Overview:

The Ambrosia consists of several parts which carries different aspects of our design, including killing component to clear the senescent cells and control element to regulate the activation and shutdown of CAR-T.

Firstly, the clearance of senescent cells is achieved by the engineered chimeric antigen receptors (CAR)-T cells, which specifically target the senescent marker, Dipeptidyl Peptidase 4 (DPP4). Previous studies reveal that DPP4 was selectively expressed on the surface of senescent (Kim et al., 2017), but not proliferating, human diploid fibroblasts, which suggests the elimination of DPP4 highly expressed cells can delay fibrosis and reverse senescence.

Besides, we were concerned about the safety of our product, so we made some designs to control the activation and shutdown of CAR-T cells. We use caffeine-operated synthetic module (COSMO) to control the switch-on of CAR-T. Several negative feedback loops based on the environmental Interlukin-6 (IL-6) were designed to achieve automated stable reversible switch-off of CAR-T to avoid the happening of inflammatory cytokine release syndrome (CRS).

Engineered CAR-T cells target DPP4 to clear senescent cells.

CAR-T is a developing immunotherapy for the treatment of several cancers. This investigational treatment is a sort of gene therapy that reroutes T lymphocytes to eliminate malignant cells (Huang et al., 2020). Its special structure, CARs, are unique receptors that are designed to target a specific antigen to functionally reprogram T lymphocytes. When the CARs recognize to their targets, it will activate and promote the proliferation of T cells and secretion of granzymes and perforins. Then, the target cells will be killed.

Since the killing properties of T cells target all target cells, targeted killing of senescent cells by CAR-T cells is possible. DPP4 was a potential senescent marker which selectively expressed on the surface of senescent, but not proliferating, human diploid fibroblasts. We engineered an anti-DPP4 CAR, containing anti-DPP4 scFv, CD8 transmembrane domain, 4-1BB co-stimulator domain, and CD3-ζ domain. When the engineered CAR-T cells recognize the DPP4 highly expressed senescent cells, the downstream 4-1BB and CD3-ζ will be activated to promote the proliferation of T cells and secretion of granzymes and perforins to achieve the goal of killing the senescent cells (FIG. 1).


Figure 1. The overview of anti-DPP4 CAR-T cells clearing senescent cells.

The experimental design is as follows:



Inducible switches regulate the activation of CAR-T cells

CRS is the most common side effects of CAR-T immunotherapy, which occurs when immune cells release too many cytokines into the environment. This will cause the damage of human body. Previous studies have shown that IL-6 is the main cause of CRS (Frey and Porter, 2019). Therefore, we design two switches to control the activation and shutdown of CAR-T cells.

The first one is caffeine-operated synthetic module (COSMO). COSMO enables chemogenetic manipulation of biological processes by caffeine and it was used in our design as a robust chemically induced dimerization system to control the activation of CAR-T cells (Wang et al., 2020). It was inserted between 4-1BB co-stimulator domain and CD3-ζ domain. In the absence of caffeine in the environment, when engineered CAR-T cells bind to their respective targets, the CD3- domain cannot be activated due to the blockage of COSMO, and the CAR T cells cannot fully activate, preventing them from carrying out their normal function of killing senescent target cells. However, if the caffeine is presented in the environment, the two engineered CARs can dimerize under the action of COSMO, resulting in the activation of downstream CD3 signaling pathway. Thus, CAR-T cells could be fully activated and perform normal functions (FIG.2). This is a human-controlled CAR-T cell turn-on via COSMO.


Figure 2. The structure of caffeine inducible CAR and its mechanism to control the activation of CAR-T cells.

IL-6 based negative feedback loop to shutdown the CAR-T cells

Several negative feedback loops based on the environmental concentration of IL-6 was designed to regulate the shutdown of CAR-T cells.

Several negative feedback loops based on the environmental concentration of IL-6 was designed to regulate the shutdown of CAR-T cells. Firstly, we designed an anti-IL-6 synthetic Notch receptor (SynNotch). The anti-IL-6 scFv was connected to the Notch regulatory domain which followed by the recombinant transcriptional factor, Gal4-KRAB. When the concentration of IL-6 in the environment is higher than a certain threshold, a large amount of IL6 will bind to anti-IL-6 scFv, leading to the conformational change of SynNotch receptor, which is cleaved by intracellular enzymes. Then the downstream Gal4-KRAB transcription factor is released and transferred to the nucleus, where it binds to the UAS-pSV40 promoter in the nucleus. Consequently, the downstream CAR gene expression was inhibited. The second design was quite similar to the first one, except that the anti-IL-6 scFv was replaced by the high affinity IL-6 receptor binding domain (FIG. 3).


Figure 3. The structure of anti-IL-6_scfv-Notch-Gal4KRAB and IL-6R-Notch-Gal4KRAB and their mechanisms of inhibiting the CAR gene expression.

Moreover, we designed a recombinant receptor which combined the IL-6 receptor extracellular binding domain and PD-1 intracellular domain. When the extracellular IL-6 concentration is high enough, it will bind to the IL-6R extracellular domain and activate the intracellular inhibitory receptors signaling modules. Therefore, the downstream signal of CD3-ζ will be inhibited and inhibit the CAR-T cells activity (FIG.4).


Figure 4. The structure of IL-6R-PD-1 and its mechanism of inhibiting the CAR downstream signaling.

To summary, via these three synthetic receptors, we can achieve automated stable reversible switch-off of CAR-T to avoid the further release of cytokines.

Ambrosia T cells

Now, we have offered a complete set of designs to develop our anti-DPP4 CAR T immunotherapy to reverse the senescent phenotype. We have made it to realize some meaningful improvements of our CAR-T cell including the inducible caffeine switches and IL-6 based negative feedback loops, which achieves the controllable switches of CAR-T cells and reduces the happening of CRS.

Therefore, our senolytic agent – Ambrosia T cell is now created (FIG. 5). After integrated and transformed the required parts (anti-DPP4 CAR, COSMO, and IL-6 based feedback loops) into the T lymphocytes, an engineered CAR-T cell targets to senescent cells was born.


Figure 5. The overall structure of the constructed Ambrosia-T cell.

EEK design for future

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

Frey, N., Porter, D., 2019. Cytokine Release Syndrome with Chimeric Antigen Receptor T Cell Therapy. Biology of Blood and Marrow Transplantation 25, e123–e127. https://doi.org/10.1016/j.bbmt.2018.12.756

Huang, R., Li, X., He, Y., Zhu, W., Gao, Lei, Liu, Y., Gao, Li, Wen, Q., Zhong, J.F., Zhang, C., Zhang, X., 2020. Recent advances in CAR-T cell engineering. J Hematol Oncol 13, 86. https://doi.org/10.1186/s13045-020-00910-5

Kim, K.M., Noh, J.H., Bodogai, M., Martindale, J.L., Yang, X., Indig, F.E., Basu, S.K., Ohnuma, K., Morimoto, C., Johnson, P.F., Biragyn, A., Abdelmohsen, K., Gorospe, M., 2017. Identification of senescent cell surface targetable protein DPP4. Genes Dev 31, 1529–1534. https://doi.org/10.1101/gad.302570.117

Wang, T., He, L., Jing, J., Lan, T., Hong, T., Wang, F., Huang, Y., Ma, G., Zhou, Y., 2020. Caffeine‐Operated Synthetic Modules for Chemogenetic Control of Protein Activities by Life Style. Adv Sci (Weinh) 8, 2002148. https://doi.org/10.1002/advs.202002148