GET THE APP

CAR-T: Expression, Expansion, and Clinical Applications
Pancreatic Disorders & Therapy

Pancreatic Disorders & Therapy
Open Access

ISSN: 2165-7092

+44 1478 350008

Editorial - (2015) Volume 0, Issue 0

CAR-T: Expression, Expansion, and Clinical Applications

Crystal DuPont, Rehan Muhammad 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

Understanding and treating cancer is one the top priorities in medical research. The use of chimeric antigen receptor T-cells (CAR-T) are increasing in popularity in the research and treatment of cancer due to their ability to utilize the expansion and killing effects of cytotoxic T cells while also having binding specificity through the chimeric antigen receptor (CAR). CAR expression and CAR-T expansion are critical for proper CAR-T functionality. Toxicity has been associated with the treatment of cancers with CAR-T however methods to control and decrease toxicity have been developed. While they first became popular for the treatment of B-cell lymphomas they are expanding to the treatment of other cancers. In this review we will discuss the current methods for CAR-T expansion, expression, and toxicity prevention while also covering current clinical applications of this therapy.

<

Keywords: CAR-T, Clinical Applications, cancer, retroviral vector

Introduction

Chimeric antigen receptor T-cells (CAR-T) are able to utilize the expansion and killing effects of cytotoxic T cells while also having binding specificity through the chimeric antigen receptor (CAR). CAR-T cells avoid issues common to T cell therapy such as the dependence on human leukocyte antigen (HLA) interactions for activation of T cell effector functions. HLA is often down regulated in cancer cells to promote immune system escape, which makes it more difficult for engineered T cells to activate an effector response [1]. The expression of CAR allows the T cell to shed its dependence on HLA mediated activation and elicit an effector response by binding to tumor specific surface proteins. The basic CAR consists of an antigen binding domain which is a single chain variable region antibody fragment (scFV), an extracellular domain, transmembrane domain and intracellular signaling domains. The intracellular domain is typically the CD3 ζ chain that is typically associated with the TCR complex. CAR-T is categorized in generations which increase as co-stimulatory molecules are added to its expression cassette. Co-stimulatory molecules enhance the efficacy and tumor trafficking of the CAR-T cell [2]. Fourth generation CAR-T contains a more specialized cassette of co-stimulatory molecules and other inhibitory molecules to control CAR-T proliferation in order to prevent CAR-T toxicity.

CAR-T Expression

The CAR can be expressed through several different mechanisms including CAR encoding retroviral vector, mRNA transfection, lentiviral vector, transposon activity, and RNA electroporation. Lentiviral vectors transduce with non-dividing cells. These vectors completely and specifically integrate into the host genome with low immunogenicity which leads to permanent transgene expression. Low immunogenicity also endows lentiviral vectors the capacity to deliver larger DNA sequences and reduced susceptibility to silencing [2].

Another popular method for integrating the CAR gene in the T cell genome is the sleeping beauty transposon system. Sleeping Beauty (SB) is a transposon made synthetically and based on sequences of transpositionally inactive elements. SB is a superfamily transposon following a cut-and-paste transpositional reaction. In this reaction the element-encoded transposase interacts with its binding sites in the terminal inverted repeats of the transposon. This then promotes the excision of the element out of its donor site and its subsequent integration into the target DNA [3]. This method has also been adopted for DNA plasmid use for integration. Combining these two vectors yielded CAR-T cells with improved plasmid integration efficiency [4].

CAR-T Expansion

The preferred T cells to undergo expansion are those that are less differentiated. Expansion requires activating signals and appropriate growth environments. Stimulation signals have been summarized in Table 1 in the appendix. Figure 1 outlines the process of CAR-T expression and expansion.

Signal Description
OKT3 (anti-CD3 clone) Can be used with anti-CD28 costimulating antibodies or activating cytokines (e.g. IL-2)
Anti-CD3/CD28 Antibodies targeting activating/stimulatory domains to T cells
Bispecific T cell Engaging antibodies (BiTEs)
IL-2/IL-21 Cytokine known to induce clonal expansion of T Cells
Artificial antigen presenting cells (e.g. Epstein-Barr virus)
(aAPCs) Cells engineered to act as endogenous APCs would actàstimulating expansion of Tcells

Table 1: Methods of CAR-T Cell Expansion [5].

pancreatic-disorders-therapy-Expression-Expansion

Figure 1: CAR-T Expression and Expansion. (A-C) CAR-T and lentiviral vector containing CAR genetic cassette transduction. (D) CAR expression and iCasp9 expression. Activating signals are introduced to induce expansion. (E) Clonal expansion of the CART cells, these cells are then infused back into patient circulation.

