Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2012 May 1;1(3):306–315. doi: 10.4161/onci.19549

Trial Watch

Adoptive cell transfer immunotherapy

Lorenzo Galluzzi 1,2,3,, Erika Vacchelli 1,2,3,, Alexander Eggermont 2, Wolf Herve´ Fridman 4,5, Jerome Galon 4,6, Catherine Sautès-Fridman 4,7,6, Eric Tartour 7,5,8, Laurence Zitvogel 2,9, Guido Kroemer 1,2,7,5,10,*
PMCID: PMC3382856  PMID: 22737606

Abstract

During the last two decades, several approaches for the activation of the immune system against cancer have been developed. These include rather unselective maneuvers such as the systemic administration of immunostimulatory agents (e.g., interleukin-2) as well as targeted interventions, encompassing highly specific monoclonal antibodies, vaccines and cell-based therapies. Among the latter, adoptive cell transfer (ACT) involves the selection of autologous lymphocytes with antitumor activity, their expansion/activation ex vivo, and their reinfusion into the patient, often in the context of lymphodepleting regimens (to minimize endogenous immunosuppression). Such autologous cells can be isolated from tumor-infiltrating lymphocytes or generated by manipulating circulating lymphocytes for the expression of tumor-specific T-cell receptors. In addition, autologous lymphocytes can be genetically engineered to prolong their in vivo persistence, to boost antitumor responses and/or to minimize side effects. ACT has recently been shown to be associated with a consistent rate of durable regressions in melanoma and renal cell carcinoma patients and holds great promises in several other oncological settings. In this Trial Watch, we will briefly review the scientific rationale behind ACT and discuss the progress of recent clinical trials evaluating the safety and effectiveness of adoptive cell transfer as an anticancer therapy.

Keywords: CD8, Tregs, cyclophosphamide, interferon γ, lymphodepletion, tumor-infiltrating lymphocytes

Introduction

For a long time, the immune system has been believed to participate in oncogenesis, tumor progression and response to therapy as a mere bystander, a notion that has now been invalidated. On one hand, components of the immune system, such a B lymphocytes and macrophages, have been shown to facilitate inflammation-driven carcinogenesis,1-3 while others, such as CD8+ T and natural killer (NK) cells, ensure a constant barrier against oncogenesis (immunosurveillance) that malignant precursors must break to develop tumors.4 On the other hand, the therapeutic efficacy of several anticancer regimens, including conventional chemotherapeutics as well as targeted agents, appear to rely (at least in part) on the activation of innate or cognate immune effector mechanisms.5 Thus, the abundance of intratumoral CD8+ and memory T cells has recently been shown to dramatically affect the clinical outcome in multiple oncological settings.6-9 Along with this conceptual shift, which occurred during the last three decades, therapeutic interventions aimed at activating the immune system against tumors begun to attract an ever increasing interest, from both researchers and clinicians. The promising field of anticancer immunotherapy had been established.10

Nowadays, cancer immunotherapy can be subdivided into three major branches: (1) approaches for the relatively “unselective” stimulation of the immune system against tumors, (2) anticancer vaccines (including protein-, peptide- and cell-based vaccines), and (3) adoptive cell transfer (ACT) protocols.11 Immunostimulatory interventions are exemplified by the systemic administration of lymphocyte-targeting growth factors such as interleukin-2 (IL-2), other pro-immunogenic cytokines such as interferon α (IFNα), or compounds that block immunosuppressive mechanisms, including monoclonal antibodies that are specific for the cytotoxic T lymphocyte antigen 4 (CTLA4) or chemotherapeutics that selectively depletes immunoregulatory cell populations. Immunostimulatory agents given as monotherapy have been associated with consistent rates of tumor regression in melanoma and renal carcinoma patients,12-15 perhaps because these cancers are able to elicit per se elevated levels of antitumor lymphocytes. Of note, several anticancer agents that are currently used in the clinic also mediate immunostimulatory effects, either by actively triggering immune effector mechanisms or by selectively inhibiting/killing immunosuppressive cells such as FOXP3+ regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs).5,16 These chemotherapeutics might de facto function as combination therapies, mediating both a cytotoxic/cytostatic effect on tumor cells and a stimulatory effect on the immune system.

Vaccines constitute a very appealing approach to cancer immunotherapy, presumably because they would be relatively easy to administer, cheap (especially in the case of peptidic vaccines) and virtually devoid of side effects.17 Nonetheless, cancer vaccines, encompassing both peptides and dendritic cell (DC)-based approaches, so far have failed to meet the high expectations that they had raised, being associated with modest and often non-reproducible clinical benefits.11 Perhaps, this can be attributed to the fact that end-stage cancer patients often exhibit immune defects that can compromise their ability to mount a vaccine-driven antitumor response. One notable exception is provided by sipuleucel-T (Provenge®), a DC-based vaccine that has been granted FDA approval for the treatment of asymptomatic or minimally symptomatic, metastatic castration-resistant (hormone refractory) prostate cancer.18-20 In addition, promising results have been observed in prostate cancer patients receiving prostate-specific antigen (PSA)-targeted poxviral vaccines (PROSTVAC-FS),21 as well as in melanoma patients treated with a peptidic vaccine combined with high-dose IL-2.22

ACT has emerged as an effective form of immunotherapy, with rates of complete durable responses (in specific clinical settings) as high as 40%.23,24 As a note, ACT must be conceptually differentiated from other cell-based immunotherapies, including the re-infusion of autologous DCs pulsed ex vivo with tumor antigens or tumor cell lysates (aimed at eliciting an anticancer T cell response in vivo) and the infusion of allogeneic T and NK cells (aimed at obtaining a curative graft-versus-disease effect).25,26

No ACT-based approach is currently approved by FDA for use in humans. In this Trial Watch, we will briefly review the scientific rationale behind ACT and discuss the progress of recent clinical studies evaluating the safety and efficacy of cell immunotherapy in oncological settings.

