Project Description

An introduction to our project.

Foundational Ideas

Abstract: Designing a novel targeted B-cell lymphoma therapeutic gene delivery platform using AAVP vectors

onCAP is a novel, phage-based cancer gene therapy using adeno-associated virus phage (AAVP) vectors to treat B-cell lymphoma. We constructed our phage combining two modified fd-tet plasmids, fMCS and f3-55nm, as well as an expression cassette engineered from the pAAV plasmid to include granzyme B and TNF-α. This plasmid encodes for a phage displaying anti-CD22 short chain variable fragment on its viral coat, which will lead to the release of the plasmid into mammalian cells. Upon plasmid release in human cancer cells, the proteins granzyme B and TNF-α will be expressed to induce cell death. We hypothesize that an AAVP therapeutic will show high specificity for cancerous B cells with limited off-target effects, because our target marker is primarily expressed on B cells and AAVPs are generally well-tolerated by the immune system. Our goal is to create a proof-of-concept demonstrating the potential of phage therapy in targeting aggressive B-cell cancers.

Rationale

Given the current limitation of B-cell lymphoma treatments and the promise of phage therapies, we hypothesize that adeno-associated virus phages (AAVPs) can provide a new specific, effective, and safe avenue of treatment.

Inspiration

Our idea started from the protocol for designing AAVPs by Hajitou et al. (Hajitou, A., Rangel, R., Trepel, M. et al. Design and construction of targeted AAVP vectors for mammalian cell transduction. Nat Protoc 2, 523–531 (2007). https://doi.org/10.1038/nprot.2007.51), from which we based our initial design and construction of our plasmid. We were fortunate to get into contact with Dr. George P. Smith, who developed the phage display method. He provided us with more current plasmids for AAVPs and helped us modify the protocol to display our targeting anti-CD22 scFv. Building from this foundation, onCAP is a novel application of AAVPs to the field of B-cell lymphoma therapeutic research.

Background

Cancer is the leading cause of death (1), however, current treatments leave the patients to suffer major side effects. Coupled with the lack of information pertaining to drug effects, alternative treatments and appropriate follow up care (2), the side effects of treatment are an unfortunate but necessary aspect of the harsh reality in cancer treatment, one that often severely limit day-to-day ability and quality of life for the patients (2). As such, our team is developing a novel cancer gene therapy using AAVP vectors, adeno-associated virus phage vectors (AAVP) to target B cell lymphoma.

Although molecular and immuno-based therapies reduce the effects that cancer treatment has on patients’ livelihoods, they possess issues of their own. Most commonly, recognition, penetration and general delivery of the drugs to the tumor sites is often subject to a multitude of forces from the body’s immune system and the tumor sites themselves (3). This proves as a barrier for these drugs to be effective on their own, requiring a multitude of treatments and their accompanying side effects to be employed on the patient.

Why are we targeting B-cell Lymphoma?

B cell lymphoma is the cancer of immune cells known as B lymphocytes or B cells. It has been estimated that 80,470 people in the United States will be diagnosed with non-hodgkin’s lymphoma (NHL) in 2022 (4). It is also known that B cell lymphomas make up around 85% of all NHLs (5), with diffuse large B cell lymphoma (DLBCL) being the most common, representing 25% - 30% of all NHLs (6). As such, our therapeutic targets B cell lymphomas with an emphasis on DLBCL. Similar to other cancers, if left untreated B cell lymphoma is fatal.

At a cellular level, this type of lymphoma compromises the normal function of these cells. As a result, B cell lymphoma patients have lowered immunity and higher susceptibility for infection. In physical manifestation, patients commonly experience fevers, night sweats, and swelling of the lymph nodes (7). Tumors are often found in the lymph nodes, and have the ability to metastasize anywhere in the body.

Under regular expression, B cells produce antibodies that can target antigens found on foreign pathogens. One specific method is by tagging these foreign antigens, allowing other immune cells to recognize and phagocytose the intruder. Each pathogen have multiple specific antigens, requiring antibodies with diverse binding for a functioning immune system (8). V(D)J recombination of the V (variable), D (diverse), and J (joining) gene segments in developing B cells is the process that rearranges antibody coding regions of the DNA, resulting in a unique binding site on the antibody protein. This process involves many breaks and ligations of the DNA sequence to produce a random combination of the V, D, and J gene segments for conversion into RNA. This unique sequence is later to be translated into the antibody protein that will be displayed on the surface of the immature B cell. The random selection of these V, D, and J segments is facilitated by the recombination activating genes (RAGs) (9).