Anti-CD3/CD28 antibodies have shown better anti-tumor activity and better persistence following infusion. Ex vivo expansion using an antigen presenting cell (APC) may have a longer culture period but produce a larger amount of T cells capable of in vivo expansion post infusion [5]. A concept that has been noted to assist in T cell expansion is “Cytokine Sinks” [6]. This concept notes that depletion of lymphocytic cells (as seen with chemotherapy treatment) before CAR-T cell infusion facilitates their expansion without having to compete with endogenous lymphocytes for cytokine stimulation [7]. Late co-stimulatory signals from CD137 or OX40 can also aid in CAR-T activation by recruiting TNF family proteins [8]. This represents a different pathway than that of the signals provided by CD28.

After the T cells undergo ex vivo clonal expansion they are infused back into patient circulation where they undergo the process of “trafficking” in which they are directed to their site of action. Trafficking to the site of action can be mediated through overexpression of chemokines or molecules that will direct the CAR-T to its target site. At the appropriate tumor site, CAR-T cells must interact with the appropriate antigen, avoid inhibitory signals in the tumor microenvironment and maintain their effector function until the tumor has been eradicated. To avoid inhibitory signals and maintain their effector activity, CAR-T can be engineered to overexpress pro-survival signals, and infused with exogenous cytokines that promote expansion while avoiding T regulatory cell induction [9]. While treatment with CAR-T has shown promising results they have been shown to induce toxicity in patients through cytokine storm, autoimmune toxicity and macrophage activation syndrome which have resulted in death of participants in clinical trials [10].

CAR-T Toxicity

On tumor off tumor toxicity is a continuous safety concern for targeted therapies and CAR-T cell treatment is not excluded. In addition to off tumor toxicity, CAR-T cells that use the retroviral method of expression run the risk of insertional mutagenesis and oncogenic expansion [5]. Some on-tumor on-target toxicities include: macrophage activation syndrome, cytokine release syndrome, and tumor lysis syndrome (TLS) [9]. TLS involves massive tumor cell lysis that releases large amounts of the tumors’ intracellular contents into systemic circulation. This often leads to hyperkalemia, hyperuricemia, hypophosphatemia, and hypocalcemia [9]. Macrophage activation syndrome denotes the condition where there is an increase in T cell expansion and elevated levels of macrophage activation in vivo. In vivo T cell clonal expansion and response to an antigen leads to a “cytokine storm”. This includes the presence of high levels of cytokines resulting in an elevated immune response (e.g. B cells, NK cells, macrophages, PMNs), inflammation, and tissue damage [3]. IL-6 is commonly elevated during a cytokine storm and at high levels can lead to trans-signaling (activation of cells not expressing IL-6 receptor).

Autoimmune toxicity or “on target, off tumor toxicity” occurs when the CAR-T attacks the correct antigen target but the tissue is non-malignant. The risk of autoimmune toxicity increases after treatment with checkpoint inhibitors. Symptoms range from mild (fever, nausea,) to severe (multiple organ failure, reflex tachycardia, death) [7].

Possible solutions include treatment with corticosteroids upon development of mild symptoms, target specific cytokine receptors in combination with treatment with CAR-T cells, and incorporation of the iCasp9 (inducible caspase 9) suicide gene. Tocilizumab is an anti-IL6R monoclonal antibody developed to help combat toxicity noted with CAR-T treatment of B cell acute lymphoblastic leukemia [2]. It has shown promising results in lowering toxicity without inhibiting the tumor killing effects of the CAR-T cell [2]. The human caspase genes are responsible for inducing cell death through apoptosis. Incorporation of the iCasp9 suicide gene using the CaspaCIDe system includes the iCasp9 and a small inert molecule that chemically induces dimerization (CID) like AP1903 [5]. Including this gene in the CAR-T expression cassette could serve as a safety switch for treatment of some cancers with high risks of CAR-T induced toxicities since only the T cells expressing this gene will undergo apoptosis upon introduction of the CID [5].

Clinical CAR-T

CD19 is the current hot target of CAR-T therapy. CD19 is a B lineage specific protein that regulates B lymphocytes activation and is expressed throughout all stages of B cell differentiation except for the hematopoietic stem cell and plasma cell stage [8]. The cell stage specific expression of CD19 makes it a good target for CAR-T treatment of B-lineage diseases. Although popular, CD19 is not the only focus of clinical trials that are making use of the CAR-T cell.

Epidermal growth factor receptor (EGFR) is another target for CAR-T therapy. EGFR is a member of the epidermal growth factor family of receptor tyrosine kinases (RTKs). There are four members of the EGFR family: ErbB2 (HER2/neu), ErbB3 (HER3), and ErbB4 (HER4). In many cancer cells, activation of EGFR leads to a signaling cascade that activates the oncogene KRAS, which is commonly mutated to a constitutively active form in oncogenic cells. [4]. Cancers involving KRAS mutations include pancreatic, adenocarcinomas, non-small cell lung cancer (NSCLC). Table 2 in the appendix outlines other current clinical trials involving CAR-T cells. Current clinical trials can be viewed on www.clinicaltrials.gov.