Scientific Background

ACT entails the (re)introduction into a conditioned patient of large amounts (often up to 1011) of lymphocytes exhibiting antitumor activity. When possible, notably in the case of melanoma and renal cell carcinoma (RCC) patients (which spontaneously manifest high number of antitumor lymphocytes), the starting material for ACT is constituted by a surgery specimen or biopsy, from which tumor-infiltrating lymphocytes (TILs) are isolated and (in some instances) selected for T-cell receptor (TCR) specificity.27 Before reinfusion, such lymphocytes are expanded ex vivo in the presence of IL-2 and other growth factors, and optionally activated with immunostimulatory compounds such as anti-CD3 antibodies, alone or combined with tumor-specific antigens.28 This said, melanoma and RCC constitute relatively privileged settings for immunotherapy, as suggested by the high rate of spontaneous TILs and by fact that immunostimulatory interventions alone are efficient against these tumors but not others.12-15

To extend the benefits of ACT to other types of cancer that are not associated with an intense endogenous immune response, genetic engineering can be employed to convert circulating lymphocytes into cells that exhibit antitumor activity. Thus, normal lymphocytes are isolated from the peripheral blood and genetically manipulated for the expression of TCRs that recognize tumor antigens with high affinity. The implementation of this strategy requires the isolation of very few endogenous cells with antitumor activity, from which rearranged TCR genes can be cloned and incorporated into highly efficient retroviral or lentiviral vectors.11 Upon transduction newly generated antitumor cells are amplified and treated similar to their tumor-derived counterparts. Importantly, as the TIL phenotype has been shown to affect both their in vivo persistence and their clinical efficacy,29-32 genetic engineering can be employed to confer additional features to lymphocytes, including (1) increased in vivo persistence, owing to the (over)expression of anti-apoptotic proteins like BCL-233 or telomerase, the enzyme that prevents the senescence-associated physiological attrition of telomeres;32 (2) increased proliferative potential, due to the (over)expression of growth factors, like IL-2, or their receptors;34 (3) enhanced homing to tumor sites, as a result of the expression of molecules involved in trafficking such as CD62L and CCR7;35 (4) enhanced antitumor potential, upon the (over)expression of co-receptors such as CD8 or co-stimulatory molecules like CD80 or the blockade of potentially immunosuppressive signals, such as those mediated by transforming growth factor β (TGFβ).36 Finally, as an alternative to TCRs, the so-called chimeric antigen receptors (CARs) can be employed. These chimeric receptors bind to tumor antigens via antibody-derived complementarity-determining regions (CDRs), yet are coupled to the intracellular machinery for TCR signaling, de facto mediating the functional activation of T cells even when antigens are not displayed in the context of MHC molecules.37

In the context of genetically engineered T cells, the choice of the tumor antigen specifically recognized by the TCR (or CAR) is critical, as it dictates both the efficacy and the safety of ACT.38 So far, encouraging results have been obtained with TCRs recognizing the melanocyte-specific markers MART1,39,40 MELOE-141,42 and gp100,43,44 the carcinoembryonic antigen (CEA),45 and cancer-testes antigens (which are expressed by a variety of epithelial cancers)46 such as NY-ESO-1, MAGE-A1 and MAGE-A3,47-50 as well as with CARs targeting the B-cell antigen CD1951-53 and the vascular endothelial growth factor receptor 2 (VEGFR2) (Table 1).54

Table 1. Examples of tumor antigens exploitable for adoptive cell transfer.

Antigen Tumor type Notes Ref.
CEA
Colorectal cancer
Preferentially expressed by gastrointestinal tumors
45
CD19
CLL
Lymphoma
B cell-specific antigen
5153
gp100
Melanoma
Melanocyte-specific antigen
43, 44
MAGE-1
Multiple epithelial malignancies
Cancer-testes antigen
46, 49
MAGE-A3
Multiple epithelial malignancies
Cancer-testes antigen
46, 50
MART-1
Melanoma
Melanocyte-specific antigen
39, 40
MELOE-1
Melanoma
Melanocyte-specific antigen
41, 42
NY-ESO-1
Melanoma
Synovial cell carcinoma
Cancer-testes antigen
47, 48
VEGFR2 Multiple types of solid tumors Overexpressed in tumor vasculature 54

Abbreviations: CEA, carcinoembryonic antigen; CLL, chronic lymphocytic leukemia; VEGFR2, vascular endothelial growth factor receptor 2.

In early trials, upon ex vivo expansion and activation, autologous TILs were re-administered to virtually untreated patients, resulting in transitory tumor regression but no durable responses.55 Perhaps, this was due to the fact that inoculated cells are subjected to a consistent degree of immunosuppression by endogenous Tregs and MDSCs, and normally fail to persist over long periods in vivo. Moreover, endogenous B, T and NK cells might compete with re-infused TILs for limited amounts of critical cytokines, including IL-7 and IL-15, a phenomenon known as “cytokine sink.”56,57 To circumvent these critical issues, pre-conditioning regimens based on cyclophosphamide (an alkylating agent that at high doses exert consistent immunosuppressive effects), fludarabine (a nucleoside analog commonly used for the therapy of hematological malignancies) and total body irradiation have been developed, resulting in different extents of lymphodepletion.58 Importantly, the intensity of lymphodepletion has been shown to directly correlate with ACT antitumor efficacy,58 leading to the introduction of preconditioning lymphodepletion into the clinical practice for ACT.

Re-infusion protocols have also been progressively refined to improve the persistence of TILs in vivo and to exacerbate their antitumor efficacy. Thus, although the co-infusion of cells with IL-2 is now a routine approach, several other possibilities are being explored. IL-2 has indeed been shown to correct the intrinsic anergy or TILs,59 and to promote the expansion of antitumor T cells in vivo.60 However, recent results indicate that this cytokine also stimulate immunosuppressive cell populations including Tregs,61-63 a phenomenon that may compromise its clinical benefits. New approach include, but are not limited to: (1) the co-infusion of cytokines other than IL-2, encompassing IL-7, IL-12, IL-15 and IFNγ, which stimulate immune effector and memory functions;64-67 (2) the co-infusion of angiogenesis inhibitors, which facilitate the extravasation of re-infused cells into the tumor;68 (3) the co-infusion of Toll-like receptor (TLR) agonists, to limit endogenous immunosuppression;69 and (4) the co-infusion of immunostimulatory chemotherapeutics, such as metronomic cyclophosphamide.5,70

Recently, ACT attempts based on B cells and NK cells have been investigated. On one hand, adoptively transferred B cells that exhibit antitumor activity in vitro reportedly mediate the generation of T-cell responses against xenografted breast cancer in mice.71 However, B cell-based ACT protocols have not yet been evaluated in clinical settings, perhaps due to the fact that B cells also mediate immunosuppressive and pro-tumorigenic effects, at least in some models of carcinogenesis.1,3 On the other hand, in spite of encouraging preclinical results and of the established efficacy of allogeneic NK cells for the therapy of acute myeloid leukemia (AML),72-74 NK cells failed to mediate antitumor effects in metastatic melanoma and RCC patients,75 perhaps owing to their limited persistence in vivo.76 Conversely, promising results have been obtained with the infusion of so-called “young TILs,” i.e., unselected, minimally cultured, bulk TILs whose production is relatively rapid and does not involve individualized tumor-reactivity screening steps.77-79 By reducing the costs and technical constraints that are associated with the ex vivo amplification and activation of TILs, this new approach may de facto increase the number of centers that will be able to offer ACT immunotherapy to eligible cancer patients. Of note, while the clinical efficacy of ACT is largely believed to depend on CD8+ T cells, the infusion of CD4+ T cells alone also mediates durable responses in melanoma patients.48 The cellular and molecular circuitries that underlie these observations remain to be precisely elucidated. As a possibility, infused cells (be they CD8+ of CD4+ cells) may initiate antitumor responses that result in the expansion of the tumor-specific TCR repertoire, and hence in the elicitation of robust immune reaction against cancer cells.48,80 Recent preclinical data demonstrate that oncogene-targeting T cells are superior to oncogene-specific drugs in the eradication of oncogene-addicted tumors, as the latter (but not the former) leave the tumor vasculature intact, allowing for the generation of resistant tumor clones.81 Perhaps, this superiority relies on the paracrine effects of IFNγ secreted by T cells, provoking the destruction of tumor vessels and impeding the growth or resistant cells.