In the case of B cell lymphoma, genetic mutations result in improperly functioning B cells. Due to the many breaks and ligations involved in V(D)J recombination, B cells are very susceptible to errors. Abnormal activity of RAGs could possibly result in inactivation of tumor suppressor genes, which compromises the cell’s ability to halt/slow down proliferation. Alternatively, malfunction of RAGs could also lead to activation of oncogenes or the creation of chimeric oncogenes; genes that cause highly accelerated and uncontrolled cell proliferation. Malfunction of RAGs has been shown to be a key contributor to the development of many human and mice lymphomas (10).

Current therapies & limitations to B cell lymphoma

Current therapies for B cell lymphoma follow those of other cancers including but not limited to chemotherapy, radiation, immunotherapy and an assortment of drugs and treatment cycles (11).

CAR-T cell therapy

A newer and rising immunotherapy called CAR-T cell treatment has gained popularity as it involves genetically modifying the patient's own highly specialized immune cells, T cells, to recognize and attack the cancer in their bodies (12). However, with 40-50% of patients reaching lasting immunity, clinical data overlooks patients who relapse (13). The main areas of concern in CAR-T cell treatment are tumor intrinsic factors, host-related factors and its inadequacy (13).

Tumour intrinsic factors refers to the changes of epitope expression, commonly found on the surface of the cells. As antibody binding to these epitopes is crucial for CAR-T treatment success, changes in this expression affect the multiple mechanisms and ultimate failure of the treatment. Similarly, the microenvironments that are crucial for CAR-T cells as they proliferate. It is possible for the increase of these cells in the microenvironment to cause inhibitory signals around them, resulting in suppression of CAR-T cells (13). Host-related factors refers to the patient's baseline characteristics, such that there is impact to the patient’s capacity to respond to the treatment.

CAR-T cell therapy is commonly followed by chemotherapy, because CAR-T and other immunotherapies are not adequate enough to treat B cell lymphoma. When chemotherapy is combined with CAR-T cell therapy, T cells with in the patient’s body may be deleted or destroyed affecting the CAR-T treatment efficacy. The general inadequacies of this treatment include the CAR-T manufacturing of the cells, changes in tumour environment, previous treatments, effects of the neighboring cells can all result in CAR-T cell exhaustion. This is when the CAR-T cells are unable to maintain previous levels of proliferation or their counterparts. Other inadequacies include the translational abilities and profile of the cells seeing as how many patients express higher levels of immunity related genes (13). The majority of the clinical research done on CAR-T cell therapy is done on successful patients, where unsuccessful patients must revert back to more common but much more harsh treatments.

Common Treatments

Meanwhile, there are major undesirable and harsh side effects to common cancer therapies for B cell lymphoma. One immune chemotherapy routine that is commonly used for B cell lymphoma is CHOP (14) and acts by (12) killing fast dividing cells such as cancerous B cells through various drugs. While this treatment has been utilized since the early 1900s (15), and can have 60-70% progression-free complete remission (16). The side effects render the treatment as a life-altering method. The patient's quality of life decreases with the progression of chemotherapy due to hair loss, nausea, mouth sores, weaker immune systems and more (15). Alternatively radiation therapy, more commonly used for treatment in early stage lymphoma, places patients at risk of experiencing similar major side effects in addition to its own set of side effects (14).

Immunotherapy, where the patient's immune system is boosted with naturally or using manmade agents to aid in phagocytosis often needs to be reevaluated on the specific of the agents and their surface profiles as the treatment process takes place (12). Predictions on success, delivery or reaction from the system are difficult to estimate due to unique immune profiles. Additional agents and/or reaction catalysts may be recruited to counter suppressive cells from the tumour site, adding on to the major side effects the patient experiences alongside increasing the risk of infection (11). While other therapies for B cell lymphoma are in use, all come with similar and unique side effects that affect the livelihood of the patient, and affect the efficiency and success of the treatments such that pursuing specific treatments are met with hesitation (11).

As treatment research progresses, coupled with the rise of CAR-T cell treatment, molecular-based treatments are being developed that have shown to have have higher specificity for cancer targets and reduced side effects on healthy cells (3,17). However, these therapies are not currently efficient enough to be the sole treatment due to difficulty of drug delivery and recognition of tumor sites (12).

