experienced acute respiratory distress and pulmonary edema and subsequently died after infusion of CAR Ts targeting ERBB2 (Her-2/neu), possibly due to expression of ERBB2 on lung tissue(Morgan et al., 2010) .

Cross-reactivity

Cross-reactivity occurs when CAR T cells recognize antigens on normal tissues similar to target antigen, possibly leading to organ damage. By far no cross reactivity of CAR T cells has been reported, instead such toxicity has been observed in clinical trials using modified TCR T cells(Cameron et al., 2013) .

Current Methods
Immunosuppressive drugs

To date the most common method for CRS management is giving immunosuppressive drugs. Tocilizumab (Actemra), an anti-interleukin-6- receptor antagonist originally developed for rheumatoid arthritis, is often effective in the management of CAR T cells related cytokine release syndrome and has been approved by the FDA for such treatments(Brudno and Kochenderfer, 2016) .

Corticosteroids are given when patients are not responsive to Tocilizumab, but they are not considered as the first option since they can interfere with T cell functions and induce T cell apoptosis(Brudno and Kochenderfer, 2016) , with some research demonstrating that prolonged administration of corticosteroids can impair CAR T therapy efficacy (Davila et al., 2014) .

A potential risk for immunosuppressive treatment is its global effect on the immune system, rendering the patients more vulnerable to opportunistic infections by bacteria, fungi and viruses. It has been demonstrated that rheumatoid arthritis patients have higher rate of respiratory infections(Hoshi et al., 2012) . In addition, a recent study suggested a correlation between CRS and rate of infection(Hill et al., 2018) . Although no causative conclusion can be drawn that the immunosuppressive treatment for CRS leads to those infection, the risk of immunosuppression- associated infections should not be overlooked, and it is desirable to mitigate adverse effects in CAR T therapies without global inhibition on the immune system.


Suicide Genes

Compared to the systemic effect of immunosuppressive drugs, genetic modification of the infused CAR T cells is able to provide a more precise control on the infused cell population alone. A straightforward method is to engineer suicide genes in CAR T cells and remove transferred cell population when giving drugs(Marin et al., 2012) . The first suicide gene evaluated in the clinical trials is thymidine kinase from herpes simplex virus, which can induce cell death by DNA synthesis inhibition upon treatment with ganciclovir. Inducible dimerization of Fas or caspase 9 serves as a second strategy for selective depletion of modified cells. The inducible caspase 9 (ICas9) molecule contains an FK506 binding domain and is able to mediate dimerization of caspase 9 domain when adding AP1903, and ultimately induce cell apoptosis(Budde et al., 2013) . A third method is achieved by programming the cells to express truncated epidermal growth factor receptors on the cell surface, which can be targeted by antibody and induce antibody-dependent cell-mediated cytotoxicity (ADCC) (Wang et al., 2011) .

However, these methods could deplete CAR-T cells and halt the expensive treatment. Repeat the treatment may be a burden on the patients, both physically and financially.

Controllable Receptors

Another strategy is to engineer the CAR receptors, rendering them requiring additional signals, typically a small molecule drug, to fully activate. Wu et al. (Wu et al., 2015) first demonstrated the such strategy by constructing a split synthetic receptor, with one chain containing the extracellular scFv, hinge and costimulatory domain and the other containing the activation domain of CD3zeta. When adding small molecule Rapalog, the heterodimerization of 2 chains reconstitutes a full CAR receptor and therefore can generate full activation signals. Similarly, Juillerat et al. (Juillerat et al., 2016) modified the hinge domain to include a FKBP/FRB dimerization pair, which separate the scFv from the cell membrane. When AP21967 (a rapamycin analog) is added, the interaction of FKBP and FRB fold the extracellular domain into an optimal conformation for activation.


However, modification of receptors may be troublesome that it may alter the structure and conformation of the receptors, which in turns may interfere with CARs' function. Such notion is exemplified in the research of Wu et al., that different split methods resulted in different signaling outcomes, with most of the receptor pairs failing to fully activate in the presence of drugs (Wu et al., 2015) . Considering a lack of conclusive information on mechanisms of CAR T cell activation(Harris and Kranz, 2016) and the diversity of existing CARs in clinical research(June et al., 2018) , a method must be developed for each type of CAR to ensure it works as expected, which can be labor-intensive.