Al least theoretically, the most prominent side effects of ACT relate to: (1) the specificity of re-infused cells, which might destroy normal tissues expressing the same antigen recognized by genetically engineered TCRs (or CARs);82 (2) the secretion by re-infused TILs of large amounts of cytokines/chemokines;83,84 (3) the possibility that transduced TCR chains might recombine with endogenous ones, resulting in the acquisition of unwarranted antigen specificity and graft-versus-host disease.85 So far, however, only a few cases of severe/lethal adverse reactions have been reported,45,83,84 suggesting that, in the vast majority of settings, ACT constitutes a safe clinical procedure.

Published Clinical Trials

Since the advent of ACT, the efficacy and safety of this intervention has been evaluated in multiple oncological settings, and the results of these early (Phase I/II) clinical studies have been published in some 30 high impact papers (Table 2).

Table 2. Published clinical trials evaluating adoptive cell transfer in cancer patients.

Site Tumor type(s) Phase Notes Ref.
Hematological neoplasms
BCL
I
IL-2-activated T cells + IL-2
2 PRs + 5 SDs out of 7 patients
103
CLL
I
Anti-CD19 CAR-engineered T cells + IL-2
1 PD, 2 SDs and 3 PRs out of 9 patients
51
CLL
Follicular lymphoma
LCL
MCL
SLL
I/II
Anti-CD19 CAR-engineered T cells + IL-2
1 CR, 1 SD, 6 ORs out of 8 patients
52, 106
Multiple myeloma
Plasma cell neoplasms
I/II
CD3/CD28-costimulated T cells ± vaccine upon HSCT
No ORs but some immunological responses
105
NHL
I
CD3/CD28-costimulated T cells following HSCT
5 CRs, 7 PRs, 4 SDs out of 16 patients
Transient dose-dependent infusion toxicities
102
Kidney
Renal cell carcinoma
I
CD3/CD28-costimulated T cells + IL-2
Some metastatic regression
Mild toxicity
96
Renal cell carcinoma
II
CD3-activated, dead tumor cells + Calmette-Guerin
bacillus-primed CD4+ cells + IL-2
9 CRs + 5 PRs out of 39 patients
95
Multiple tumors
Metastatic melanoma
Metastatic SCC
II
Anti-NY-ESO-1 TCR-engineered T cells + IL-2
ORs in 4/6 SCC patients and 5/11 melanoma patients
47
Advanced solid tumors
NHL
I
CD3-stimualted CD4+ cells + IL-2
1 CR, 2 PRs and 8 MRs out of 31 patients
101
Skin
Melanoma
n.a.
MART-1-specific CTLs generated in vitro using aAPCs
1 CR, 3 PRs, 3 SDs out of 10 patients
Some IL-2- and lymphopenia-associated toxicity
94
I
CD8+-enriched young TILs ± myeloablative lymphodepletion
9/33 ORs (3 CRs) without TBI; 11/23 ORs (2 CRs) with TBI
79
Metastatic melanoma
I
TILs + IL-2 upon non-myeloblative lymphodepletion
18 ORs (3 CRs + 15 PRs) out of 35 patients
Some IL-2- and lymphopenia-associated toxicity
86
I
Anti-MART-1 TCR-engineered PBMCs
2 ORs out of 15 patients and durable engraftment
43
I
MART-1-specific cells generated upon co-culture
with MART-1-pulsed DCs + low dose IL-2
3 ORs out of 11 patients
Mild (grade 1–2) adverse effects
87
I
Autologous CTLs upon previous fludarabine or not
3 MRs or SDs out of 9 patients
Fludarabine improved in vivo persistence
104
I
TILs + IL-2 upon non-myeloblative lymphodepletion
20 CRs out of 93 patients, which exhibited 100% 3 y survival
23
I/II
IL-2-engineered TILs + IL-2
1 PR out of 12 patients and persistent IL-2 expression
88
I/II
MART-1-specific T cells + IL-2 and IFNα
6 ORs (2 LTCRs) out of 14 patients
91
I/II
PBMC-derived T cells generated upon co-culture
with melanoma cells + low dose IFNα
1 CR, 1 PR, 3 SDs out of 10 patients
93
II
Autologous TILs + IL-2 ± myeloablative lymphodepletion
Up to 72% ORs (with TBI) out of 93 patients
1 death
89
II
Anti-MART-1 and anti-gp100 human or
murine TCR-engineered PBMCs
30% (human TCR) and 19% (murine TCR) ORs
Toxicity to normal melanocytes in the ear and skin
82
II
Young TILs + IL-2
10 ORs and 4 SDs out of 20 patients
Transient and manageable toxicity
77
II
Autologous TILs or MART1-specific
PBMC-derived T cells + IL-2
9 CRs and PRs in the brain out of 26 patients
1 subarachnoid hemorrhage without consequences
92
Various Colorectal cancer
I/II
Anti-CEA murine TCR-engineered T cells
1 ORs out of 3 patients
Transient colitis in all patients
45
HCC
I
iDCs, CIK cells and CTLs + tumor lysate-pulsed DCs
Early increase in the CD8+/FOXP3+ ratio
97
HNSCC
II
NKT cells in combination with alphaGalCer-pulsed DCs
5 ORs out of 10 patients
100
Neuroblastoma
I
Anti-GD2 CAR-engineered EBV-specific CTLs
ORs in 50% of patients
99
Ovarian cancer I Anti-FR TCR-engineered T cells + IL-2
No anticancer responses
IL-2 related mild (grade 3–4) toxicity in 5/8 patients
98

Abbreviations: aAPC, artificial antigen-presenting cell; BCL, B cell lymphoma; CAR, chimeric antigen receptor; CEA, carcinoembryonic antigen; CIK, cytokine-induced killer; CLL, chronic lymphocytic leukemia; CR, complete response; CTL, cytotoxic lymphocyte; DC, dendritic cell; EBV, Epstein-Barr virus; FR, folate receptor; HCC, hepatocellular carcinoma; HNSCC, head and neck squamous cell carcinoma; HSCT, hematopoietic stem cell transplantation; iDC, immature DC; LCL, large cell lymphoma; LTCR, long-term complete response; MCL, mantle cell lymphoma; n.a., not available; NHL, non-Hodgkin lymphoma; NK, natural killer: NKT, Vα24 NK; IFN, interferon; IL, interleukin; MR, minor response; n.a., not available; OR, objective response; PBMC, peripheral blood mononuclear cell; PD, progressive disease; PR, partial response; SCC, synovial cell carcinoma; SD, stable disease; SLL, small lymphocytic leukemia; TBI, total body irradiation; TIL, tumor infiltrating lymphocyte.