AAVP in onCAP: why we chose it

Bringing attention to onCAP, we use adeno-associated virus phage vectors (AAVP) as our novel cancer gene therapeutic delivery platform. AAVPs contain single-stranded DNA with up to 5 kb pairs of space for the insertion of therapeutic genes (18). These are eukaryotic viruses created by combining adenoviruses and bacteriophages. Normally bacteriophages are only able to infect bacteria, as such AAVPs typically co-infect with adenoviruses to permit entry of the virus into mammalian hosts. However, genetic and chemical modifications allow them to infect human cells and transduce therapeutic DNA cassettes for gene therapy in human cells (19, 20, 21).

Compared to previous vectors, AAVPs are advantageous due to controlled expression as a result of replication inability. Their capsids initiate weaker immune responses than other therapeutic phages, and their simple structure allows for greater efficiency during infection (18, 22).

Like any phage vector, AAVPs meet the requirements of infection through target cell entry through receptor binding, lysosomal breakdown evasion in the intracellular environment, and the transport of genetic information to the host cell nucleus (23, 24). Current limitations of AAVP use include their susceptibility to proteasomal degradation (25). As such, the AAVP may have to be combined with proteasome-inhibitors for effective anticancer treatment.

In brief, this vector should allow for target recognition and delivery while preventing grand immune reactions. Additionally, we should observe infection into the cancer cells and the release of desired genetic information. We hope to use this evector to release an expression cassette that should induce apoptosis of the cancer cell.

Results of previous literature are able to further support the use of AAVPs. Specifically, one article (26) looked at the transduction efficacy of AAVPs to transduce cells in a mice model of human glioblastoma. Their vectors carried either cytotoxic tumor necrosis factor (TNF) transgenes or a theranostic approach consisting of Herpes simplex virus thymidine kinase (HSVtk) followed by ganciclovir (GCV) transgenes. Efficacy of both vectors were comparable to preclinical efficacy.

Therefore, we hypothesize that our AAVP vector will demonstrate high specificity and immunotolerance (27) to deliver therapeutic genes to induce the apoptosis of cancerous cells. We hope our AAVP vector-based therapeutic delivery platform will decrease side effects and efficiency issues of current B cell lymphoma therapies to increase patients’ quality of life while undergoing treatment.

