GET THE APP

CAR-T: The Current and the Future
Pancreatic Disorders & Therapy

Pancreatic Disorders & Therapy
Open Access

ISSN: 2165-7092

+44 1478 350008

Mini Review - (2015) Volume 5, Issue 2

CAR-T: The Current and the Future

Kunal Mishra, Shu-Feng Zhou and Jiazhi Sun*
Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, USA
*Corresponding Author: Jiazhi Sun, Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, USA, Tel: +813-974-7468, Fax: 813-905-9885 Email:

Abstract

Chimeric antigen receptors (CARs) are recombinant receptors that are expressed on autologous T-cells. CAR-T usage has grown in recent years as a way to combat hematological and solid tumors. The purpose of this review is to describe new studies on CAR-T treatment along with our lab’s ideas on potential uses and pitfalls that could be investigated. The usage of TREG and antiCLTA4 as means for regulating CAR-T will also be looked at along with OX40, CD137 and CD27 receptors as means for increasing the efficacy of the treatment overall. In addition the usage of iCasp9 as a ‘suicide switch’ for the CAR-T treatment and the potential for a 4th generation CAR will be touched upon. Then the review will lead into talks about how such a treatment could be of potential use in treating solid and hematological cancer.

<

Keywords: Immunotherapy, Chimeric antigen receptors, CAR-T,

The Current

Immunotherapy research has long looked for a way to harness an individual’s own immune system to fight off tumors and cancers. Many ideas have made tried to make use of natural killer cells or B-cells, however research has not moved past conceptual thinking. However current research on chimeric antigen receptor T cells may change this. With several clinical trials already in place, CAR-T research is a growing field, with physicians and scientists looking for its expansion to a variety of cancers with cytokine storm cautious [1]. The idea behind CAR-T is the modified T-Cell. The patient’s own T-cells are harvested and then genetically modified to have a receptor for a particular antigen that a particular tumor cell may display. The particular antigen that has been used is CD-19, a common tumor antigen [2]. However without an initiating signal the CAR-T cells would remain naïve within the body. This signal is provided by dendritic cells. DC cells are professional antigen presenting cells that allow for a more directed immune response than B-cells could provide [3]. However DC cells cannot pick up the CD-19 antigen to present to the CAR-T cells because it is masked by the PD1-PDL1 interaction in tumor cells [4]. Naturally, the PD1-PDL1 interaction is used in the body to prevent the immune system from attacking a particular cell; for example, the PD1-PDL1 interaction prevents the mother’s immune system from attacking the fetus’s cells during pregnancy [5]. The tumor cells have harnessed this interaction to mask themselves from the body’s immune system. Breaking this interaction is a key component of CAR-T treatment as once done tumor cells would be vulnerable to the immune system [6]. The CAR-T’s function lies in providing an anti-PD1-anti-PDL1 interaction which would allow the tumor cell to be susceptible to the host’s immune system1 [7-17].

The Future

To combat solid tumors, the CAR-T treatment being designed must not only be specific and effective but also be administered within the window in which the patient is preconditioned [18-21]. New studies have however found that when used in combination with adenovirus Ad5Δ24, CAR-T therapy with anti-PD1/PDL1 and CLTA4 strategy can be more effective [1,19,22]. Ad5Δ24, an oncolytic virus that contains the chemokine RANTES along with IL15 cytokine has shown via the intratumoral release of these two substituents, to not only attract CAR-T cells to the tumor, but also promote their local survival in the immunosuppressive environment [19]. The non-HLA CAR-T target of stromal solid tumor (i.e. ErbB) has been in study [1]. By combining Ad5Δ24 with CAR-T treatment, while the study will need to progress further before clinical trials, it does shed light on the future of CAR-T cell therapy’s role in treating solid tumors.