One half of these studies were performed in small cohorts of (often metastatic) melanoma patients,23,43,47,52,77,82,86-94 while the other half tested ACT in clinical settings as diverse as hematological malignancies,23,43,47,52,77,82,86-90,92-94 RCC,47,95,96 hepatocellular carcinoma,97 ovarian cancer,98 neuroblastoma,99 metastatic colorectal carcinoma,45 and head and neck squamous cell carcinoma.100

In about one third of these studies, patients were re-infused with autologous cells that had been non-specifically activated in vitro, for instance by the administration of IL-2 or anti-CD3 antibodies, alone or in combination with anti-CD28 antibodies.23,86,89,92,96,101-105 Alternatively, patients received lymphocytes that had been specifically activated against the tumor by the ex vivo administration of dead cancer cells in the presence of the Calmette-Guerin bacillus,95 or by the co-culture with living tumor cells,93 with DCs pulsed with tumor antigens,87,91 with DCs pulsed with cancer cell lysates,97 or with an artificial antigen-presenting cell (APC) system that can educate antitumor lymphocytes to acquire both a central memory and an effector memory phenotype.94 In two studies, patients received young TILs,77,79 whereas in one trial ex vivo expanded Vα24 NK (NKT) cells were intravenously administered in tumor-feeding arteries, in conjunction with the nasal administration of αGalCer-pulsed APCs.100 In the remaining studies, the re-infused material consisted in genetically modified cells, including peripheral blood mononuclear cell (PBMC)-derived T cells engineered to express tumor antigen-specific TCRs,43,45,47,82,92,98 CARs51,52,99,106 or IL-2.88

In most cases, patients received classical lymphodepleting regimens based on cyclophosphamide + fludarabine, alone or in combination with total body irradiation,23,47,52,79,82,86,89,92,101,104,106 and were given cells together with high-dose IL-2.23,25,43,45,47,51,52,77,79,82,86,88,89,91,92,94-98,100-102,104-106 As an alternative, cells were co-infused with a course of low-dose IL-2 over 6 d,87 or low-dose IFNγ.93

In two of these trials no antitumor effects were observed,97,98 and in two other studies ACT was associated with limited (though assessable) therapeutic responses.96,105 Apart from these notable exceptions, the results of the clinical trials conducted so far on ACT are very encouraging, reporting response rates as high as 70% and a very low incidence of severe side effects (Table 2). Although they were often performed in small patient cohorts and the therapeutic protocols often differed from trial to trial, these phase I/II studies demonstrate that ACT is efficient and safe for the treatment of some types of cancer, in particular melanoma. This paved the way for ongoing studies that evaluate alternative ACT protocols or the applicability of ACT to other oncological settings.

Ongoing Clinical Trials

At present, there are around 35 ongoing, early (Phase I/II) clinical trials that test the safety and efficacy of ACT in oncological indications (source www.clinicaltrials.gov). Thirteen of these studies are performed in melanoma and RCC patients. In addition, ACT, alone or combined with established procedures, is being evaluated as a therapy for tumors as different as hematological malignancies (including multiple myeloma and several types of leukemia and lymphoma), sarcomas, cancers of the reproductive trait (including cervical, Fallopian tube, ovarian and prostate cancer), neoplasms of the central nervous system (including malignant glioma, glioblastoma, medulloblastoma and neuroectodermal tumors), as well as nasopharyngeal, breast, lung and hepatocellular carcinomas (Table 3).

Table 3. Ongoing clinical trials evaluating adoptive cell transfer in cancer patients*.

Site Tumor type(s) Phase Notes Ref.
Breast
HER2+ breast cancer
I/II
Autologous ex vivo expanded HER2-specific T cells
NCT00791037
I
Autologous ex vivo expanded HER2-specific
T cells + cyclophosphamide
NCT01219907
TNBC
n.a.
CIK cells ± DCs
NCT01232062
NCT01395056
CNS
Glioblastoma
I
CMV-activated T cells alone or
combined with a DC-based vaccine
NCT00693095
Malignant glioma
I
Autologous CD8+ T cells expressing inducible suicide fusion protein and an IL-13 chimeric immunoreceptor
NCT00730613
Medulloblastoma
Neuroectodermal tumors
I/II
Tumor-specific T cells alone or
combined with a DC-based vaccine
NCT01326104
Hematological neoplasms
AML
CML
MDS
II
Autologous CIK cells ± imatinib
NCT00815321
BCL
I
Anti-CD19 CAR-engineered CD8+ cells + IL-2 upon HSCT
NCT00968760
BCL
B-CLL
Multiple myeloma
I
Anti-κ light chain CAR-engineered T cells
NCT00881920
CLL
I
Anti-CD19 CAR-engineered CD8+ cells after chemotherapy
NCT01416974
NHL
I/II
Anti-CD19 CAR-engineered CD8+ cells upon
myeloablative conditioning and PBSCT
NCT01318317
Multiple myeloma
I
CD3/CD28-costimulated T cells following HSCT
NCT01239368
Multiple myeloma
I/II
Vaccine primed-autologous T cells following HSCT
NCT00834665
Multiple
sites
Breast carcinoma
Colorectal carcinoma
Gastric cancer
Lung cancer
Ovarian cancer
Pancreatic cancer
I
Anti-CEA TCR-engineered T cells + IL-2
NCT01212887
Breast carcinoma
HCC
Nasopharyngeal cancer
I
CD8+-enriched young TILs + IL-2
NCT01462903
Metastatic melanoma
RCC
I/II
Anti-VEGFR2 CAR-engineered CD8+ cells + IL-2
NCT01218867
Reproductive tract
Cervical neoplasms
I/II
Low-dose radiotherapy followed by autologous TILs
NCT01194609
Fallopian tube cancer
Ovarian cancer
Primary peritoneal cancer
I
CD3/CD28-costimulated vaccine-primed
autologous T cells
NCT01312376
Prostate cancer
I
Anti-PSMA TCR-engineered T cells
NCT01140373
Skin
Metastatic melanoma
II
TILs + IL-2 upon non-myeloblative lymphodepletion
NCT00604136
I/II
MART-1-specific TILs + IL-2 followed by IL-2 and IFNα
NCT00720031
I/II
NY-ESO-1-specific TILs + IL-2 ± ipilimumab
NCT00871481
II
Anti-MART-1 TCR-engineered PBMCs plus MART-126–35 peptide-pulsed DCs + IL-2
NCT00910650
I
Melanoma reactive (DMF5) TILs + IL-2
NCT00924001
n.a.
TILs + IL-2 upon non-myeloblative lymphodepletion
NCT01005745
I/II
IFNγ-engineered TILs + IL-2
NCT01082887
II
CD8+-enriched young TILs + IL-2
NCT01118091
I/II
IL-12-engineered TILs + IL-2
NCT01236573
II
Young TILs + IL-2
NCT01319565
I/II
Young TILs + IL-15
NCT01369888
II
TILs + IL-2 upon non-myeloblative lymphodepletion
NCT01468818
Soft tissues Adult liposarcoma
Adult soft tissue sarcoma
Adult synovial sarcoma
I NY-ESO-1-specific CD8+ T cells + IL-2, upon conditioning with low-dose IFNγ and cyclophosphamide NCT01477021

