Part1: Minimalist controllable switch

1. Background

CD3ζ is a fixed module in all existing chimeric antigen receptor (CAR) as the primary transmitter of endogenous T-cell receptor (Wu et al., 2020). This part is an improved part of cytoplasmic domain of CD3ζ chain (BBa_K3244012) posted by iGEM19_AFCM-Egypt.

Synthetic biology requires the specific control systems that are functionalized to perform user-defined, precisely controlled regulation. Furthermore, considering that anti-aging therapy needs to be continuous, stable, and gentle, choosing common and no-threatening molecules would be more beneficial for public recognition as well as circumventing the adverse effects of drugs.

Using caffeine to modulate CD3ζ signaling is a great potential application in CAR due to its non-toxicity, low cost and easy obtain. COSMO is an optimized caffeine antibody based on wild-type acVHH. It is a robust chemically induced dimerization system with high affinity towards caffeine (EC50 = 16.9 × 10−9 m) (Wang et al., 2021).

2. Design

In this experiment, we added a COSMO before CD3ζ to regulate its activation, we can construct a minimal and precisely controllable CAR-M (Fig.1, BBa_K4175052).


Figure 1. Schematic diagram depicting the activation of CAR modules under different conditions.

CAR-M cells will only kill normally when both target cells and caffeine are present. When only caffeine is present, CD3ζ will be activated solely; while when only target cells are available, inactivated COSMO will block upstream signaling and only 4-1BB domain will be activated. As CD3ζ and 4-1BB cannot be activated simultaneously, the signal for cell activation is too weak to initiate downstream pathway for secreting perforin, granzyme, etc. (Philipson et al., 2020; Wu et al., 2020).

3. Map


Figure 2. Map of COSMO (BBa_K4175052)

4. Results

The CAR-COSMO-M killing system showed good killing after the addition of coffee, but the CAR-COSMO modulation system did not seem to completely inhibit the killing of Raji cell lines by CAR-M. Also, the CAR-M system did not show killing in the absence of coffee, which is not as expected. Considering the effect of coffee color on measured value of the zymograph, we believe that this result is in error with the real result, although the CAR-COSMO-M killing system exhibited good killing in the presence of caffeine.

We will explore the removal of the pigment in caffeine or the use of flow-through assays to eliminate the effect of coffee color on the experimental results.


Figure 3. Results of COSMO

Part2: Negative feedback loop


Figure 4. Overview of Negative feedback loop

Module1: Blocking expression

1. Introduction

It is an improvement over part of NMU_China 2021 design of “Toggle Macrophage: Synthetic Antiviral Macrophages with Immune-regulatory Capacity” (BBa K4040021 and BBa K4040020). This system utilizes IL6R and gp130 to sense IL-6 concentration in the environment (Fig.1①). When low-affinity IL6R binds to IL6 with high concentration, it will recruit gp130, IL-6 cytokine family signal transducer. Its downstream-linked TEV protease will bind to TCS cleavage site, downstream of IL6R, releasing Gal4-KRAB. Eventually, Gal4-KRAB will combine with UAS-pSV40, inhibiting the following gene transcription.

However, due to three long plasmids, it is hard to have high transfection efficiency and difficulty in obtaining enough cells with high positive rates. We hope to simplify the protein structure to shorten the length of plasmids.

Furthermore, cytokine release syndrome (CRS) has been observed in patients with CAR-T treatment, including numerous successful trials, causing severe or even lethal releases of inflammatory cytokine levels, especially IL-6, one of CRS biomarkers (Rafiq et al., 2020). To alleviate CRS, we designed to apply this improved system to inhibit CAR expression by recognizing high concentration of IL-6 to slow down the progress of CRS; and, at the same time, restarts CAR expression at low concentrations to continue killing tumor cells.