OX40, CD137 and CD27’s Role in CAR-T

Expression of OX40 will prevent CAR T-cells from dying and can maintain their immune response onwards to a memory response [23,24]. OX40 is required to further augment CD4 and CD8 priming. OX40 receptors can be promoted to be expressed in T cells first by the TCR engagement. The pitfall with OX40 however is the association of increasing cytokine production. OX40 binding has been shown to initiate cytokine storms in certain infections and will presumably add to the cytokine storm already made via CAR-T therapy [23]. CD137 and CD27 both TNF receptors, are expressed in the same fashion as OX40, generally within 24-48 hours after TCR engagement [25]. They also make use of the DC cells to promote the expression of their required ligand. CD137 is expressed by primarily CD8 cells [7]. CD137 acts as a co-stimulant that enhances T-cell proliferation and has been shown to enhance immune activity to eliminate tumors [26,27]. By allowing our CAR constructs to express CD137, we will increase the efficacy of the treatment. CD27 works in a similar fashion and is also a TNF receptor; however this works more towards maintaining T-cell immunity [23]. It also helps regulate B and DC cell activation, which can allow for further DC cell activation past the initial stimulation [23,24].

Conclusions: The Ideal CAR Model

CAR-T treatment is fascinating prospect in the field of immunotherapy due to its potential uses. New research is being conducted as to improving current CAR constructs and minimizing on target, off tumor toxicities. Based on all the current research we can present an ideal CAR model (Figure 1). This model will include an anti-PD1 region, anti-CD19, and OX40, CD137 and CD27 co-stimulatory domains. In addition the CAR-T cell will have Treg and anti CLTA4 to regulate the CAR-T cell numbers and maintain a basal level. This CAR construct will also incorporate the iCasp9/AP1903 suicide gene technology as a self-withdrawal mechanism [28]. This ideal CAR construct would be used as a treatment that will be administered via intratumoral injection to maximize the efficacy and minimize any side effects. New research is being conducted on a similar kind of model by Zhu et al and the results of this study will help advance our knowledge about CAR-T treatment and better mold the ideal CAR construct [29,30].

pancreatic-disorders-therapy-cross-talk

Figure 1: Figure above is a summary figure of our CAR-T model. The figure shows the CAR-T cell with the CD137, anti-CLTA4, CD27,iCasp9, OX40, anti-PD1 and CD19 receptors. The varying receptors all are contributing in one way or another to overall efficacy of the treatment. The figure shows the CAR-T cell binding to the melanoma site via the CD-19 antigen, and also shows the breaking of the PD1-PDL1 interaction. Additionally the Tregulatory cell (TREG) is shown to be interacting and regulating with the CAR-T cell via cytokines which act as signals for cross talk between.

