Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 May 12.
Published in final edited form as: Curr Opin Immunol. 2010 Feb 17;22(2):251–257. doi: 10.1016/j.coi.2010.01.020

Adoptive T Cell Therapy of Cancer

Malcolm K Brenner 1, Helen E Heslop 1
PMCID: PMC3093371  NIHMSID: NIHMS289628  PMID: 20171074

Summary

Adoptive transfer of T cells specific for antigens expressed on tumor cells is an attractive strategy for producing targeted and long-lived anti-tumor activity. T-cell therapies have shown activity in selected clinical applications but broader application is limited by inadequate persistence of transferred T cells and by tumor-evasion strategies. Current research focuses on defining the optimum type of cell for transfer, genetically modifying infused T cells to augment function and overcome tumor evasion strategies and modulating the host environment.

Introduction

Harnessing the cytotoxic power and targeting ability of the cellular immune system could significantly advance cancer therapy. While monoclonal antibody therapies are now well established in clinical practice, [1] exploitation of the cellular immune response to cancer has been slower, largely because of its greater complexity. Of the multiple forms of cellular immunotherapy, [2;3] adoptive transfer of T lymphocytes has perhaps been the most successful. For example, unmanipulated donor lymphocyte infusions (DLI) have been widely used to treat patients with relapsed hematologic malignancies after allogeneic hemopoietic stem cell transplantation (HSCT), [4] while antitumor responses post HSCT correlate with development of a diverse T-cell response specific for minor histocompatibility[5] or tumor-associated antigens such as WT1. [6] Moreover, ex-vivo expanded donor-derived cytotoxic specific T lymphocytes (CTLs) have proved highly effective in preventing or treating viral infections and Epstein-Barr virus (EBV) lymphomas developing post-transplant. [7**;8] In the autologous setting clinical responses have been observed following T cell therapies in patients with melanoma, lymphoma and nasopharyngeal cancer[912], providing insights into requirements for effective immunotherapy.

T cell immunotherapy for cancer nonetheless faces significant obstacles. For example, potentially immunogenic tumors have evolved a range of passive and active immune evasion strategies to avoid the consequences of immune activation, [13;14] and the immune response can even promote tumor growth. [15] Passive evasion tactics include the failure to present tumor antigens appropriately to the immune system and persistent tolerance to the self-antigens present on tumor cells. It is increasingly evident that an immune response may also select tumor variants that have lost the targeted antigens. For example, in 5/17 patients who relapsed with AML after haploidentical HSCT, the region of the host Chromosome 6 encoding the mismatched HLA haplotype was deleted, with consequent loss of the major target for the anti-tumor effect of the donor T cells. [16**] Active subversion of immunity may include the presence of factors such as TGFβ in the tumor microenvironment that diminish T-cell survival and function, or the secretion of chemokines that attract regulatory or inhibitory T cells rather than antitumor T effector cells. Recent laboratory insights have elucidated the molecular basis of many of these evasion strategies, allowing the development of potentially effective countermeasures. [13]

What is The Optimal Cell to Transfer?

One limitation of immunotherapy has been the suboptimal persistence of the adoptively transferred cells. During the last year, the optimal type of T cell for transfer to ensure optimal persistence and function has been extensively studied. In a primate model of CMV infection Berger et al reported that only antigen-specific CD8 clones derived from central memory T cells persisted long-term in vivo and accessed memory T cell niches. [17*] In a murine model using transgenic or retrovirally transduced T cells engineered to express a tumor-specific T-cell receptor, however, superior antitumor activity was seen in effector cells derived from naive T cells. [18*] Other studies showed that induction of Wnt-beta-catenin expression promoted generation of memory “stem” cells with enhanced proliferative and antitumor capacities which were more effective for adoptive transfer than other T cell subsets. [19*] These different conclusions about the optimal subset likely reflect the models used and emphasize that multiple factors determine the best phenotype for adoptive transfer into a complex immune network. For the moment we should be wary of placing excessive faith in broad assertions.

In practical terms, it is clearly possible to obtain long term reconstitution even with “suboptimal” subsets of cells. Long term follow up of gene-marked, donor derived, EBV specific cytotoxic T lymphocytes (EBV-CTLs) given to HSCT recipients to prevent or treat EBV lymphoma, showed marked cells persisting beyond 9 years, so that the transferred cells had entered the memory pool, even though the infused lines were predominantly effector memory phenotype. [7**] The transferred product in this study contained CD4 as well as CD8 cells, potentially facilitating long term persistence and function [20] and enabling T cell entry into infected tissue. [21*] In terms of identifying T cell subsets with the greatest cytotoxic potency, CD4 cells alone can induce clinical remissions of cancer, as documented in a report in which a CD4 clone specific for NY-ESO-1 induced a durable compete response in a patient with melanoma. [11*] Of note “epitope spreading” was seen, and the transferred NY-ESO specific immune response was followed by increased immune activity directed to the MAGE-3 and MART-1 antigens also expressed by the patient’s tumor cells. [11*]

