Coding
anti-CD19

Part:BBa_K2549005

Designed by: Rongrong Du   Group: iGEM18_Fudan   (2018-10-02)


anti-CD19

Anti-CD19 (αCD19) is a CD19 scFv. Its heavy-chain variable region (αCD19a) and light-chain variable region (αCD19b) are fused using a glycine-rich peptide linker (3 repeats of GGGGS, or G4S for short)[1]. It was used as the extracellular domain of the SynNotch, thus accomplishing the contact-dependent signal input against CD19 (surCD19 was used in our project, Part:BBa_K2549001).

Sequence and Features


Assembly Compatibility:
  • 10
    COMPATIBLE WITH RFC[10]
  • 12
    COMPATIBLE WITH RFC[12]
  • 21
    COMPATIBLE WITH RFC[21]
  • 23
    COMPATIBLE WITH RFC[23]
  • 25
    COMPATIBLE WITH RFC[25]
  • 1000
    INCOMPATIBLE WITH RFC[1000]
    Illegal BsaI site found at 444


Biology

Clinical significance of CD19

As summarized on wikipedia page[2]: B-lymphocyte antigen CD19, also known as CD19 molecule (Cluster of Differentiation 19), B-Lymphocyte Surface Antigen B4, T Cell Surface Antigen Leu-12 and CVID3 is a transmembrane protein that in humans is encoded by the gene CD19. In humans, CD19 is expressed in all B lineage cells, except for plasma cells, and in follicular dendritic cells. CD19 plays two major roles for B cells: (1) it acts as an adaptor protein to recruit cytoplasmic signaling proteins to the membrane; (2) it works within the CD19/CD21 complex to decrease the threshold for B cell receptor signaling pathways. Due to its presence on all B cells, it is a biomarker for B lymphocyte development, lymphoma diagnosis and can be utilized as a target for leukemia immunotherapies.

CAR-T CD19

CD19-targeted chimeric antigen receptor T cell therapy[3]: Acute lymphoblastic leukemia (ALL) remains difficult to treat, with minimal improvement in more than 2 decades. Adoptive transfer of T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a powerful targeted immunotherapy. Complete remission rates as high as 90% have been reported in children and adults with relapsed and refractory ALL treated with CAR-modified T cells targeting the B-cell–specific antigen CD19. For more details, please check Maude SL et al.

cancer.gov[4]: CAR T cells and TCR T cells are engineered to produce special receptors on their surfaces. They are then expanded in the laboratory and returned to the patient. Image credit: National Cancer Institute.

CAR T cell therapy can cause several worrisome. One of the most frequent is cytokine release syndrome (CRS). Ironically, CRS is considered an “on-target” effect of CAR T cell therapy. Patients experiencing severe CRS all had particularly high levels of IL-6, a cytokine that is secreted by T cells and macrophages in response to inflammation. And, doctors have been using IL-6 blockade to manage IL-6 CRS[5]. Another potential side effect of CAR T cell therapy is a mass die off of B cells, known as B-cell aplasia. CD19 is expressed on normal B cells, which are responsible for producing antibodies that kill pathogens. These normal B cells are also often killed by the infused CAR T cells. To compensate, many patients must receive immunoglobulin therapy, which provides them with the necessary antibodies to fight off infections[6].

α-CD19 works extremely well in Royal KT et al 2016

Please refer the original article for more details[7]. Below is our summary of their article to explain why we focus on CD19.