  1. Cancer [Internet]. [cited 2022 Oct 10]. Available from: https://www.who.int/news-room/fact-sheets/detail/cancer
  2. Zebrack B, Kent EE, Keegan THM, Kato I, Smith AW. ‘Cancer Sucks,’ and Other Ponderings by Adolescent and Young Adult Cancer Survivors. J Psychosoc Oncol. 2014;32(1):1–15.
  3. Kausch I, Doehn C. Molecular Therapy. Encyclopedia of Cancer. 2011;:2364-2367.
  4. Key Statistics for Non-Hodgkin Lymphoma [Internet]. [cited 2022 Oct 2]. Available from: https://www.cancer.org/cancer/non-hodgkin-lymphoma/about/key-statistics.html
  5. Epidemiology in B-Cell Malignancies. Spec Rep [Internet]. 2014 Jul 9 [cited 2022 Oct 2];1(1). Available from: https://www.targetedonc.com/view/epidemiology-in-b-cell-malignancies
  6. Padala SA, Kallam A. Diffuse Large B Cell Lymphoma. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 [cited 2022 Oct 2]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK557796/
  7. Jamil A, Mukkamalla SKR. Lymphoma. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 [cited 2022 Aug 30]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK560826/
  8. LeBien TW, Tedder TF. B lymphocytes: how they develop and function. Blood. 2008 Sep 1;112(5):1570–80.
  9. Roth DB. V(D)J Recombination: Mechanism, Errors, and Fidelity. Microbiol Spectr. 2014 Dec;2(6):10.1128/microbiolspec.MDNA3-0041–2014.
  10. Haines BB, Ryu CJ, Chen J. Recombination Activating Genes (RAG) in Lymphoma Development. Cell Cycle. 2006 May;5(9):913–6.
  11. Clayton Boldt P. Why doesn’t immunotherapy work for everyone? [Internet]. MD Anderson Cancer Center. 2020 [cited 10 October 2022]. Available from: https://www.mdanderson.org/cancerwise/why-doesnt-immunotherapy-work-for-everyone.h00-159385101.html
  12. CAR T-Cell Therapy for Lymphoma - Dana-Farber Cancer Institute | Boston, MA [Internet]. Dana-farber.org. [cited 10 October 2022]. Available from: https://www.dana-farber.org/cellular-therapies-program/car-t-cell-therapy/car-t-cell-therapy-for-lymphoma/#:~:text=CAR%20T%2Dcell%20therapy%20is,cells%20to%20attack%20their%20cancer
  13. Bradley S. Why patients with large B-cell lymphoma fail CAR T-cell therapy & how to manage them [Internet]. LymphomaHub. 2019 [cited 10 October 2022]. Available from: https://lymphomahub.com/medical-information/why-patients-with-large-b-cell-lymphoma-fail-car-t-therapy-and-how-to-manage-them
  14. Watson S. Treatments for B-Cell Lymphoma [Internet]. WebMD. 2022 [cited 10 October 2022]. Available from: https://www.webmd.com/cancer/lymphoma/treatment-options
  15. The American Cancer Society. History of Cancer Treatments: Chemotherapy [Internet]. Cancer.org. 2014 [cited 10 October 2022]. Available from: https://www.cancer.org/treatment/understanding-your-diagnosis/history-of-cancer/cancer-treatment-chemo.html
  16. Jakobsen L, Øvlisen A, Severinsen M, Bæch J, Kragholm K, Glimelius I et al. Patients in complete remission after R-CHOP(-like) therapy for diffuse large B-cell lymphoma have limited excess use of health care services in Denmark. Blood Cancer Journal. 2022;12(1).
  17. Methods of Treatment [Internet]. Www1.udel.edu. 2022 [cited 10 October 2022].
  18. Naso MF, Tomkowicz B, Perry WL, Strohl WR. Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs. 2017 Aug 1;31(4):317–34.
  19. Elias A, Kassis H, Elkader SA, Gritsenko N, Nahmad A, Shir H, et al. HK022 bacteriophage Integrase mediated RMCE as a potential tool for human gene therapy. Nucleic Acids Res. 2020 Dec 16;48(22):12804–16.
  20. Łobocka M, Dąbrowska K, Górski A. Engineered Bacteriophage Therapeutics: Rationale, Challenges and Future. BioDrugs. 2021 May 1;35(3):255–80.
  21. Veeranarayanan S, Azam AH, Kiga K, Watanabe S, Cui L. Bacteriophages as Solid Tumor Theragnostic Agents. Int J Mol Sci. 2021 Dec 30;23(1):402.
  22. Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019 May;18(5):358–78.
  23. Hajitou A, Rangel R, Trepel M, Soghomonyan S, Gelovani JG, Alauddin MM, et al. Design and construction of targeted AAVP vectors for mammalian cell transduction. Nat Protoc. 2007 Mar 1;2(3):523–31.
  24. Suwan Keittisak, Yata Teerapong, Waramit Sajee, Przystal Justyna M., Stoneham Charlotte A., Bentayebi Kaoutar, et al. Next-generation of targeted AAVP vectors for systemic transgene delivery against cancer. Proc Natl Acad Sci. 2019 Sep 10;116(37):18571–7.
  25. Przystal, J. M. et al. Proteasome inhibition in cancer is associated with enhanced tumor targeting by the adeno-associated virus/phage. Molecular Oncology 7, 55–66 (2012).
  26. Staquicini FI, Smith TL, Tang FHF, Gelovani JG, Giordano RJ, Libutti SK, et al. Targeted AAVP-based therapy in a mouse model of human glioblastoma: a comparison of cytotoxic versus suicide gene delivery strategies. Cancer Gene Ther. 2020 May;27(5):301–10.
  27. Suwan K, Yata T, Waramit S, Przystal JM, Stoneham CA, Bentayebi K, et al. Next-generation of targeted AAVP vectors for systemic transgene delivery against cancer. Proc Natl Acad Sci. 2019 Sep 10;116(37):18571–7.