If it turns out that it will be preferable to transfer specific subsets for a given application, then manipulation of the cytokine environment may make this feasible. In a study to optimize ex vivo culture conditions to ensure that allogeneic donor T cells maintained their anti tumor activity (in this case measured by alloreactivity) following genetic modification to express the TK suicide gene, IL-7 and IL-15 were found to be optimal. [22*] Similarly, primate studies have shown that IL15 can increase both CD4 and CD8 cells in vivo with minimal toxicity [23] while administration of IL-7 has been well tolerated in a clinical trial where it preferentially expanded naive T cells. [24] These cytokines may therefore be administered in conjunction with adoptively transferred T cells of the desired subset

Gene Transfer to Enhance and Simplify T-cell Therapy

Most tumor associated or tumor-specific antigens [25] are self-proteins to which the immune system has limited responsiveness, due to the development of tolerance by clonal deletion or anergy. Hence, tumor antigen–specific T cells isolated from patients with cancer prior to expansion and adoptive transfer may have low-affinity T-cell receptors (TCRs), limiting their cytotoxic activity against tumor cells. Investigators have overcome this limitation by using gene transfer to express transgenic TCR α and β chains of high affinity, or by expressing a synthetic chimeric antigen receptor. [2]

Artificial αβ T Cell Receptors

The cDNAs for the α and β chains of the TCR are cloned from class I HLA-restricted TCRs of tumor-reactive cytotoxic T cells and transferred to fresh T cells by an integrating vector, potentially giving the recipient cells the same antigen specificity as the donor T cells. [2] This approach allows rapid production of large numbers of tumor antigen–specific T cells. Preclinical studies have shown that infusion of αβTCR transgenic T cells can eradicate tumors in vivo. Recently, Morgan and colleagues treated melanoma patients with T cells genetically modified with MART-1–specific TCRs and reported regression of metastatic lesions in two patients together with prolonged persistence of CTLs, [26] although they have also noted toxicities to melanin expressing cells in the inner ear and the retina. [27**] Off target effects are not a sole preserve of TCR studies however as T cells specific for the minor histocompatibility antigens given to treat leukemic relapse produced transient benefit but also toxicity in some cases due to the unexpected presence of the target antigens on normal tissue, in this case affecting the lung. [28] One major constraint of TCR gene transfer is the development of hybrid TCR, which contain a mixture of native and transgenic receptors. These usually have loss of function, but in preclinical models, gain of function can be observed, producing autoreactivity and fatal graft-versus-host-disease–like syndrome. Efforts are now being made to prevent cross pairing and are summarized in Table 1.

Table 1.

Strategies to Prevent Cross Pairing in TCRs

Strategy Reference
Introduce cysteine residues in the transgenic receptors to favor the desired partner formation Kuball et al 2007[51]
Cohen et al 2007[52]
Transcribe the transgenic chains as a single RNA molecule with a 2a sequence between α and β sequences that ensures 1:1 stoichometry of production and requires post translational cleavage, that will increase the probability of the forming the desired chain combination. T Schumacher personal communication.
Incorporate CD3zeta in TCR Sebestyén et al 2008[53]
Edit endogenous TCR by using lentiviral delivery of TCR-zinc finger nucleases or small interfering RNAs Okamoto et al 2009[54]
Transfer αβ TCR to γ δ T cells Van der Veken et al 2009[55]

TCR: T cell receptor

Chimeric Antigen Receptors (CARs)

Instead of transducing T cells with additional αβTCR, it is possible to transfer chimeric TCRs, which may be generated by joining the light and heavy chain variable regions of a monoclonal antibody expressed as a singlechain Fv (scFv) molecule with the transmembrane and cytoplasmic signaling domains derived from CD3 ζ chain or Fc receptor γ chain through a flexible spacer. Thus they combine the antigen specificity of an antibody and the cytotoxic properties of a T cell in a single fusion molecule.

Since CARs bind to target antigens in an HLA-unrestricted manner, they are resistant to many tumor-immune evasion mechanisms, such as down-regulation of HLA class I molecules or failure to process or present proteins. First generation CARs incorporated the cytoplasmic region (endodomains) from the CD3 ζ or the Fc receptor γ chains as their signaling domain. Although these receptors successfully redirected T-cell cytotoxicity, they failed to stimulate T-cell proliferation and survival in vivo, likely because of the lack of appropriate costimulatory signals to T cells following engagement of their CAR. Efficacy was therefore modest in clinical trials in subjects with lymphoma, ovarian, neuroblastoma or renal cancer summarized in Table 2. [29*31]

Table 2.