2. Design

We utilized ‘notch’ to replace ‘TCS’ and ‘TEV’, functioning as freeing up Gal4-KRAB as shown in Fig1 ② and ③. The ‘notch’ part is the minimal core transmembrane domain of the wild-type notch, controlling proteolysis (Morsut et al., 2016). With activation of high concentration of IL-6 and sending signaling to the ‘notch’, ‘notch’ will change the conformation and expose multiple proteolytic cleavage site. Various cells have the necessary proteolytic machinery for ‘notch’ activation such that Gal4-KRAB can be released to combine with the UAS-pSV40. Another improved part is that we changed IL6R to IL-6 scFv (Tan Hong Ji and Campana, 2021). ScFv with lower affinity could make sure the negative feedback system only launch when sensing aberrantly high IL-6 levels.

3. Map

a.



b.


Figure 5. Results of COSMO

Module2: Disrupts CAR signaling downstream

1. Background

Blocking expression of CAR-T only can slow down the onset of CRS. If T cell activation can be inhibited rather than blocking new expression of CAR on the cell membrane through high IL-6 level, CRS will be alleviated and relieved.

Inhibitory CAR (i.e., iCAR) utilized intracellular domain of inhibitory receptors, PD-1 or CTLA-4, to disrupt T cell activation, proliferation, and cytokine release (Fedorov et al., 2013). It is designed as a dynamic safety guard to prevent ‘off-target’ toxicities when CAR-T cells are recognized to bind to normal cells. Compared with CTLA-4-based iCARs, PD-1-based iCARs showed more efficiency to inhibit numbers, proliferation and cytokine released of CAR-T cells.

2. Design

Learning from iCAR, We designed a synthetic receptor with extracellular and transmembrane domains of IL-6R and intracellular domain of PD-1 (Fig.1 ④). The amino acid sequence of PD-1 was obtained with the Ensembl sequence, amino acids 191 to 289. With high level of IL-6, intracellular domain of PD-1 will be activated to inhibit downstream of T cell activation signals.

3. Map


Figure 6. Map of PD1 (BBa K4175011)

Results


Figure 7. Results of PD1 (blue, Raji; orange, Raji cocultured with untransfected Jurkat; gray, Raji cocultured with transfected Jurkat). The title of each panel represents IL6 concentration (pg/ml).

In humans, the concentration of IL6 is usually maintained at 0.00-2.90 pg/ml. With this in mind, we set up two sets of normal concentrations and three sets of abnormal concentrations to treat the cells that were then transferred. We found that the killing ability of Jurkat was enhanced after IL6 treatment, as shown in Figure 7. In the design concept of module1, we were expecting the expression of CAR to be inhibited at abnormal IL6 concentrations, thus reducing the side effects of killing; hence the problem with the system we designed. As the results of PD1 are shown in Figure 7, we can see that at high concentrations, PD1 successfully disrupts the signaling downstream of CAR-T, reducing the killing ability.

Although the experiment successfully proved that our system is problem-free, the activity of transfected Jurkat is not really high and the choice of dual promoter in the plot resulted in not high CAR expression, thus his killing is not very obvious. In addition, the concentration gradient in the normal group was set low and additional experimental groups should be set up in subsequent experiments.

Part3: Engineering Erythrocyte Killing

1. Background

In order to address the problem of high anti-aging CAR-T cost as reflected from human practices, we systematically analyzed the promising ways to solve the problem. The most widely used universal CAR-T (a type of CAR-T cell with TCR knockdown) is currently on the market. But given its potential host resistance problem and the technical challenges faced by secondary gene editing is beyond our capabilities (T cells start to be exhausted in about two weeks after isolation). We would like to explore a simpler approach, and another solution strategy is the in vivo production of CAR-T cells. Most of the previous studies have produced CAR-T cells in vivo by nanocarriers or liposomes, but they face problems of immunogenicity as well as organ delivery efficiency that are difficult to solve. Recently, exosomes have been widely studied as an excellent small molecule carrier, so we hope to solve this problem by exosomes. Also, considering that exosomes are secreted by cells, based on the idea of capsule cells in synthetic biology, we would like to develop a synthetic cell based on this and use it to produce CAR-T cells in vivo. Exosomes were first found in reticulocytes, while red blood cells, as one of the main components of blood transfusion, have a high degree of versatility among people of the same blood type. Therefore, we wanted to rely on erythrocytes as a medium (immature erythrocytes produce a large number of exosomes), and we considered erythrocytes as a potential engineered vector, considering their short self-life and the fact that they are not easily edited to become tumorigenic.