Furthermore, such methods are limited to CAR Ts alone, which have synthetic receptors. In contrast, TCR Ts rely on the endogenous T cell receptor complex to function properly, and TCRs, as part of the complex, requires native conformation to function, leaving little, if any, space for modification.

Reference

Brentjens, R.J., Davila, M.L., Riviere, I., Park, J., Wang, X., Cowell, L.G., Bartido, S., Stefanski, J., Taylor, C., Olszewska, M., et al. (2013). CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Science translational medicine 5, 177ra138.
Brudno, J.N., and Kochenderfer, J.N. (2016). Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 127, 3321-3330.
Budde, L.E., Berger, C., Lin, Y., Wang, J., Lin, X., Frayo, S.E., Brouns, S.A., Spencer, D.M., Till, B.G., Jensen, M.C., et al. (2013). Combining a CD20 chimeric antigen receptor and an inducible caspase 9 suicide switch to improve the efficacy and safety of T cell adoptive immunotherapy for lymphoma. PloS one 8, e82742.
Cameron, B.J., Gerry, A.B., Dukes, J., Harper, J.V., Kannan, V., Bianchi, F.C., Grand, F., Brewer, J.E., Gupta, M., Plesa, G., et al. (2013). Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Science translational medicine 5, 197ra103.
Davila, M.L., Riviere, I., Wang, X., Bartido, S., Park, J., Curran, K., Chung, S.S., Stefanski, J., Borquez-Ojeda, O., Olszewska, M., et al. (2014). Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Science translational medicine 6, 224ra225.
Harris, D.T., and Kranz, D.M. (2016). Adoptive T Cell Therapies: A Comparison of T Cell Receptors and Chimeric Antigen Receptors. Trends in pharmacological sciences 37, 220-230.
Hill, J.A., Li, D., Hay, K.A., Green, M.L., Cherian, S., Chen, X., Riddell, S.R., Maloney, D.G., Boeckh, M., and Turtle, C.J. (2018). Infectious complications of CD19-targeted chimeric antigen receptor-modified T-cell immunotherapy. Blood 131, 121-130.
Hoshi, D., Nakajima, A., Inoue, E., Shidara, K., Sato, E., Kitahama, M., Seto, Y., Tanaka, E., Urano, W., Ichikawa, N., et al. (2012). Incidence of serious respiratory infections in patients with rheumatoid arthritis treated with tocilizumab. Modern rheumatology 22, 122-127.
Juillerat, A., Marechal, A., Filhol, J.M., Valton, J., Duclert, A., Poirot, L., and Duchateau, P. (2016). Design of chimeric antigen receptors with integrated controllable transient functions. Scientific reports 6, 18950.
June, C.H., O'Connor, R.S., Kawalekar, O.U., Ghassemi, S., and Milone, M.C. (2018). CAR T cell immunotherapy for human cancer. Science 359, 1361-1365.
June, C.H., and Sadelain, M. (2018). Chimeric Antigen Receptor Therapy. The New England journal of medicine 379, 64-73.
Lamers, C.H., Sleijfer, S., van Steenbergen, S., van Elzakker, P., van Krimpen, B., Groot, C., Vulto, A., den Bakker, M., Oosterwijk, E., Debets, R., et al. (2013). Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Molecular therapy : the journal of the American Society of Gene Therapy 21, 904-912.
Marin, V., Cribioli, E., Philip, B., Tettamanti, S., Pizzitola, I., Biondi, A., Biagi, E., and Pule, M. (2012). Comparison of different suicide-gene strategies for the safety improvement of genetically manipulated T cells. Human gene therapy methods 23, 376- 386.
Maude, S.L., Frey, N., Shaw, P.A., Aplenc, R., Barrett, D.M., Bunin, N.J., Chew, A., Gonzalez, V.E., Zheng, Z., Lacey, S.F., et al. (2014). Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England journal of medicine 371, 1507-1517.
Morgan, R.A., Yang, J.C., Kitano, M., Dudley, M.E., Laurencot, C.M., and Rosenberg, S.A. (2010). Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Molecular therapy : the journal of the American Society of Gene Therapy 18, 843-851.
Wang, X., Chang, W.C., Wong, C.W., Colcher, D., Sherman, M., Ostberg, J.R., Forman, S.J., Riddell, S.R., and Jensen, M.C. (2011). A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood 118, 1255-1263.
Wu, C.Y., Roybal, K.T., Puchner, E.M., Onuffer, J., and Lim, W.A. (2015). Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 350, aab4077.