Clinical Trials with T cells Expressing Chimeric Antigen Receptors

Reference Type of T cell CAR construct Cell Dose Targeted Cancer/Number of patients Serious Adverse Effects Persistence Responses
Kershaw et al[30] OKT3 activated T cells (8 patients) Alloantigen activated T cells (6 patients) α-folate receptor CAR retroviral vector with neomycin resistance gene 3 × 109 to 5 × 1010 (OKT3) 4.0 × 109 to 1.69 × 1011 (alloantigen) Ovarian cancer/14 patients None (IL2 effects in cohort receiving high dose IL2 Up to 3 weeks in 13 patients 12 months in 1 patient None
Park et al[56] OKT3 activated T cells (clones) CE7R-CAR plasmid with HyTK 108 to 109 cells/m2 Neuroblastoma/6 patients None 1–42 days 1 of 6 with evaluable tumor had a PR
Lamers et al[31] G250-CAR retroviral vector 0.38 to 2.13 × 109 Renal cancer Grade 2-4 liver toxicity Up to 53 days None
Till et al[29*] OKT3 activated T cells (clones in 3 and lines in 4) CD20-CAR plasmid with neomycin resistance gene 108 to 3.3 × 109 cells/m2 CD20+ low grade B cell lymphoma/7 patients None 1 to 3 weeks (clones) 5 to 9 weeks (T cell lines and low dose IL2) 4 of 5 with evaluable disease had stable disease and one a PR
Pule et al [34**] OKT3 activated T cells and EBV specific CTLs GD2-CAR retroviral vector 2 × 107 to 2 × 108 cells/m2 of each product Neuroblastoma/11 patients None Up to 3 weeks for the activated T cells and up to 6 months for CTLs 4 of 8 with evaluable tumor had necrosis or responses with 1 CR

EBV. Epstein Barr Virus

CTL: cytotoxic T lymphocyte

CR: complete remission

PR: partial remission

HyTK: hygromycin thymidine kinase

Second-generation CARs were constructed by incorporating signaling domains from individual costimulatory molecules such as CD28, OX40, and 4-1BB within the endodomain, and improved antigen-specific T cell activation and expansion. Third generation CARs include a combination of co-stimulatory endodomains[32], but there are concerns that such receptors may either be too easily triggered by low avidity “off-target’ binding or may produce too potent an activation signal, producing potentially lethal consequences from the resulting cytokine storm. [33] An alternative approach is to express CARs in antigen-specific T cells, which will then also be activated and expanded through engagement of their native αβTCR by antigen on professional antigen presenting cells, with attendant co-stimulation. [34**;35] For example, subjects receiving EBV-specific CTLs engineered with a CAR (CAR-CTLs) specific for the disialoganglioside antigen GD2a on neuroblastoma cells show longer in vivo persistence of CAR-CTLs compared with unselected T cells engineered with the identical CAR, since the CAR-CTLs encounter (persistent) Epstein-Barr virus antigens. [34**] (Table 2) Longer persistence is associated with tumor responses including complete remission.

Overcoming Immune Evasion Strategies

One of the main challenges to effective adoptive T-cell therapy is the lack of in vivo expansion and maintenance of ex-vivo manipulated, adoptively transferred T cells because of tumor-induced immune evasion mechanisms. Gene transfer technologies allow us to modify T cells and restore their functionality in a hostile environment. For example, many tumor cells or their associated stroma produce TGF-β which favors the development of immune tolerance and T-cell anergy, inducing T effector cell growth arrest with induction of Tregs. [15] Bollard and coworkers showed that human and murine antigen-specific T cells could express a dominant negative (dn) TGF-β receptor following retroviral transduction and become resistant to the anti-proliferative effects of TGF- β retaining their effector function in vivo. [36;37] T cells may also be modified to express cytokine or cytokine receptor genes that mimic the milieu found during lymphoid regeneration and restoration of homeostasis, such as IL-2, IL-7, or IL-15. [3840] Preliminary clinical data are encouraging but as yet, we do not know for certain how safe or effective these transgenic cytokines and their receptors will be. An alternative strategy is administer antibodies to receptors on T cells that have an inhibitory effect on T cell activation such as CTLA4 (cytotoxic T lymphocyte antigen-4) and PD-1 (programmed death-1).

