Trends in Biotechnology
Volume 29, Issue 11, November 2011, Pages 550-557
Journal home page for Trends in Biotechnology

Review
Treating cancer with genetically engineered T cells

https://doi.org/10.1016/j.tibtech.2011.04.009Get rights and content

Administration of ex vivo cultured, naturally occurring tumor-infiltrating lymphocytes (TILs) has been shown to mediate durable regression of melanoma tumors. However, the generation of TILs is not possible in all patients and there has been limited success in generating TIL in other cancers. Advances in genetic engineering have overcome these limitations by introducing tumor-antigen-targeting receptors into human T lymphocytes. Physicians can now genetically engineer lymphocytes to express highly active T-cell receptors (TCRs) or chimeric antigen receptors (CARs) targeting a variety of tumor antigens expressed in cancer patients. In this review, we discuss the development of TCR and CAR gene transfer technology and the expansion of these therapies into different cancers with the recent demonstration of the clinical efficacy of these treatments.

Introduction

The ability of lymphocytes to eradicate tumor cells in cancer patients has been demonstrated in metastatic melanoma for which the T cell cytokine interleukin (IL)-2 (aldesleukin), now an FDA-approved therapy, can mediate measurable responses in 15% of patients treated 1, 2. The immunogenic nature of melanoma tumors has served as the foundation for the development of other immune-based therapies for the treatment of this and other cancers. Nonspecific immune stimulation with IL-2 and anti-cytotoxic T-lymphocyte antigen-4 (Ipilimumab) antibody leads to activation of antitumor lymphocytes in vivo, and has been shown to mediate tumor regression in metastatic melanoma and renal cell cancer [3]. Currently, the most effective immune-based therapy for melanoma is adoptive cell therapy involving the generation of T lymphocytes with antitumor activity. When these TILs are infused into patients along with IL-2 and reduced-intensity chemotherapy to knock down temporarily the patient's circulating immune cells, TILs can mediate tumor responses in up to 70% of patients, with a significant portion of these being durable complete responses (defined as the disappearance of all target lesions) [4].

The protein that T cells utilize to identify foreign epitopes (or in the case of TILs, tumor antigens) is the T-cell receptor (TCR). The TCR is a member of the immunoglobulin gene super family and is a heterodimer composed of an α and a β chain. TCR genes can be isolated from tumor-reactive T cell clones (clones that mediate clinical responses), inserted into gene transfer vectors, and used to genetically engineer normal T lymphocytes to redirect them with antitumor specificity. These genetically engineered T cells were shown to result in objective responses in a small number of metastatic melanoma patients in 2006 [5]. Progress in the ability to mediate responses with the above immune-based therapies in metastatic melanoma has prompted the translation of these therapies to treat cancers of other tissues and organs. Recently, a series of new clinical trials have shown that measurable responses can be achieved using gene-modified T cells in cancers other than melanoma, including colorectal cancer, lymphoma, neuroblastoma, and synovial sarcoma 6, 7, 8, 9, 10. In this review, we discuss the development of T cell genetic engineering, two specific gene modifications, and the clinical applications of these biotechnologies.

Section snippets

Initial studies using natural antitumor T-cell therapy

Adoptive immunotherapy using the transfer of viral-antigen-specific T cells is now a well-established procedure that results in effective treatment of transplant-associated viral infections and rare viral-related malignancies. In these approaches, allogeneic peripheral blood lymphocytes (PBLs) are first isolated from the bone marrow donor. PBLs with reactivity to human cytomegalovirus (CMV) or Epstein–Barr virus (EBV) are isolated and expanded, and then intravenously infused into patients

Development of engineered T cells: TCR gene transfer

As an alternative to TIL therapy, highly avid TCRs can be cloned from naturally occurring T cells and, by using gene transfer vectors, introduced into patient's lymphocytes, thus creating the opportunity to generate large quantities of antigen-specific T cells for treatment (Figure 1) 14, 15. The first step in TCR gene therapy is to isolate a high-affinity T-cell clone for a defined target antigen. These TCRs can be isolated from patients with rare, highly reactive T-cell clones that recognize