CAR-T target* Clinical Trials Cancers** Supplementary Reference
BCMA NCT02215967 Myeloma,Plasma-Cell/ Myeloma-Multiple S21, S22, S23
CEA NCT02416466 Liver Metastasis  
NCT02349724 Lung, colorectal, gastric, breast, and pancreatic  
CD19 + Mesothilin NCT02465983 Pancreatic Cancer  
CD 19 (CD19 clinical trial information can be found in supplementary Table 1)
CD 20 NCT01735604 Hematopoietic/Lymphoid Cancer S24
CD 28 NCT00881920 Lymphoma/ Myeloma/ Leukemia  
CD 123 NCT02159495 AML  
CD 138 NCT01886976 Relapsed and/or Chemotherapy Resistant Multiple Myeloma  
Myeloma, toxicity noted: inflammation [5].
CD171 NCT02311621 Neuroblastoma / Ganglioneuroblastoma  
CD30 NCT02259556 Hodgkin's Lymphoma  
NCT02274584 NCT01316146 NHL
NCT01192464  
   
  Note: Relapsed patients retain high CD30 expression making them good candidates for CAR-T targets[5].
CD33 NCT01864902 Relapsed AML/ Chemotherapy Refractory AML S4
cMET NCT01837602 Metastatic BreastCancer/ Triple Negative Breast Cancer  
ROR1 NCT02194374 CLL/SLL  
EGFR NCT01869166 Advanced malignant gliomas, Advanced EGFR-positive Solid Tumors  
NCT02331693
EGFRvIII NCT01454596 Malignant Glioma/ Glioblastoma/ Brain Cancer S16, S17, S18, S19, S20
GPC3 NCT02395250 Hepatocellular Carcinoma  
GD-2 NCT02107963 Sarcoma/ Osteosarcoma/ Rhabdomyosarcoma/ Neuroblastoma/ Melanoma S1,S2,S3
NCT01822652
NCT01953900
VEGFR2 NCT01218867 Metastatic cancer, metastatic melanoma, renal cancer S11, S12, S13
Mesothelin NCT01897415 Metastatic PDA  
NCT01583686 Metastatic Cancer, Pancreatic Cancer, Mesothelioma, Ovarian Cancer S14, S15
NCT02414269 Malignant Pleural Disease/ Mesothelioma/ Metastases/ Lung Cancer/ Breast Cancer  
NCT02159716 Metastatic PDA/  
Epithelial Ovarian Cancer/ Malignant Epithelial Pleural Mesothelioma
NCT02388828 LTSFU  
HER-2 NCT02442297 Glioblastomas  
NCT01109095 Glioblastoma Multiforme S19
PSMA NCT01140373 Prostate Cancer  

Table 2: Clinical trials on tumor antigen targets for CAR-T and their associated diseases. * All clinical trial information and supplementary references were obtained from clinicaltrials.gov. ** Abbreviations can be found in supplementary Table 2.

References

  1. Cheadle EJ, Gornall H, Baldan V, Hanson V, Hawkins RE, et al. (2014) CAR T cells: driving the road from the laboratory to the clinic. Immunol Rev 257: 91-106.
  2. Gill S, June CH (2015) Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies. Immunol Rev 263: 68-89.
  3. DeFrancesco L (2014) CAR-T cell therapy seeks strategies to harness cytokine storm. Nat Biotechnol 32: 604.
  4. Ferguson KM (2008) Structure-based view of epidermal growth factor receptor regulation. Annu Rev Biophys 37: 353-373.
  5. 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.
  6. Davila ML, Riviere I, Wang X, Bartido S, Park J, et al. (2014). Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. SciTransl Med, 6: 224-225.
  7. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, et al. (2014) Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124: 188-195.
  8. Ramos CA, Savoldo B, Dotti G (2014) CD19-CAR trials. Cancer J 20: 112-118.
  9. Wagner J, Arora S (2014) Oncologic metabolic emergencies. Emerg Med Clin North Am 32: 509-525.
  10. Singh H, Huls H, Kebriaei P, Cooper LJ (2014) A new approach to gene therapy using Sleeping Beauty to genetically modify clinical-grade T cells to target CD19. Immunol Rev 257: 181-190.
Citation: DuPont C, Muhammad R, Sun J (2015) CAR-T: Expression, Expansion, and Clinical Applications. Pancreat Disord Ther 5:S5e001.

Copyright: © 2015 Crystal DuPont. 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