Cytotoxic T cells can also be made resistant to small molecule cancer therapeutics, many of which cause profound immunosuppression. Several possibilities exist, from the introduction of drug metabolizing enzymes to expression of rapalog resistant MTOR. Other investigators have made T cells that express transgenic calcineurin molecules resistant to commonly used post-transplant immunosuppressive drugs such as cyclosporine or silenced the FK506-binding protein with a specific small interfering RNA [41;42] potentially allowing administration of allogeneic cancer specific T cells to subjects with cancer whose own immune system is prevented from rejecting the allogeneic cells by immunosuppressive drugs. The overall value of this approach remains to be established. Another approach is to combine cellular immunotherapy with antibody treatment. In a recent report in a mouse model of immunotherapy targeting CD20, concurrent antibody therapy that depletes antigen-expressing normal tissues enhanced the ability of cTCR(+) T cells to survive and control tumors. [43]

Since transduced T cells have the potential to last the lifetime of the host and even to expand in number, any adverse effect attributable to gene transfer or gene modified cells may worsen over time. As a consequence, “suicide” strategies use a second transgene that accompanies the gene of interest, and which allows the cell to be destroyed on exposure to a specific signal. The most widely used is the herpes simplex viral thymidine kinase (Tk) gene, the product of which will phosphorylate ganciclovir or acyclovir to the active moiety, which interferes with DNA synthesis. The Tk gene has been introduced into allogeneic T lymphocytes used as donor lymphocyte infusions following stem cell transplantation. [44*] If the infused cells produce graft-versus-host disease rather than the desired antiviral and antileukemic activity, they can be inactivated by administration of the ganciclovir prodrug. The safety and benefits of this approach appear substantial and the approach has now reached phase III clinical trial. [44*] Though apparently effective, the Tk gene may itself be immunogenic, leading to undesired elimination of a transduced cell population. Moreover, gancyclovir is a useful drug for immunocompromised patients who develop cytomegalovirus (CMV) infections; in these patients, administration of GCV to treat CMV would produce cell suicide irrespective of need. Finally, Tk/GCV may have limited ability to actually kill cell populations, particularly those that are post mitotic. Investigators are therefore developing alternatives such as inducible Caspase9 (icasp9). Since icasp9 is a naturally occurring component of the caspase pathway it should be non-immunogenic and produce apoptosis even in non-dividing cells. [45] The molecule can be triggered by administration of a small molecule dimeriser that brings together two non-functional icasp9 molecules to form the active enzyme. The approach has begun clinical study.

Broader Implementation of T-cell Therapy

As described above, complex biological therapies are indeed intricate to implement. Fortunately, the successes described in the preceding paragraphs have spurred methodological developments to simplify and accelerate manufacture of engineered viral- or tumor-specific CTLs. Several investigators have evaluated artificial antigen presenting cells to provide a rapid source of antigen. [4648] and clinical trials are underway using T cells generated by these stimulator cells. Investigators have also developed methods to generate CTL lines that have anti-viral activity for multiple antigens in a single culture, [49] and mini-bioreactors in which to prepare these cells in a closed system. [50] In combination, these techniques allow sufficient CTLs for patient treatment to be made in less than 10 days instead of longer than 10 weeks. Moreover, these lines can be banked and given to partially HLA matched patients as “off the shelf” reagents, and a multicenter study is evaluating whether tri-virus specific CTL lines may have similar activity against EBV, CMV and adenovirus in partially HLA-matched allogeneic patients. If successful, extension to the treatment of malignant disease would follow.

Acknowledgments

This work was supported by NIH grants PO1 CA94237, P50CA126752, U54HL081007, and aSpecialized Center of Research Award from the Leukemia Lymphoma Society, and a Doris Duke Distinguished Clinical Scientist Award to HEH