Abbreviations: AML, acute myeloid leukemia; BCL, B cell lymphoma; B-CLL, B cell chronic lymphocytic leukemia; CAE, carcinoembryonic antigen; CAR, chimeric antigen receptor; CIK, cytokine-induced killer; CML, chronic myeloid leukemia; CMV, cytomegalovirus; CNS, central nervous system; DC, dendritic cell; HCC, hepatocellular carcinoma; HSCT, hematopoietic stem cell transplantation; IFN, interferon; IL, interleukin; MDS, myelodysplastic syndrome; n.a., not available; NHL, non-Hodgkin lymphoma; PBMC, peripheral blood mononuclear cell; PBSCT, peripheral blood stem cell transplantation; PSMA, prostate-specific membrane antigen; TBI, total body irradiation; TCR, T-cell receptor; TILs, tumor-infiltrating lymphocytes; TNBC, triple negative breast cancer; VEGFR2, vascular endothelial growth factor receptor 2. *started after January, 1st 2008 and not completed or terminated at the day of submission.

In many cases, the infused material consists of ex vivo expanded and activated TILs, though the use of young TILs (NCT01118091; NCT01319565; NCT01369888) and of cytokine-induced killer (CIK, CD3+CD56+ non-MHC-restricted, NK-like T lymphocytes)107 cells (NCT00815321; NCT01232062; NCT01395056) is also being investigated. Sometimes, TILs are collected a few weeks after the administration of tumor antigen-specific vaccines (NCT00791037; NCT00834665; NCT01312376). Moreover, distinct approaches of genetic engineering are being undertaken, including the production of lymphocytes expressing tumor antigen-specific TCRs (NCT00720031; NCT00871481; NCT00910650; NCT01212887), CARs (NCT00968760; NCT01218867; NCT01318317; NCT01416974) IL-12 (NCT01236573) or IFNγ (NCT01082887). One particularly interesting study entails the engineering of lymphocytes for the expression of an inducible suicide fusion protein, which might be employed to resolve ACT-related toxicity (NCT00730613). Conditioning regimens largely overlap with those employed in previous successful studies, with a high prevalence of non-myeloablative lymphodepletion (cyclophosphamide + fludarabine), alone or combined with total body irradiation. In one instance, cells are infused in the absence of conditioning and after three cycles of low-dose (sub-efficient) radiotherapy, aimed at stimulating immune responses (NCT01194609). Most often patients receive cells together with high-dose IL-2, though the co-infusion of IL-15 (NCT01369888) or DC-based vaccines (NCT00693095; NCT00910650; NCT01326104) is also being tested.

Concluding Remarks

Thanks to the work from several laboratories worldwide, our knowledge on the molecular and cellular circuitries that underlie the long-term effectiveness of ACT has greatly advanced. ACT based on autologous TILs has already been associated with consistent rates of durable remissions in exquisitely immunosensitive cancers such as melanoma. Moreover, the use of genetically engineered circulating T cells constitutes a promising approach for the treatment of several other (solid and hematological) malignancies. Results from ongoing trials will clarify whether the clinical benefits of ACT truly extend to poorly immunosensitive tumors. Unfortunately, ACT is far from becoming a routine clinical practice, as its implementation is laborious and associated with elevated costs. In this sense, the development of simplified techniques for the ex vivo expansion, activation and genetic engineering of lymphocytes might allow an increasing number of cancer centers to offer ACT as a therapeutic option.

Acknowledgments

Authors are supported by the Ligue contre le Cancer (e´quipes labelise´es), AXA Chair for Longevity Research, Cance´ropôle Ile-de-France, Institut National du Cancer (INCa), Fondation Bettencourt-Schueller, Fondation de France, Fondation pour la Recherche Me´dicale, Agence National de la Recherche, the European Commission (Apo-Sys, ArtForce, ChemoRes. Death-Train) and the LabEx Immuno-Oncology.

Glossary

Abbreviations:

ACT

adoptive cell transfer

AML

acute myeloid leukemia

APC

antigen-presenting cell

CAR

chimeric antigen receptor

CDR

complementarity-determining region

CIK

cytokine-induced killer

CTLA4

cytotoxic T lymphocyte antigen 4

DC

dendritic cell

IFNγ

interferon γ

IL

interleukin

NK

natural killer

NKT

Vα24 NK

MDSC

myeloid-derived suppressor cell

PBMC

peripheral blood mononuclear cell

PSA

prostate-specific antigen

RCC

renal cell carcinoma

TCR

T-cell receptor

TGFβ

transforming growth factor β

TILs

tumor-infiltrating lymphocytes

TLR

Toll-like receptor

Tregs

FOXP3+ regulatory T cells

VEGFR2

vascular endothelial growth factor receptor 2

Footnotes

References

  • 1.de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell. 2005;7:411–23. doi: 10.1016/j.ccr.2005.04.014. [DOI] [PubMed] [Google Scholar]
  • 2.Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70:7465–75. doi: 10.1158/0008-5472.CAN-10-1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Schioppa T, Moore R, Thompson RG, Rosser EC, Kulbe H, Nedospasov S, et al. B regulatory cells and the tumor-promoting actions of TNF-α during squamous carcinogenesis. Proc Natl Acad Sci U S A. 2011;108:10662–7. doi: 10.1073/pnas.1100994108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006;6:715–27. doi: 10.1038/nri1936. [DOI] [PubMed] [Google Scholar]
  • 5.Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secrete ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626. [DOI] [PubMed] [Google Scholar]
  • 6.Pagès F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353:2654–66. doi: 10.1056/NEJMoa051424. [DOI] [PubMed] [Google Scholar]
  • 7.Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–4. doi: 10.1126/science.1129139. [DOI] [PubMed] [Google Scholar]
  • 8.Mlecnik B, Tosolini M, Kirilovsky A, Berger A, Bindea G, Meatchi T, et al. Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol. 2011;29:610–8. doi: 10.1200/JCO.2010.30.5425. [DOI] [PubMed] [Google Scholar]
  • 9.Mlecnik B, Bindea G, Pagès F, Galon J. Tumor immunosurveillance in human cancers. Cancer Metastasis Rev. 2011;30:5–12. doi: 10.1007/s10555-011-9270-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–15. doi: 10.1038/nm1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rosenberg SA. Cell transfer immunotherapy for metastatic solid cancer--what clinicians need to know. Nat Rev Clin Oncol. 2011;8:577–85. doi: 10.1038/nrclinonc.2011.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Robert C, Thomas L, Bondarenko I, O’Day S, M D JW, Garbe C, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
  • 14.Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghausen SE, et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med. 1985;313:1485–92. doi: 10.1056/NEJM198512053132327. [DOI] [PubMed] [Google Scholar]
  • 15.Smith FO, Downey SG, Klapper JA, Yang JC, Sherry RM, Royal RE, et al. Treatment of metastatic melanoma using interleukin-2 alone or in conjunction with vaccines. Clin Cancer Res. 2008;14:5610–8. doi: 10.1158/1078-0432.CCR-08-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tartour E, Pere H, Maillere B, Terme M, Merillon N, Taieb J, et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev. 2011;30:83–95. doi: 10.1007/s10555-011-9281-4. [DOI] [PubMed] [Google Scholar]
  • 17.Finn OJ. Cancer vaccines: between the idea and the reality. Nat Rev Immunol. 2003;3:630–41. doi: 10.1038/nri1150. [DOI] [PubMed] [Google Scholar]
  • 18.Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. IMPACT Study Investigators Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 19.Small EJ, Schellhammer PF, Higano CS, Redfern CH, Nemunaitis JJ, Valone FH, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. 2006;24:3089–94. doi: 10.1200/JCO.2005.04.5252. [DOI] [PubMed] [Google Scholar]
  • 20.Beer TM, Bernstein GT, Corman JM, Glode LM, Hall SJ, Poll WL, et al. Randomized trial of autologous cellular immunotherapy with sipuleucel-T in androgen-dependent prostate cancer. Clin Cancer Res. 2011;17:4558–67. doi: 10.1158/1078-0432.CCR-10-3223. [DOI] [PubMed] [Google Scholar]
  • 21.Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, et al. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28:1099–105. doi: 10.1200/JCO.2009.25.0597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 2011;364:2119–27. doi: 10.1056/NEJMoa1012863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17:4550–7. doi: 10.1158/1078-0432.CCR-11-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gattinoni L, Powell DJ, Jr., Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol. 2006;6:383–93. doi: 10.1038/nri1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Shlomchik WD. Graft-versus-host disease. Nat Rev Immunol. 2007;7:340–52. doi: 10.1038/nri2000. [DOI] [PubMed] [Google Scholar]
  • 26.Tacken PJ, de Vries IJ, Torensma R, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol. 2007;7:790–802. doi: 10.1038/nri2173. [DOI] [PubMed] [Google Scholar]
  • 27.Bouquie´ R, Bonnin A, Bernardeau K, Khammari A, Dre´no B, Jotereau F, et al. A fast and efficient HLA multimer-based sorting procedure that induces little apoptosis to isolate clinical grade human tumor specific T lymphocytes. Cancer Immunol Immunother. 2009;58:553–66. doi: 10.1007/s00262-008-0578-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yun YS, Hargrove ME, Ting CC. In vivo antitumor activity of anti-CD3-induced activated killer cells. Cancer Res. 1989;49:4770–4. [PubMed] [Google Scholar]
  • 29.Aebersold P, Hyatt C, Johnson S, Hines K, Korcak L, Sanders M, et al. Lysis of autologous melanoma cells by tumor-infiltrating lymphocytes: association with clinical response. J Natl Cancer Inst. 1991;83:932–7. doi: 10.1093/jnci/83.13.932. [DOI] [PubMed] [Google Scholar]
  • 30.Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K, Cardones AR, Finkelstein SE, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A. 2005;102:9571–6. doi: 10.1073/pnas.0503726102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Schwartzentruber DJ, Hom SS, Dadmarz R, White DE, Yannelli JR, Steinberg SM, et al. In vitro predictors of therapeutic response in melanoma patients receiving tumor-infiltrating lymphocytes and interleukin-2. J Clin Oncol. 1994;12:1475–83. doi: 10.1200/JCO.1994.12.7.1475. [DOI] [PubMed] [Google Scholar]
  • 32.Zhou J, Shen X, Huang J, Hodes RJ, Rosenberg SA, Robbins PF. Telomere length of transferred lymphocytes correlates with in vivo persistence and tumor regression in melanoma patients receiving cell transfer therapy. J Immunol. 2005;175:7046–52. doi: 10.4049/jimmunol.175.10.7046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kalbasi A, Shrimali RK, Chinnasamy D, Rosenberg SA. Prevention of interleukin-2 withdrawal-induced apoptosis in lymphocytes retrovirally cotransduced with genes encoding an antitumor T-cell receptor and an antiapoptotic protein. J Immunother. 2010;33:672–83. doi: 10.1097/CJI.0b013e3181e475cd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liu K, Rosenberg SA. Transduction of an IL-2 gene into human melanoma-reactive lymphocytes results in their continued growth in the absence of exogenous IL-2 and maintenance of specific antitumor activity. J Immunol. 2001;167:6356–65. doi: 10.4049/jimmunol.167.11.6356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hinrichs CS, Borman ZA, Gattinoni L, Yu Z, Burns WR, Huang J, et al. Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood. 2011;117:808–14. doi: 10.1182/blood-2010-05-286286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zhang Q, Yang X, Pins M, Javonovic B, Kuzel T, Kim SJ, et al. Adoptive transfer of tumor-reactive transforming growth factor-beta-insensitive CD8+ T cells: eradication of autologous mouse prostate cancer. Cancer Res. 2005;65:1761–9. doi: 10.1158/0008-5472.CAN-04-3169. [DOI] [PubMed] [Google Scholar]