Royal KT at al stated: SynNotch receptors have a custom ligand binding domain that detects a cell-surface antigen of interest (e.g., scFvs targeted to CD19 or Her2 or nanobodies to GFP), the core regulatory region of Notch that controls proteolysis, and a cytoplasmic orthogonal transcription factor (e.g., Gal4 VP64). The corresponding response elements for the orthogonal transcription factor controlling custom transcriptional programs are also engineered into the T cell.
Royal KT at al stated: CD4+ and CD8+ primary human T cells were engineered with the α-CD19 synNotch Gal4VP64 receptor and 5x Gal4 response elements control- ling the expression of a BFP reporter. Histogram showing selective induction of the BFP reporter in α-CD19 synNotch receptor receiver CD4+ T cells in response to stimulation with sender cells with CD19- or CD19+ K562s.
Royal KT at al stated: CD4+ AND CD8+ primary human T cells were engineered with the α-CD19 nanobody synNotch Gal4VP64 receptors and 5x Gal4 response elements controlling the expression of a BFP reporter. The percentages of synNotch T cells that upregulate the BFP reporter after 24 hr of stimulation with the indicated sender cells is given (n >= 3 for all conditions, error bars, SEM).
Royal KT at al stated: CD4+ T cells were engineered with the α-CD19 synNotch Gal4VP64 receptor and the corresponding response elements controlling the expression of either IL-2, IL-10, IL-12, or combined IL-2/MIP-1a. The cells were co-cultured with target CD19+ K562s or CD19- non-target K562s.
Royal KT at al stated: CD4+ α-CD19 synNotch T cells were engineered to regulate the expression Tbet and thus Th1 fate choice by T cells. The synNotch T cells were co-cultured with target CD19+ or non-target CD19- K562 cells for 11 days to determine if synNotch driven Tbet expression could skew CD4+ T cells to Th1 fate in a CD19- dependent manner.
Royal KT at al stated: (C) Histograms showing the selective expression of Tbet T2A EGFP after 24 hr of CD4+ α-CD19 synNotch T cells with CD19+ K562s (representative of at least three experiments). (D) Two-dimensional dot plots of intracellular stained CD4+ α-CD19 synNotch Gal4VP64 T cells for Tbet and IFNg after 11 days of culture with either CD19+ or CD19- K562s. T cells were stimulated with PMA/Ionomycin for 4 hr prior to staining to drive cytokine production (representative of at least three experiments). (E) The percentage of IFNg+ (Th1) T cells after 11 days of the indicated treatment (n >= 3 for all treatments, error bars, SEM, significance determined by Student’s t test, n.s. p >= 0.05).
Royal KT at al stated: CD4+ T cells were engineered with the α-CD19 synNotch receptor controlling the expression of PD-L1 and IL-10.
Royal KT at al stated: Quantification of the percentage of synNotch T cells that express PD-L1 and intracellular IL-10 after co-culture with CD19+ or CD19- K562s for 24 hr is given. The amount of IL-10 in the supernatant was also determined by ELISA (n = 3; error bars, SEM).
Royal KT at al stated: NSG mice were subcutaneously injected with CD19- non-target K562s and target CD19+ in the left and right flank, respectively. α-CD19 synNotch T cells in control of IL-2 iRES mCherry expression were injected into the mice after tumors were established and tumors were harvested at the indicated time point to determine whether the synNotch T cells had infiltrated the tumor and expression of IL-2 and mCherry reporter was induced.
Royal KT at al stated: Histograms of IL-2 IRES mCherry reporter levels in tumor and spleen infiltrated CD4+ synNotch T cells injected i.v. showing selective expression of the mCherry reporter in target CD19+ tumors (data representative of three replicate mice).


References

  1. Chimeric Antigen Receptor-Modified T Cells in Chronic Lymphoid Leukemia; Chimeric Antigen Receptor-Modified T Cells for Acute Lymphoid Leukemia; Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N Engl J Med, 2016 Mar;374(10):998 PMID: 26962747; DOI: 10.1056/NEJMx160005
  2. https://en.wikipedia.org/wiki/CD19
  3. CD19-targeted chimeric antigen receptor T cell therapy for acute lymphoblastic leukemia. Maude SL, Teachey DT, Porter DL, Grupp SA. Blood, 2015 Jun;125(26):4017-23 PMID: 25999455; DOI: 10.1182/blood-2014-12-580068
  4. https://www.cancer.gov/about-cancer/treatment/research/car-t-cells
  5. https://en.wikipedia.org/wiki/Tocilizumab
  6. https://primaryimmune.org/treatment-information/immunoglobulin-therapy
  7. Engineering T Cells with Customized Therapeutic Response Programs Using Synthetic Notch Receptors. Roybal KT, Williams JZ, Morsut L, ..., McNally KA, Lim WA. Cell, 2016 Oct;167(2):419-432.e16 PMID: 27693353; DOI: 10.1016/j.cell.2016.09.011
[edit]
Categories
//cds
//cds/receptor/antibody
Parameters
None