References

  • 1.Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC. Monoclonal antibody successes in the clinic. Nat Biotechnol. 2005;23:1073–1078. doi: 10.1038/nbt0905-1073. [DOI] [PubMed] [Google Scholar]
  • 2.Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8:299–308. doi: 10.1038/nrc2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Disis ML, Bernhard H, Jaffee EM. Use of tumour-responsive T cells as cancer treatment. Lancet. 2009;373:673–683. doi: 10.1016/S0140-6736(09)60404-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371–4383. doi: 10.1182/blood-2008-03-077974. [DOI] [PubMed] [Google Scholar]
  • 5.Nishida T, Hudecek M, Kostic A, Bleakley M, Warren EH, Maloney D, Storb R, Riddell SR. Development of tumor-reactive T cells after nonmyeloablative allogeneic hematopoietic stem cell transplant for chronic lymphocytic leukemia. Clin Cancer Res. 2009;15:4759–4768. doi: 10.1158/1078-0432.CCR-09-0199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rezvani K, Yong AS, Savani BN, Mielke S, Keyvanfar K, Gostick E, Price DA, Douek DC, Barrett AJ. Graft-versus-leukemia effects associated with detectable Wilms tumor-1 specific T lymphocytes after allogeneic stem-cell transplantation for acute lymphoblastic leukemia. Blood. 2007;110:1924–1932. doi: 10.1182/blood-2007-03-076844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7**.Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA, Bollard CM, Liu H, Wu MF, Rochester RJ, Amrolia PJ, Hurwitz JL, Brenner MK, Rooney CM. Long term outcome of EBV specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood. 2009 doi: 10.1182/blood-2009-08-239186. epub online. Provides long term follow-up on patients who received donor-derived EBV-specific cytotoxic T lymphocytes (CTLs) showing that they were effective in 101 high-risk patients as prophylaxis and 11 of 13 as therapy. Gene-marking showed persistence of functional CTLs for up to 9 years. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Leen AM, Myers GD, Sili U, Huls MH, Weiss H, Leung KS, Carrum G, Krance RA, Chang CC, Molldrem JJ, Gee AP, Brenner MK, Heslop HE, Rooney CM, Bollard CM. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med. 2006;12:1160–1166. doi: 10.1038/nm1475. [DOI] [PubMed] [Google Scholar]
  • 9.Louis CU, Straathof K, Bollard CM, Gerken C, Huls MH, Gresik MV, Wu MF, Weiss HL, Gee AP, Brenner MK, Rooney CM, Heslop HE, Gottschalk S. Enhancing the in vivo expansion of adoptively transferred EBV-specific CTL with lymphodepleting CD45 monoclonal antibodies in NPC patients. Blood. 2009;113:2442–2450. doi: 10.1182/blood-2008-05-157222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, Robbins PF, Huang J, Citrin DE, Leitman SF, Wunderlich J, Restifo NP, Thomasian A, Downey SG, Smith FO, Klapper J, Morton K, Laurencot C, White DE, Rosenberg SA. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26:5233–5239. doi: 10.1200/JCO.2008.16.5449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11*.Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, Jungbluth A, Gnjatic S, Thompson JA, Yee C. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med. 2008;358:2698–2703. doi: 10.1056/NEJMoa0800251. Sustained complete remission in a melanoma patient treated with CD4 clones specific for NY-ESO despite this antigen only being expressed by 50–75% of tumor cells. This was likely due to epitope spreading as increased immune activity directed to at least two other antigens expressed on the patient’s tumor cells - MAGE-3 and MART-1 -was seen. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bollard CM, Gottschalk S, Leen AM, Weiss H, Straathof KC, Carrum G, Khalil M, Wu MF, Huls MH, Chang CC, Gresik MV, Gee AP, Brenner MK, Rooney CM, Heslop HE. Complete responses of relapsed lymphoma following genetic modification of tumor-antigen presenting cells and T-lymphocyte transfer. Blood. 2007;110:2838–2845. doi: 10.1182/blood-2007-05-091280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5:263–274. doi: 10.1038/nrc1586. [DOI] [PubMed] [Google Scholar]
  • 14.Stewart TJ, Abrams SI. How tumours escape mass destruction. Oncogene. 2008;27:5894–5903. doi: 10.1038/onc.2008.268. [DOI] [PubMed] [Google Scholar]
  • 15.Petrausch U, Jensen SM, Twitty C, Poehlein CH, Haley DP, Walker EB, Fox BA. Disruption of TGF-beta signaling prevents the generation of tumor-sensitized regulatory T cells and facilitates therapeutic antitumor immunity. J Immunol. 2009;183:3682–3689. doi: 10.4049/jimmunol.0900560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16**.Vago L, Perna SK, Zanussi M, Mazzi B, Barlassina C, Stanghellini MT, Perrelli NF, Cosentino C, Torri F, Angius A, Forno B, Casucci M, Bernardi M, Peccatori J, Corti C, Bondanza A, Ferrari M, Rossini S, Roncarolo MG, Bordignon C, Bonini C, Ciceri F, Fleischhauer K. Loss of mismatched HLA in leukemia after stem-cell transplantation. N Engl J Med. 2009;361:478–488. doi: 10.1056/NEJMoa0811036. Describes suppression of mismatched HLA antigens as an immune evasion strategy in recipients of haploidentical HSCT. In 5 of the 17 patients with relapsed AML post transplant, the mismatched HLA antigens in leukemia cells had been replaced withan HLA haplotype derived from the donor parent so there was no longer any HLA mismatch to serve as a target for the donor immune cells. [DOI] [PubMed] [Google Scholar]