  • 37.Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86:10024–8. doi: 10.1073/pnas.86.24.10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Li LP, Lampert JC, Chen X, Leitao C, Popovicá J, Müller W, et al. Transgenic mice with a diverse human T cell antigen receptor repertoire. Nat Med. 2010;16:1029–34. doi: 10.1038/nm.2197. [DOI] [PubMed] [Google Scholar]
  • 39.Chhabra A, Yang L, Wang P, Comin-Anduix B, Das R, Chakraborty NG, et al. CD4+CD25- T cells transduced to express MHC class I-restricted epitope-specific TCR synthesize Th1 cytokines and exhibit MHC class I-restricted cytolytic effector function in a human melanoma model. J Immunol. 2008;181:1063–70. doi: 10.4049/jimmunol.181.2.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ray S, Chhabra A, Chakraborty NG, Hegde U, Dorsky DI, Chodon T, et al. UCLA-CALTECH-CHLA-USC-UCONN Consortium on Translational Program in Engineered Immunity MHC-I-restricted melanoma antigen specific TCR-engineered human CD4+ T cells exhibit multifunctional effector and helper responses, in vitro. Clin Immunol. 2010;136:338–47. doi: 10.1016/j.clim.2010.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Godet Y, Moreau-Aubry A, Guilloux Y, Vignard V, Khammari A, Dreno B, et al. MELOE-1 is a new antigen overexpressed in melanomas and involved in adoptive T cell transfer efficiency. J Exp Med. 2008;205:2673–82. doi: 10.1084/jem.20081356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Godet Y, Desfrançois J, Vignard V, Schadendorf D, Khammari A, Dreno B, et al. Frequent occurrence of high affinity T cells against MELOE-1 makes this antigen an attractive target for melanoma immunotherapy. Eur J Immunol. 2010;40:1786–94. doi: 10.1002/eji.200940132. [DOI] [PubMed] [Google Scholar]
  • 43.Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–9. doi: 10.1126/science.1129003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Morgan RA, Dudley ME, Yu YY, Zheng Z, Robbins PF, Theoret MR, et al. High efficiency TCR gene transfer into primary human lymphocytes affords avid recognition of melanoma tumor antigen glycoprotein 100 and does not alter the recognition of autologous melanoma antigens. J Immunol. 2003;171:3287–95. doi: 10.4049/jimmunol.171.6.3287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DA, Feldman SA, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19:620–6. doi: 10.1038/mt.2010.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer. 2005;5:615–25. doi: 10.1038/nrc1669. [DOI] [PubMed] [Google Scholar]
  • 47.Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29:917–24. doi: 10.1200/JCO.2010.32.2537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med. 2008;358:2698–703. doi: 10.1056/NEJMoa0800251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Willemsen R, Ronteltap C, Heuveling M, Debets R, Bolhuis R. Redirecting human CD4+ T lymphocytes to the MHC class I-restricted melanoma antigen MAGE-A1 by TCR alphabeta gene transfer requires CD8alpha. Gene Ther. 2005;12:140–6. doi: 10.1038/sj.gt.3302388. [DOI] [PubMed] [Google Scholar]
  • 50.Chinnasamy N, Wargo JA, Yu Z, Rao M, Frankel TL, Riley JP, et al. A TCR targeting the HLA-A*0201-restricted epitope of MAGE-A3 recognizes multiple epitopes of the MAGE-A antigen superfamily in several types of cancer. J Immunol. 2011;186:685–96. doi: 10.4049/jimmunol.1001775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–28. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116:4099–102. doi: 10.1182/blood-2010-04-281931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–33. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chinnasamy D, Yu Z, Theoret MR, Zhao Y, Shrimali RK, Morgan RA, et al. Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenic tumors in mice. J Clin Invest. 2010;120:3953–68. doi: 10.1172/JCI43490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988;319:1676–80. doi: 10.1056/NEJM198812223192527. [DOI] [PubMed] [Google Scholar]
  • 56.Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med. 2005;202:907–12. doi: 10.1084/jem.20050732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Klebanoff CA, Khong HT, Antony PA, Palmer DC, Restifo NP. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol. 2005;26:111–7. doi: 10.1016/j.it.2004.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wrzesinski C, Paulos CM, Kaiser A, Muranski P, Palmer DC, Gattinoni L, et al. Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother. 2010;33:1–7. doi: 10.1097/CJI.0b013e3181b88ffc. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tartour E, Latour S, Mathiot C, Thiounn N, Mosseri V, Joyeux I, et al. Variable expression of CD3-zeta chain in tumor-infiltrating lymphocytes (TIL) derived from renal-cell carcinoma: relationship with TIL phenotype and function. Int J Cancer. 1995;63:205–12. doi: 10.1002/ijc.2910630210. [DOI] [PubMed] [Google Scholar]
  • 60.Tartour E, Mathiot C, Fridman WH. Current status of interleukin-2 therapy in cancer. Biomed Pharmacother. 1992;46:473–84. doi: 10.1016/0753-3322(92)90005-R. [DOI] [PubMed] [Google Scholar]
  • 61.Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood. 2006;107:2409–14. doi: 10.1182/blood-2005-06-2399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V, et al. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med. 2011;365:2067–77. doi: 10.1056/NEJMoa1105143. [DOI] [PubMed] [Google Scholar]
  • 63.Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP, 3rd, et al. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med. 2011;365:2055–66. doi: 10.1056/NEJMoa1108188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mignot G, Ullrich E, Bonmort M, Me´nard C, Apetoh L, Taieb J, et al. The critical role of IL-15 in the antitumor effects mediated by the combination therapy imatinib and IL-2. J Immunol. 2008;180:6477–83. doi: 10.4049/jimmunol.180.10.6477. [DOI] [PubMed] [Google Scholar]
  • 65.Ullrich E, Bonmort M, Mignot G, Jacobs B, Bosisio D, Sozzani S, et al. Trans-presentation of IL-15 dictates IFN-producing killer dendritic cells effector functions. J Immunol. 2008;180:7887–97. doi: 10.4049/jimmunol.180.12.7887. [DOI] [PubMed] [Google Scholar]
  • 66.Liu DL, Håkansson CH, Seifert J. Immunotherapy in liver tumors: II. Intratumoral injection with activated tumor-infiltrating lymphocytes, intrasplenic administration of recombinant interleukin-2 and interferon alpha causes tumor regression and lysis. Cancer Lett. 1994;85:39–46. doi: 10.1016/0304-3835(94)90236-4. [DOI] [PubMed] [Google Scholar]
  • 67.Helms MW, Prescher JA, Cao YA, Schaffert S, Contag CH. IL-12 enhances efficacy and shortens enrichment time in cytokine-induced killer cell immunotherapy. Cancer Immunol Immunother. 2010;59:1325–34. doi: 10.1007/s00262-010-0860-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Dings RP, Vang KB, Castermans K, Popescu F, Zhang Y, Oude Egbrink MG, et al. Enhancement of T-cell-mediated antitumor response: angiostatic adjuvant to immunotherapy against cancer. Clin Cancer Res. 2011;17:3134–45. doi: 10.1158/1078-0432.CCR-10-2443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yang Y, Huang CT, Huang X, Pardoll DM. Persistent Toll-like receptor signals are required for reversal of regulatory T cell-mediated CD8 tolerance. Nat Immunol. 2004;5:508–15. doi: 10.1038/ni1059. [DOI] [PubMed] [Google Scholar]