Project Design

Backbones

Our AAVP is a recombination of three different backbones: f3-55nm, fMCS, and pAAV-GFP . f3-55 and fMCS are derived from fd-tet, a phage cloning vector first developed in 1980 by a team at the University of Missouri including Dr. George P. Smith. (4) The phage itself is a modified tetracycline-resistance carrying filamentous phage of the fd species, which are almost identical genetically and functionally to the more commonly known M13 phage. These phages are defined by their long, filamentous shape; approximately 6nm wide and 900nm long. (1) They infect E.coli cells that contain the F factor by using one of their five pIII coat proteins at one end of the filament to bind to the F-pilus expressed on the surface of the E.coli, causing it to retract into the cell. pIII then interacts with the bacterial TolQRA protein to release the genome into the cytoplasm of the cell. (2) By modifying the pIII coat protein, alternative proteins can be displayed on the N-terminus of pIII, allowing the phage to bind to receptors other than the F-pilus. This technique is commonly associated with phage displays, first done by Dr. George P. Smith in 1985. (3) The other modification required is the ability to insert the gene expression cassette into the plasmid. This can be achieved via the utilization of a multiple cloning site (MCS), a section of the sequence containing various different restriction enzyme sites. These restriction enzyme sites allow for the cleavage and insertions of DNA at unique sites insertion of the expression cassette.

The final backbone is an AAV containing plasmid called pAAV-GFP. This plasmid contains the inverted terminal repeats (ITRs) needed for effective transduction of our cassette into mammalian cells. (5) By inserting the expression cassette into the region bordered by the ITRs, we can remove the entire sequence and insert it into the MCS of our primary backbone.

f3-55nm

f3-55nm is a phage display vector derived from fd-tet. Its distinguishing feature is a 14bp stuffer sequence, bordered by SfiI restriction sites in the pIII coat protein coding region. The stuffer sequence disrupts the pIII reading frame, preventing the formation of infective phages, and allowing for propagation of the plasmid. This stuffer sequence can be removed, and the sequence for a protein can be inserted in its place, restoring the reading frame and causing the creation of infective phages that are able to both bind to the F-pilus of F+ bacteria, and display the inserted protein.

fMCS

fMCS is a phage cloning vector derived from fd-tet. Its distinguishing feature is the inclusion of an MCS in one of the non-coding regions of the sequence. This MCS contains a unique PvuII site, which is used for the insertion of the ITR bordered expression cassette. Another feature of fMCS is its common derivativation from fd-tet with f3-55nm. This allows for the splicing together of their sequences without modifying any of the coding regions. This is done using the BamHI site in the middle of the pIII coding region, and the SacII site near where the MCS is located. This results in a final backbone consisting of approximately half of f3-55nm, and half of fMCS, creating f3-55nm-MCS.

The double stranded DNA we received from Dr. Smith had not yet been sequenced, requiring us to verify it ourselves. The results of our Sanger sequencing results are below, along with the .ab1 files with the results. 7 base pair changes over the reference sequence were found, as well as 1 base pair deletion.4 of the base pair changes were silent mutations, while 3 did change the amino acid coded for. The first was at amino acid 249 in the pII coding region, changing out Glutamic Acid for Lysine.The second was at 87 in the pIV coding region, changing out Isoleucine for Asparagine.The third was at 312 also in the pIV coding region, changing out Valine for Isoleucine. Interestingly, 2 of the silent mutations also occurred in the pIV coding region, in the amino acid directly before the mutated ones. The base pair deletion occurred in the non-coding region between TetA and TetR.

pAAV-GFP

pAAV-GFP is an AAV containing plasmid derived from M13. Its use comes from the inclusion of AAV in its plasmid, which can be modified and removed. This region consists of 2 ITRs bordering a region that includes promoters, enhancers, and other essential sequences for transgene expression. This region also contains the code for the green fluorescent protein. All together, these sequences allow for the effective transcription of the coding genes inside mammalian cells. Essential for our uses is the MCS after this protein, allowing for the insertion of our expression cassette in the transcribed sequence of the region. The ITRs bordering the region each end with a PvuII site, allowing for their removal from the pAAV plasmid, and subsequent insertion into the MCS of the f3-55nm-MCS backbone.

  1. Straus S, Bo H. Filamentous Bacteriophage Proteins and Assembly. Subcellular Biochemistry [Internet]. 2018;:261-279. Available from: https://doi.org/10.1007%2F978-981-10-8456-0_12
  2. Hoffmann-Thoms S, Jakob R, Schmid F. Energetic Communication between Functional Sites of the Gene-3-Protein during Infection by Phage fd. Journal of Molecular Biology [Internet]. 2014;426(8):1711-1722. Available from: https://doi.org/10.1016/j.jmb.2014.01.002
  3. Smith G. Filamentous Fusion Phage: Novel Expression Vectors That Display Cloned Antigens on the Virion Surface. Science [Internet]. 1985;228(4705):1315-1317. Available from: https://www.science.org/doi/10.1126/science.4001944
  4. Zacher A, Stock C, Golden J, Smith G. A new filamentous phage cloning vector: fd-tet. Gene [Internet]. 1980;9(1-2):127-140. Available from: https://www.sciencedirect.com/science/article/abs/pii/0378111980901717?via%3Dihub
  5. Wilmott P, Lisowski L, Alexander I, Logan G. A User's Guide to the Inverted Terminal Repeats of Adeno-Associated Virus. Human Gene Therapy Methods [Internet]. 2019;30(6):206-213. Available from: https://www.liebertpub.com/doi/full/10.1089/hgtb.2019.276