  • 17*.Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305. doi: 10.1172/JCI32103. In a primate model of CMV infection only T cells derived from cells with central memory phenotype persist long term. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18*.Hinrichs CS, Borman ZA, Cassard L, Gattinoni L, Spolski R, Yu Z, Sanchez-Perez L, Muranski P, Kern SJ, Logun C, Palmer DC, Ji Y, Reger RN, Leonard WJ, Danner RL, Rosenberg SA, Restifo NP. Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc Natl Acad Sci USA. 2009;106:17469–17474. doi: 10.1073/pnas.0907448106. In a murine model using transgenic or retrovirally transduced T cells engineered to express a tumor-specific T-cell receptor, superior antitumor activity was seen in effector cells derived from naive T cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19*.Gattinoni L, Zhong XS, Palmer DC, Ji Y, Hinrichs CS, Yu Z, Wrzesinski C, Boni A, Cassard L, Garvin LM, Paulos CM, Muranski P, Restifo NP. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat Med. 2009;15:808–813. doi: 10.1038/nm.1982. Induction of Wnt-beta-catenin signaling promoted the generation of self-renewing multipotent CD8(+) memory stem cells with proliferative and antitumor capacities exceeding those of central and effector memory T cell subsets. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Pulendran B, Ahmed R. Translating innate immunity into immunological memory: implications for vaccine development. Cell. 2006;124:849–863. doi: 10.1016/j.cell.2006.02.019. [DOI] [PubMed] [Google Scholar]
  • 21*.Nakanishi Y, Lu B, Gerard C, Iwasaki A. CD8(+) T lymphocyte mobilization to virus-infected tissue requires CD4(+) T-cell help. Nature. 2009 doi: 10.1038/nature08511. epub online. A new role for CD4 cells in mobilizing effector CTL to peripheral sites of infection where they help to eliminate infected cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22*.Kaneko S, Mastaglio S, Bondanza A, Ponzoni M, Sanvito F, Aldrighetti L, Radrizzani M, La Seta-Catamancio S, Provasi E, Mondino A, Nagasawa T, Fleischhauer K, Russo V, Traversari C, Ciceri F, Bordignon C, Bonini C. IL-7 and IL-15 allow the generation of suicide gene-modified alloreactive self-renewing central memory human T lymphocytes. Blood. 2009;113:1006–1015. doi: 10.1182/blood-2008-05-156059. Defines culture conditions that result in generation of central memory T cells that maintain alloreactivity after transduction with the Tk retroviral vector. [DOI] [PubMed] [Google Scholar]
  • 23.Berger C, Berger M, Hackman RC, Gough M, Elliott C, Jensen MC, Riddell SR. Safety and immunologic effects of IL-15 administration in nonhuman primates. Blood. 2009;114:2417–2426. doi: 10.1182/blood-2008-12-189266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sportes C, Hakim FT, Memon SA, Zhang H, Chua KS, Brown MR, Fleisher TA, Krumlauf MC, Babb RR, Chow CK, Fry TJ, Engels J, Buffet R, Morre M, Amato RJ, Venzon DJ, Korngold R, Pecora A, Gress RE, Mackall CL. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. J Exp Med. 2008;205:1701–1714. doi: 10.1084/jem.20071681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, Mellman I, Prindiville SA, Viner JL, Weiner LM, Matrisian LM. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15:5323–5337. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA, Rosenberg SA. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–129. doi: 10.1126/science.1129003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27**.Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, Kammula US, Royal RE, Sherry RM, Wunderlich JR, Lee CC, Restifo NP, Schwarz SL, Cogdill AP, Bishop RJ, Kim H, Brewer CC, Rudy SF, VanWaes C, Davis JL, Mathur A, Ripley RT, Nathan DA, Laurencot CM, Rosenberg SA. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114:535–546. doi: 10.1182/blood-2009-03-211714. Reports clinical trial of T cells expressing TCRs highly reactive to melanoma antigens with objective cancer regressions seen in 30% and 19% of patients who received the human or mouse TCRs, respectively. However, patients exhibited destruction of normal melanocytes in the skin, eye, and ear, and sometimes required local steroid administration to treat uveitis and hearing loss. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Riddell SR, Bleakley M, Nishida T, Berger C, Warren EH. Adoptive transfer of allogeneic antigen-specific T cells. Biol Blood Marrow Transplant. 2006;12:9–12. doi: 10.1016/j.bbmt.2005.10.025. [DOI] [PubMed] [Google Scholar]