Development of engineered T cells: chimeric antigen receptors

Redirection of T-cell specificity by TCRs is constrained by HLA restriction, which limits the applicability of TCR therapy to patients who express the particular HLA type (similar to organ or bone marrow transplantation). In addition, tumors can lose their antigen expression by downregulation of HLA [38]. CARs can avoid these limitations because they can confer non-HLA restricted specificity to T cells based on antibody recognition. CARs consist of a tumor-antigen-binding domain of a

Clinical trials using engineered T cells

As first documented in melanoma, genetically engineered T cells can recognize and destroy large established tumors in cancer patients; an example of this is shown in Figure 3 (this particular patient had complete elimination of melanoma tumors and remained disease free >4 years post-treatment). Recently, several clinical trials have been reported documenting the clinical efficacy of gene-modified T cells for treatment of other cancers (Table 1). These trials used both TCR- and CAR-engineered T

Future directions

Active efforts are being made in the basic research arena to improve TCR and CAR function. For TCRs, site-directed mutagenesis in the CDR region to improve antigen affinity and manipulations to improve TCR chain pairing are active areas of research. For CARs, second and third generation CARs with additional co-stimulatory signaling domains are being investigated for their ability to improve cell signaling, survival, and proliferation. Lessons learned from the clinical responses in patients

Acknowledgements

All of the clinical trials results reported from the Surgery Branch of the National Cancer Institute were performed by principal investigator and Branch Chief, Steve A. Rosenberg, MD, PhD. We thank Nicholas Restifo for the creation of Figure 1 in this review and James Kochenderfer for helpful discussions.

References (62)

  • M. Sadelain

    The promise and potential pitfalls of chimeric antigen receptors

    Curr. Opin. Immunol.

    (2009)
  • B. Jena

    Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor

    Blood

    (2010)
  • C.H. Lamers

    Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells

    Blood

    (2011)
  • L.A. Johnson

    Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen

    Blood

    (2009)
  • S.A. Rosenberg

    Of mice, not men: no evidence for graft-versus-host disease in humans receiving T-cell receptor-transduced autologous T cells

    Mol. Ther.

    (2010)
  • S. Pontvert-Delucq

    Characterization and functional analysis of adult human bone marrow cell subsets in relation to B-lymphoid development

    Blood

    (1993)
  • F.M. Uckun

    Detailed studies on expression and function of CD19 surface determinant by using B43 monoclonal antibody and the clinical potential of anti-CD19 immunotoxins

    Blood

    (1988)
  • D.B. Kohn

    CARs on track in the clinic

    Mol. Ther.

    (2011)
  • J.N. Kochenderfer

    A phase I clinical trial of treatment of B-cell malignancies with autologous anti-CD19-CAR-transduced T cells

    Blood

    (2010)
  • R. Brentjens

    Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial

    Mol. Ther.

    (2010)
  • R.A. Morgan

    Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2

    Mol. Ther.

    (2010)
  • S.A. Rosenberg

    Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2: identification of the antigens mediating response

    Ann. Surg.

    (1998)
  • M.B. Atkins

    High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993

    J. Clin. Oncol.

    (1999)
  • A.A. Sarnaik et al.

    Recent advances using anti-CTLA-4 for the treatment of melanoma

    Cancer J.

    (2009)
  • M.E. Dudley

    Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens

    J. Clin. Oncol.

    (2008)
  • R.A. Morgan

    Cancer regression in patients after transfer of genetically engineered lymphocytes

    Science

    (2006)
  • M.A. Pule

    Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma

    Nat. Med.

    (2008)
  • P.F. Robbins

    Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1

    J. Clin. Oncol.

    (2011)
  • S.A. Rosenberg

    Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report

    N. Engl. J. Med.

    (1988)
  • M.E. Dudley

    CD8+ enriched “young” tumor infiltrating lymphocytes can mediate regression of metastatic melanoma

    Clin. Cancer Res.

    (2010)
  • S. Thomas

    Molecular immunology lessons from therapeutic T-cell receptor gene transfer

    Immunology

    (2010)
  • Cited by (198)

    • Advances in adoptive T-cell therapy for metastatic melanoma

      2023, Current Research in Translational Medicine
    • HBV as a target for CAR or TCR-T cell therapy

      2020, Current Opinion in Immunology
    View all citing articles on Scopus
    View full text