scfv-CD22 targeting sequence

How it works

Our anti-CD22 scFv (BBa_K4415001) would specifically recognize and bind to anti-CD22, a receptor expressed in B cells. Therefore, the anti-CD22 scFv allows the AAVP to bind to cancerous B cells, and through constitutive CD22 clathrin-mediated endocytosis allows for internalization of the AAVP vector and subsequent expression of the transgene cassette.

Why we chose CD22 as a biomarker

CD22 is uniquely expressed in B lymphocytes, and is expressed in higher concentrations in mature B lymphocytes than in precursor B cells (1). It is strongly expressed in the marginal zone, mantle, and follicular B cells. In cancerous B-cells, CD22 is overexpressed, hence, it is characteristic of B-cell-related blood cancers such as B-cell acute lymphoblastic leukemia (B-ALL) and Diffuse Large B-Cell Lymphoma (DLBCL), where it is used as a target. Constitutive CD22 clathrin-mediated endocytosis enables the targeted intracellular delivery of immunotoxins to treat B-cell-related blood cancers such as leukemia, and autoimmune diseases (2).

The feasibility and efficacy of CD22 targeting have been demonstrated. Previous CAR T cell treatments for B cell malignancies have shown clinical success by targeting CD22 (3). Furthermore, anti-CD22 antibody-drug conjugates have had some clinical success within the past decade, including a phase-two trial on pertuzumab vedotin (pina), which combined the microtubule inhibitor (monomethyl auristatin E; MMAE) conjugated via a protease-cleavable peptide linker to monoclonal antibodies targeting CD22. Of the 42 patients with DLBCL who received R-pina, 25 (60%, 95% CI 43–74) experienced an objective response and 11 (26%, 95% CI 14–42) underwent a complete response (3). Preceding this finding, Pfeifer et al.’s study on antibody-drug conjugates in different molecular DLBCL subtypes found that anti-CD22-MMAE was highly active and induced cell death in the vast majority of activated B-cell-like (ABC) and germinal center B-cell-like (GCB) DLBCL cell lines (4).

Why we chose to use a scFv

We propose the use of a short-chain variable fragment (scFv) to recognize the target. scFvs are produced from phage display and can be genetically fused to marker proteins. Following fusion, these proteins will have both antigen-binding capacity and marker activity allowing for immunodetection of biological agents (5). Thus, anti-CD22 scFvs will be displayed on the AAVPs for selective phage binding to the cancer target. The phage vector should then be internalized intracellularly given the intrinsic endocytic capability of CD22.

  1. Shah NN, Sokol L. Targeting CD22 for the treatment of B-cell malignancies [Internet]. ImmunoTargets and therapy. Dove; 2021 [cited 2022 Jun 13]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8275043/#cit0004
  2. Ereño-Orbea J, Sicard T, Cui H, Mazhab-Jafari MT, Benlekbir S, Guarné A, et al. Molecular basis of human CD22 function and therapeutic targeting [Internet]. Nature News. Nature Publishing Group; 2017 [cited 2022 Jun 13]. Available from: https://www.nature.com/articles/s41467-017-00836-6#ref-CR20
  3. Ponterio E, De Maria R, Haas TL. Identification of targets to redirect car T cells in glioblastoma and colorectal cancer: An arduous venture [Internet]. Frontiers. Frontiers; 1AD [cited 2022 Jun 13]. Available from: https://www.frontiersin.org/articles/10.3389/fimmu.2020.565631/full#B35
  4. Pfeifer M, Zheng B, Erdmann T, Koeppen H, McCord R, Grau M, et al. Anti-cd22 and anti-CD79B antibody drug conjugates are active in different molecular diffuse large B-cell lymphoma subtypes [Internet]. Nature News. Nature Publishing Group; 2015 [cited 2022 Jun 13]. Available from: https://www.nature.com/articles/leu201548
  5. Ahmad ZA, Yeap SK, Ali AM, Ho WY, Alitheen NBM, Hamid M. ScFv antibody: Principles and clinical application [Internet]. Clinical and Developmental Immunology. Hindawi; 2012 [cited 2022 Jun 13]. Available from: https://www.hindawi.com/journals/jir/2012/980250/

Expression Cassette

The key players in our transgene cassette consists of Granzyme B (GrzB) linked by a 2A linker to the tumor necrosis factor-α (TNFα) and flanked by inverted tandem repeats (ITRs).