  • 29*.Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, Qian X, James SE, Raubitschek A, Forman SJ, Gopal AK, Pagel JM, Lindgren CG, Greenberg PD, Riddell SR, Press OW. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood. 2008;112:2261–2271. doi: 10.1182/blood-2007-12-128843. Report of a clinical trial using a plasmid containing a selectable marker to transfer a CD20 CAR into T cells expanded non-specifically with OKT3 and IL2. Persistence was short in 3 patients who received an oligoclonal product but longer in 4 patients who received polyclonal populations and 1 PR was obtained. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, Rogers-Freezer L, Chen CC, Yang JC, Rosenberg SA, Hwu P. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12:6106–6115. doi: 10.1158/1078-0432.CCR-06-1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol. 2006;24:e20–e22. doi: 10.1200/JCO.2006.05.9964. [DOI] [PubMed] [Google Scholar]
  • 32.Zhao Y, Wang QJ, Yang S, Kochenderfer JN, Zheng Z, Zhong X, Sadelain M, Eshhar Z, Rosenberg SA, Morgan RA. A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. J Immunol. 2009;183:5563–5574. doi: 10.4049/jimmunol.0900447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Minutes of the December 1st 2009 RAC meeting. 2009 http://oba.od.nih.gov/rdna_rac/rac_meetings.html. Ref Type: Electronic Citation.
  • 34**.Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, Huls MH, Liu E, Gee AP, Mei Z, Yvon E, Weiss HL, Liu H, Rooney CM, Heslop HE, Brenner MK. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–1270. doi: 10.1038/nm.1882. Report of a clinical trial transferring a distinguishable CAR targeting GD2 into OKT3 activated T cells and EBV CTLs. Persistence of the EBV-CTLs was greater with clinical activity in 4 of 8 patients with active disease including 1 sustained CR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.van Loenen MM, Hagedoorn RS, Kester MG, Hoogeboom M, Willemze R, Falkenburg JH, Heemskerk MH. Kinetic preservation of dual specificity of coprogrammed minor histocompatibility antigen-reactive virus-specific T cells. Cancer Res. 2009;69:2034–2041. doi: 10.1158/0008-5472.CAN-08-2523. [DOI] [PubMed] [Google Scholar]
  • 36.Foster AE, Dotti G, Lu A, Khalil M, Brenner MK, Heslop HE, Rooney CM, Bollard CM. Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-beta receptor. J Immunother. 2008;31:500–505. doi: 10.1097/CJI.0b013e318177092b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bollard CM, Rossig C, Calonge MJ, Huls MH, Wagner HJ, Massague J, Brenner MK, Heslop HE, Rooney CM. Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunity. Blood. 2002;99:3179–3187. doi: 10.1182/blood.v99.9.3179. [DOI] [PubMed] [Google Scholar]
  • 38.Vera JF, Hoyos V, Savoldo B, Quintarelli C, Giordano Attianese GM, Leen AM, Liu H, Foster AE, Heslop HE, Rooney CM, Brenner MK, Dotti G. Genetic manipulation of tumor-specific cytotoxic T lymphocytes to restore responsiveness to IL-7. Mol Ther. 2009;17:880–888. doi: 10.1038/mt.2009.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Heemskerk B, Liu K, Dudley ME, Johnson LA, Kaiser A, Downey S, Zheng Z, Shelton TE, Matsuda K, Robbins PF, Morgan RA, Rosenberg SA. Adoptive cell therapy for patients with melanoma, using tumor-infiltrating lymphocytes genetically engineered to secrete interleukin-2. Hum Gene Ther. 2008;19:496–510. doi: 10.1089/hum.2007.0171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Quintarelli C, Vera JF, Savoldo B, Giordano Attianese GM, Pule M, Foster AE, Heslop HE, Rooney CM, Brenner MK, Dotti G. Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes. Blood. 2007;110:2793–2802. doi: 10.1182/blood-2007-02-072843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Brewin J, Mancao C, Straathof K, Karlsson H, Samarasinghe S, Amrolia PJ, Pule M. Generation of EBV-specific cytotoxic T-cells that are resistant to calcineurin inhibitors for the treatment of post-transplant lymphoproliferative disease. Blood. 2009 doi: 10.1182/blood-2009-07-228387. epub online. [DOI] [PubMed] [Google Scholar]