2. Design

The entire project is shown in the figure below:



At the technical level, the genetic editing of cells required for the production of exosomes by engineered cell carriers includes 1. promoting exosome production, 2. packaging mRNA exosomes, and 3. enhancing exosome targeting. As a well-studied vector, the commercial pCDH-CMV-exosome booster-EF1-copGFP-Puro plasmid can do the job of promoting exosome production and has also been validated many times, Designer exosomes produced by implanted cells The article intracerebrally deliver therapeutic cargo for Parkinson's disease treatment describes an approach to engineer exosomes that allows for packaging and targeting of exosomes. We therefore wish to construct a cellular platform for the efficient production of exosomes as shown in the figure below.



Materials:

Plasmids:

1. pCDH-CMV-exosome booster-EF1-copGFP-Puro

2. EEK-CD5-CAR



3. EEK-CD5-nluc



4. EEK-CD160-CAR



5. EEK-CD160-nluc



Cell Lines and Cells

HEK 293T cell line

Primary erythroblasts

(Isolation of MNCs and enrichment of primary erythroblasts from adult peripheral blood)

T cells & NK cells

(Separation and Enrichment from adult peripheral blood)

Animal Models

Modelled haematological cancers by systemically injecting luciferase-expressing Eμ-ALL01 leukaemia cells into 4–6-week-old female albino B6 (C57BL/6J-Tyr < c-2J>) mice and allowing them to develop for 1 week

The whole experimental design is described as following:



3. Critical Thinking

Our future projects have the following advantages.

  • The exosomes lack both nuclear and mitochondrial DNA
  • Obtained from any human subject readily
  • Used safely and routinely for blood transfusions
  • Low cancerization risk

In addition, other issues are faced here to address.

  • Whether antibody modification delivery specificity is ideal
  • Whether immune cells can be effectively killed in aged/tumor patients
  • The persistence of exosome release from engineered erythrocytes is unknown
  • Whether the immune system can be repaired by delivery of certain molecules/genes
  • Whether delivery errors in tumor therapy can lead to super tumor cells
  • Is it possible to directly deliver a killing substance to kill tumor cells

References

Fedorov, V.D., Themeli, M., and Sadelain, M. (2013). PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med 5, 215ra172.

Morsut, L., Roybal, K.T., Xiong, X., Gordley, R.M., Coyle, S.M., Thomson, M., and Lim, W.A. (2016). Engineering Customized Cell Sensing and Response Behaviors Using Synthetic Notch Receptors. Cell 164, 780-791.

Philipson, B.I., O'Connor, R.S., May, M.J., June, C.H., Albelda, S.M., and Milone, M.C. (2020). 4-1BB costimulation promotes CAR T cell survival through noncanonical NF-kappaB signaling. Sci Signal 13.

Rafiq, S., Hackett, C.S., and Brentjens, R.J. (2020). Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 17, 147-167.

Tan Hong Ji, A., and Campana, D. (2021). Neutralization of Human Cytokines with Membrane-Bound Anti-Cytokine Non-Signaling Binders Expressed in Immune Cells (US: NAT UNIV SINGAPORE).

Wang, T., He, L., Jing, J., Lan, T.H., Hong, T., Wang, F., Huang, Y., Ma, G., and Zhou, Y. (2021). Caffeine-Operated Synthetic Modules for Chemogenetic Control of Protein Activities by Life Style. Adv Sci (Weinh) 8, 2002148.

Wu, W., Zhou, Q., Masubuchi, T., Shi, X., Li, H., Xu, X., Huang, M., Meng, L., He, X., Zhu, H., et al. (2020). Multiple Signaling Roles of CD3epsilon and Its Application in CAR-T Cell Therapy. Cell 182, 855-871 e823.