References

  1. Gill S, June CH (2015) Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies. Immunol Rev 263: 68-89.
  2. Shi H, Sun M, Liu L, Wang Z (2014) Chimeric antigen receptor for adoptive immunotherapy of cancer: latest research and future prospects. Mol Cancer 13: 219.
  3. Wieder E (2003). Dendritic cells: a basic review. International Society for Cellular Therapy.
  4. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, et al. (2013) Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res 19: 5636-5646.
  5. Guleria I, Khosroshahi A, Ansari MJ, Habicht A, Azuma M, et al. (2005) A critical role for the programmed death ligand 1 in fetomaternal tolerance. J Exp Med 202: 231-237.
  6. John LB, Kershaw MH, Darcy PK (2013) Blockade of PD-1 immunosuppression boosts CAR T-cell therapy. Oncoimmunology 2: e26286.
  7. Kalos M, Levine BL, Porter DL, Katz S, Grupp, et al. (2011) T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science translational medicine, 3: 93.
  8. Monnier P, Vigouroux R, Tassew N (2013) In Vivo Applications of Single Chain Fv (Variable Domain) (scFv) Fragments. Antibodies 2: 193-208.
  9. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, et al. (2013) Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368: 1509-1518.
  10. Geurts AM, Yang Y, Clark KJ, Liu G, Cui Z, et al. (2003) Gene transfer into genomes of human cells by the sleeping beauty transposon system. Mol Ther 8: 108-117.
  11. Hackett PB, Ekker SC, Largaespada DA, McIvor RS (2005) Sleeping beauty transposon-mediated gene therapy for prolonged expression. Adv Genet 54: 189-232.
  12. Scholler J, Bushman FD, Hege KM, PlesaG, Hwang W, et al. (2012). Decade-Long Safety and Function of Retroviral-Modified Chimeric Antigen Receptor T Cells. Science Translational Medicine, 4: 132-53.
  13. O’Keefe EP(2013). Nucleic Acid Delivery: Lentiviral and Retroviral Vectors. Materials and Methods, 3: 1-15.
  14. Fitz M (2013) Chimeric Antigen Receptor (CAR)-Engineered T Cells for Cancer Therapy.
  15. Gargett T, Brown MP (2014) The inducible caspase-9 suicide gene system as a "safety switch" to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front Pharmacol 5: 235.
  16. Gilham DE, Debets R, Pule M, Hawkins RE, Abken H (2012) CAR-T cells and solid tumors: tuning T cells to challenge an inveterate foe. Trends Mol Med 18: 377-384.
  17. Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, et al. (2013) Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther 21: 904-912.
  18. Textor A, Listopad JJ, Wührmann LL, Perez C, Kruschinski A, et al. (2014) Efficacy of CAR T-cell therapy in large tumors relies upon stromal targeting by IFNγ. Cancer Res 74: 6796-6805.
  19. Nishio N, Diaconu I, Liu H, Cerullo V, Caruana I, et al. (2014) Armed Oncolytic Virus Enhances Immune Functions of Chimeric Antigen Receptor–Modified T Cells in Solid Tumors. Cancer research, 74: 5195-5205.
  20. Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, et al. (2014) Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res.
  21. Okada M, Hibino S, Someya K, Yoshmura A (2014) Regulation of regulatory T cells: epigenetics and plasticity. Adv Immunol 124: 249-273.
  22. Lopez-Pastrana J, Shao Y, Chernaya V, Wang , Yang XF (2015) Epigenetic enzymes are the therapeutic targets for CD4(+)CD25(+/high)Foxp3(+) regulatory T cells. Transl Res 165: 221-240.
  23. Croft M (2003) Costimulation of T cells by OX40, 4-1BB, and CD27. Cytokine Growth Factor Rev 14: 265-273.
  24. Redmond W, Ruby CE, Weinberg AD (2009) The role of OX40-mediated co-stimulation in T-cell activation and survival. Crit Rev Immunol 29: 187-201.
  25. Prasad KV, Ao Z, Yoon Y, Wu MX, Rizk M, et al. (1997) CD27, a member of the tumor necrosis factor receptor family, induces apoptosis and binds to Siva, a proapoptotic protein. Proc Natl Acad Sci U S A 94: 6346-6351.
  26. Milone MC, FishJD, Carpenito C, CarrollRG, BinderGK, et al. (2009) Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Molecular Therapy, 17: 1453-1464.
  27. Song DG, Ye Q, Carpenito C, Poussin M, Wang LP, et al. (2011) In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4-1BB). Cancer research, 71: 4617-4627.
  28. Bonini C, Bondanza A, Perna SK, Kaneko S, Traversari C, et al. (2007) The suicide gene therapy challenge: how to improve a successful gene therapy approach. Mol Ther 15: 1248-1252.
  29. Zhu J (2015). Evaluation of 4th Generation Safety-designed CAR T Cells Targeting High-risk and Refractory B Cell Lymphomas.
  30. Kakarla S, Gottschalk S (2014) CAR T cells for solid tumors: armed and ready to go? Cancer J 20: 151-155.
Citation: Mishra K, Zhou S-F, Sun J (2015) CAR-T: The Current and the Future. Pancreat Disord Ther 5:154.

Copyright: © 2015 Kunal Mishra, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top