TNFα Homo sapiens

Tumor necrosis factor-α (TNF-α) (BBa_K4415007) is a 17 kDa, proinflammatory cytokine produced by activated macrophages, natural killer (NK) cells, and T-lymphocytes (1). Upon acute inflammation, TNF binds to two receptors on the cell membrane, TNFR1, and TNR2, resulting in necrosis or apoptosis (2). TNFR1 primarily signals pro-inflammatory and apoptosis activity whereas TNFR2 signals anti-inflammatory and cell proliferation activity (3). When TNF receptors bind to the ligand, a trimeric complex forms and the receptor undergoes a conformational change (4). As seen in Figure 1, this interaction causes the inhibitory protein SODD to separate from the intracellular death allowing the adaptor protein TRADD to bind to the death domain along with FADD (4,5). This interaction can further initiate a cascade of signaling pathways including the induction of death signaling via cysteine protease caspase 8 activation (6). Several studies have investigated the transgene delivery of TNF-α into cancerous cells to induce apoptosis. As a result, our circuit aims to deliver TNF-α into target cells to initiate apoptotic mechanisms.

Granzyme B Isoform 1

Granzyme B (GrzB) (BBa_K4415002) is a serine protease that is produced and secreted by immune, non-immune, and tumor cells (7). Activated CD8 and CD4 T cells as well as NK and NKT cells GrzB upon infections and inflammation (8). GrzB exhibits multifunctional pro-inflammatory activity by mediating cellular apoptosis through cleavage of caspases, extracellular matrix (ECM) components, cytokines, and cell receptors (7). Similarly, the granule exocytosis pathway plays a crucial role in controlling tumor development and progression (9). As seen in Figure 2, upon interaction with perforin, a glycoprotein located in the granules of NK cells and cytotoxic T lymphocytes (CTLs), the granule exocytosis pathway transports granzymes to the cytosol which leads to a series of events most notably, the cleavage of substrates necessary for apoptosis (5,10,11). Our circuit leverages the transgene delivery of GrzB into cancerous cells through receptor binding to mediate apoptosis in lymphoma cells.

Human eGFP

The eGFP (BBa_K4415005) is an enhanced green fluorescent protein sequence derived from pAAV-GFP for localizing proteins and monitoring cells (12). eGFP was developed from wild-type GFP to improve expression in mammalian systems and increase fluorescence production (13). We will use eGFP in our circuit to identify cells that translated the transfected cassette.

T2A linker

The T2A linker sequence (BBa_K1537017) expresses a self-cleaving oligopeptide and is situated between TNF-α and GrzB proteins. Upon translation, this self-cleaving oligopeptide induces the ribosome to skip the synthesis of a peptide bond at the C-terminus of a 2A element (14). This leads to a self-cleaving mechanism between the TNF-a and GrzB peptides. In our circuit, we will leverage the 2A peptide interaction to effectively secrete GrzB and TNF-α.

E2A linker

The E2A linker sequence (BBa_K4415003), which comes from equine rhinitis A virus 2A, expresses a self cleaving oligopeptide and is situated between GrzB and eGFP (15). Upon translation, this self-cleaving oligopeptide induces the ribosome to skip the synthesis of a peptide bond at the C-terminus of a 2A element (14). This leads to a self-cleaving mechanism between the GrzB and eGFP which we will leverage in our circuit.

Kozak Sequence

The kozak sequence (BBa_K4415004) is a crucial component required for initiation of translation in eukaryotic mRNA (16). This sequence contains base pairs both before and after the start codon. In our circuit this sequence is required for the expression of the transgene cassette.

TNF-a expression pathway to induce apoptosis. Figure was created using Biorender adapted from references (4,5).

Granzyme-b performin cytoxic pathway to induce apoptosis. Figure was created using Biorender adapted from references 5,10,11.