  • 42.De Angelis B, Dotti G, Quintarelli C, Huye LE, Zhang L, Zhang M, Pane F, Heslop HE, Brenner MK, Rooney CM, Savoldo B. Generation of Epstein-Barr-virus-specific cytotoxic T lymphocytes resistant to the immunosuppressive drug tacrolimus (FK506) Blood. 2009 doi: 10.1182/blood-2009-07-230482. epub online. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.James SE, Orgun NN, Tedder TF, Shlomchik MJ, Jensen MC, Lin Y, Greenberg PD, Press OW. Antibody mediated B cell depletion prior to adoptive immunotherapy with T cells expressing CD20-specific chimeric T cell receptors facilitates eradication of leukemia in immunocompetent mice. Blood. 2009 doi: 10.1182/blood-2009-08-232967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44*.Ciceri F, Bonini C, Stanghellini MT, Bondanza A, Traversari C, Salomoni M, Turchetto L, Colombi S, Bernardi M, Peccatori J, Pescarollo A, Servida P, Magnani Z, Perna SK, Valtolina V, Crippa F, Callegaro L, Spoldi E, Crocchiolo R, Fleischhauer K, Ponzoni M, Vago L, Rossini S, Santoro A, Todisco E, Apperley J, Olavarria E, Slavin S, Weissinger EM, Ganser A, Stadler M, Yannaki E, Fassas A, Anagnostopoulos A, Bregni M, Stampino CG, Bruzzi P, Bordignon C. Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): a non-randomised phase I–II study. Lancet Oncol. 2009;10:489–500. doi: 10.1016/S1470-2045(09)70074-9. Report of a study administering donor lymphocytes expressing the herpes-simplex thymidine kinase suicide gene to recipients of haploidentical transplant. 22 of 28 patients obtained immune reconstitution and in 10 patients who developed acute GVHD and one who developed chronic GVHD, the complication was controlled by administration of the suicide prodrug. [DOI] [PubMed] [Google Scholar]
  • 45.Tey SK, Dotti G, Rooney CM, Heslop HE, Brenner MK. Inducible caspase 9 suicide gene to improve the safety of allodepleted T cells after haploidentical stem cell transplantation. Biol Blood Marrow Transplant. 2007;13:913–924. doi: 10.1016/j.bbmt.2007.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hasan AN, Kollen WJ, Trivedi D, Selvakumar A, Dupont B, Sadelain M, O’Reilly RJ. A panel of artificial APCs expressing prevalent HLA alleles permits generation of cytotoxic T cells specific for both dominant and subdominant viral epitopes for adoptive therapy. J Immunol. 2009;183:2837–2850. doi: 10.4049/jimmunol.0804178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Butler MO, Lee JS, Ansen S, Neuberg D, Hodi FS, Murray AP, Drury L, Berezovskaya A, Mulligan RC, Nadler LM, Hirano N. Long-lived antitumor CD8+ lymphocytes for adoptive therapy generated using an artificial antigen-presenting cell. Clin Cancer Res. 2007;13:1857–1867. doi: 10.1158/1078-0432.CCR-06-1905. [DOI] [PubMed] [Google Scholar]
  • 48.June CH, Blazar BR, Riley JL. Engineering lymphocyte subsets: tools, trials and tribulations. Nat Rev Immunol. 2009;9:704–716. doi: 10.1038/nri2635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gerdemann U, Christin AS, Vera JF, Ramos CA, Fujita Y, Liu H, Dilloo D, Heslop HE, Brenner MK, Rooney CM, Leen AM. Nucleofection of DCs to generate Multivirus-specific T cells for prevention or treatment of viral infections in the immunocompromised host. Mol Ther. 2009;17:1616–1625. doi: 10.1038/mt.2009.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Vera JF, Brenner LJ, Gerdemann U, Ngo MC, Sili U, Wilson J, Dotti G, Heslop HE, Leen AM, Rooney CM. Accelerated production of antigen-specific T-cells for pre-clinical and clinical applications using Gas-permeable Rapid Expansion cultureware (G-Rex) J Immunother. 2009 doi: 10.1097/CJI.0b013e3181c0c3cb. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kuball J, Dossett ML, Wolfl M, Ho WY, Voss RH, Fowler C, Greenberg PD. Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood. 2007;109:2331–2338. doi: 10.1182/blood-2006-05-023069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cohen CJ, Li YF, El-Gamil M, Robbins PF, Rosenberg SA, Morgan RA. Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. Cancer Res. 2007;67:3898–3903. doi: 10.1158/0008-5472.CAN-06-3986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sebestyen Z, Schooten E, Sals T, Zaldivar I, San JE, Alarcon B, Bobisse S, Rosato A, Szollosi J, Gratama JW, Willemsen RA, Debets R. Human TCR that incorporate CD3zeta induce highly preferred pairing between TCRalpha and beta chains following gene transfer. J Immunol. 2008;180:7736–7746. doi: 10.4049/jimmunol.180.11.7736. [DOI] [PubMed] [Google Scholar]
  • 54.Okamoto S, Mineno J, Ikeda H, Fujiwara H, Yasukawa M, Shiku H, Kato I. Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR. Cancer Res. 2009 doi: 10.1158/0008-5472.CAN-09-1450. [DOI] [PubMed] [Google Scholar]
  • 55.van der Veken LT, Coccoris M, Swart E, Falkenburg JH, Schumacher TN, Heemskerk MH. Alpha beta T cell receptor transfer to gamma delta T cells generates functional effector cells without mixed TCR dimers in vivo. J Immunol. 2009;182:164–170. doi: 10.4049/jimmunol.182.1.164. [DOI] [PubMed] [Google Scholar]
  • 56.Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet HB, Bautista C, Chang WC, Ostberg JR, Jensen MC. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–833. doi: 10.1038/sj.mt.6300104. [DOI] [PubMed] [Google Scholar]

RESOURCES