  1. S; PNP. Tumor necrosis factor-α signaling in macrophages [Internet]. Critical reviews in eukaryotic gene expression. U.S. National Library of Medicine; [cited 2022May10]. Available from: https://pubmed.ncbi.nlm.nih.gov/21133840/
  2. Idriss H, Naismith J. TNFΑ and the TNF receptor superfamily: Structure‐function relationship(s): Semantic scholar [Internet]. undefined. 1970 [cited 2022May12]. Available from: https://www.semanticscholar.org/paper/TNF%CE%B1-and-the-TNF-receptor-superfamily%3A-Idriss-Naismith/926cad1f1568bc7019bcf97a7c64c652fad364c3
  3. D; HRS. TNF-mediated homeostatic synaptic plasticity: From in vitro to in vivo models [Internet]. Frontiers in cellular neuroscience. U.S. National Library of Medicine; [cited 2022May12]. Available from: https://pubmed.ncbi.nlm.nih.gov/33192311/
  4. Wajant, H., pfizenmaier, K. and scheurich, P. (2003) tumor necrosis factor signaling. cell death and differentiation, 10, 45-56. - references - scientific research publishing. [cited 2022May11]. Available from: https://www.scirp.org/reference/ReferencesPapers.aspx?ReferenceID=2025414
  5. Biorender [Internet]. BioRender. [cited 2022May2]. Available from: https://biorender.com/
  6. DV; CGG. TNF-R1 signaling: A beautiful pathway [Internet]. Science (New York, N.Y.). U.S. National Library of Medicine; [cited 2022May10]. Available from: https://pubmed.ncbi.nlm.nih.gov/12040173/
  7. DE; KFCJ. Delivery and therapeutic potential of human granzyme B [Internet]. Immunological reviews. U.S. National Library of Medicine; [cited 2022May10]. Available from: https://pubmed.ncbi.nlm.nih.gov/20536562/
  8. Lin L;Couturier J;Yu X;Medina MA;Kozinetz CA;Lewis DE; Granzyme B secretion by human memory CD4 T cells is less strictly regulated compared to memory CD8 T cells [Internet]. BMC immunology. U.S. National Library of Medicine; [cited 2022May12]. Available from: https://pubmed.ncbi.nlm.nih.gov/25245659/
  9. Pardo J;Aguilo JI;Anel A;Martin P;Joeckel L;Borner C;Wallich R;Müllbacher A;Froelich CJ;Simon MM; The biology of cytotoxic cell granule exocytosis pathway: Granzymes have evolved to induce cell death and inflammation [Internet]. Microbes and infection. U.S. National Library of Medicine; [cited 2022 May 6]. Available from: https://pubmed.ncbi.nlm.nih.gov/19249384/
  10. Antibody-granzyme B fusion protein [Internet]. Creative Biolabs. [cited 2022Jun6]. Available from: https://www.creative-biolabs.com/adc/antibody-granzyme-b-fusion-protein.htm
  11. Cao X;Cai SF;Fehniger TA;Song J;Collins LI;Piwnica-Worms DR;Ley TJ; Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance [Internet]. Immunity. U.S. National Library of Medicine; [cited 2022 May 6]. Available from: https://pubmed.ncbi.nlm.nih.gov/17919943/
  12. Paav-GFP (plasmid #32395) [Internet]. Addgene. [cited 2022Sep29]. Available from: https://www.addgene.org/32395/
  13. Ganini D;Leinisch F;Kumar A;Jiang J;Tokar EJ;Malone CC;Petrovich RM;Mason RP; Fluorescent proteins such as egfp lead to catalytic oxidative stress in cells [Internet]. Redox biology. U.S. National Library of Medicine; [cited 2022Sep29]. Available from: https://pubmed.ncbi.nlm.nih.gov/28334681/
  14. Self-cleaving 2A peptides allow for expression of multiple genes in ... [Internet]. [cited 2022Sep29]. Available from: https://www.biorxiv.org/content/10.1101/2022.03.08.482734v1.full.pdf
  15. Chng J, Wang T, Nian R, Lau A, Hoi KM, Ho SCL, et al. Cleavage efficient 2A peptides for high level monoclonal antibody expression in Cho Cells [Internet]. mAbs. Taylor & Francis; 2015 [cited 2022Sep29]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4622431/
  16. Kozak sequence [Internet]. GeneMedi. [cited 2022Sep29]. Available from: https://www.genemedi.net/i/kozak-sequence