  • 70.North RJ. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J Exp Med. 1982;155:1063–74. doi: 10.1084/jem.155.4.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Li Q, Lao X, Pan Q, Ning N, Yet J, Xu Y, et al. Adoptive transfer of tumor reactive B cells confers host T-cell immunity and tumor regression. Clin Cancer Res. 2011;17:4987–95. doi: 10.1158/1078-0432.CCR-11-0207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Pegram HJ, Jackson JT, Smyth MJ, Kershaw MH, Darcy PK. Adoptive transfer of gene-modified primary NK cells can specifically inhibit tumor progression in vivo. J Immunol. 2008;181:3449–55. doi: 10.4049/jimmunol.181.5.3449. [DOI] [PubMed] [Google Scholar]
  • 73.Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
  • 74.Velardi A, Ruggeri L, Mancusi A, Aversa F, Christiansen FT. Natural killer cell allorecognition of missing self in allogeneic hematopoietic transplantation: a tool for immunotherapy of leukemia. Curr Opin Immunol. 2009;21:525–30. doi: 10.1016/j.coi.2009.07.015. [DOI] [PubMed] [Google Scholar]
  • 75.Parkhurst MR, Riley JP, Dudley ME, Rosenberg SA. Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clin Cancer Res. 2011;17:6287–97. doi: 10.1158/1078-0432.CCR-11-1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Geller MA, Cooley S, Judson PL, Ghebre R, Carson LF, Argenta PA, et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy. 2011;13:98–107. doi: 10.3109/14653249.2010.515582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Besser MJ, Shapira-Frommer R, Treves AJ, Zippel D, Itzhaki O, Hershkovitz L, et al. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clin Cancer Res. 2010;16:2646–55. doi: 10.1158/1078-0432.CCR-10-0041. [DOI] [PubMed] [Google Scholar]
  • 78.Donia M, Junker N, Ellebaek E, Andersen MH, Straten PT, Svane IM. Characterization and comparison of “Standard” and “Young” tumor infiltrating lymphocytes for adoptive cell therapy at a Danish Translational Research Institution. Scand J Immunol. 2011 doi: 10.1111/j.1365-3083.2011.02640.x. in press. [DOI] [PubMed] [Google Scholar]
  • 79.Dudley ME, Gross CA, Langhan MM, Garcia MR, Sherry RM, Yang JC, et al. CD8+ enriched “young” tumor infiltrating lymphocytes can mediate regression of metastatic melanoma. Clin Cancer Res. 2010;16:6122–31. doi: 10.1158/1078-0432.CCR-10-1297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Vignard V, Lemercier B, Lim A, Pandolfino MC, Guilloux Y, Khammari A, et al. Adoptive transfer of tumor-reactive Melan-A-specific CTL clones in melanoma patients is followed by increased frequencies of additional Melan-A-specific T cells. J Immunol. 2005;175:4797–805. doi: 10.4049/jimmunol.175.7.4797. [DOI] [PubMed] [Google Scholar]
  • 81.Anders K, Buschow C, Herrmann A, Milojkovic A, Loddenkemper C, Kammertoens T, et al. Oncogene-targeting T cells reject large tumors while oncogene inactivation selects escape variants in mouse models of cancer. Cancer Cell. 2011;20:755–67. doi: 10.1016/j.ccr.2011.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114:535–46. doi: 10.1182/blood-2009-03-211714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Brentjens R, Yeh R, Bernal Y, Riviere I, Sadelain M. 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;18:666–8. doi: 10.1038/mt.2010.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–51. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bendle GM, Linnemann C, Hooijkaas AI, Bies L, de Witte MA, Jorritsma A, et al. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat Med 2010; 16:565-70, 1p following 70. [DOI] [PubMed]
  • 86.Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005;23:2346–57. doi: 10.1200/JCO.2005.00.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. J Clin Oncol. 2006;24:5060–9. doi: 10.1200/JCO.2006.07.1100. [DOI] [PubMed] [Google Scholar]
  • 88.Heemskerk B, Liu K, Dudley ME, Johnson LA, Kaiser A, Downey S, et al. 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]
  • 89.Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26:5233–9. doi: 10.1200/JCO.2008.16.5449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Disis ML, Bernhard H, Jaffee EM. Use of tumour-responsive T cells as cancer treatment. Lancet. 2009;373:673–83. doi: 10.1016/S0140-6736(09)60404-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Khammari A, Labarrière N, Vignard V, Nguyen JM, Pandolfino MC, Knol AC, et al. Treatment of metastatic melanoma with autologous Melan-A/MART-1-specific cytotoxic T lymphocyte clones. J Invest Dermatol. 2009;129:2835–42. doi: 10.1038/jid.2009.144. [DOI] [PubMed] [Google Scholar]
  • 92.Hong JJ, Rosenberg SA, Dudley ME, Yang JC, White DE, Butman JA, et al. Successful treatment of melanoma brain metastases with adoptive cell therapy. Clin Cancer Res. 2010;16:4892–8. doi: 10.1158/1078-0432.CCR-10-1507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Verdegaal EM, Visser M, Ramwadhdoebe´ TH, van der Minne CE, van Steijn JA, Kapiteijn E, et al. Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ T cells in combination with low-dose interferon-alpha. Cancer Immunol Immunother. 2011;60:953–63. doi: 10.1007/s00262-011-1004-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Butler MO, Friedlander P, Milstein MI, Mooney MM, Metzler G, Murray AP, et al. Establishment of antitumor memory in humans using in vitro-educated CD8+ T cells. Sci Transl Med. 2011;3:80ra34. doi: 10.1126/scitranslmed.3002207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chang AE, Li Q, Jiang G, Sayre DM, Braun TM, Redman BG. Phase II trial of autologous tumor vaccination, anti-CD3-activated vaccine-primed lymphocytes, and interleukin-2 in stage IV renal cell cancer. J Clin Oncol. 2003;21:884–90. doi: 10.1200/JCO.2003.08.023. [DOI] [PubMed] [Google Scholar]
  • 96.Thompson JA, Figlin RA, Sifri-Steele C, Berenson RJ, Frohlich MW. A phase I trial of CD3/CD28-activated T cells (Xcellerated T cells) and interleukin-2 in patients with metastatic renal cell carcinoma. Clin Cancer Res. 2003;9:3562–70. [PubMed] [Google Scholar]
  • 97.Zhou P, Liang P, Dong B, Yu X, Han Z, Xu Y. Phase I clinical study of combination therapy with microwave ablation and cellular immunotherapy in hepatocellular carcinoma. Cancer Biol Ther. 2011;11:450–6. doi: 10.4161/cbt.11.5.14669. [DOI] [PubMed] [Google Scholar]
  • 98.Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12:6106–15. doi: 10.1158/1078-0432.CCR-06-1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–70. doi: 10.1038/nm.1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Yamasaki K, Horiguchi S, Kurosaki M, Kunii N, Nagato K, Hanaoka H, et al. Induction of NKT cell-specific immune responses in cancer tissues after NKT cell-targeted adoptive immunotherapy. Clin Immunol. 2011;138:255–65. doi: 10.1016/j.clim.2010.11.014. [DOI] [PubMed] [Google Scholar]
  • 101.Curti BD, Ochoa AC, Powers GC, Kopp WC, Alvord WG, Janik JE, et al. Phase I trial of anti-CD3-stimulated CD4+ T cells, infusional interleukin-2, and cyclophosphamide in patients with advanced cancer. J Clin Oncol. 1998;16:2752–60. doi: 10.1200/JCO.1998.16.8.2752. [DOI] [PubMed] [Google Scholar]
  • 102.Laport GG, Levine BL, Stadtmauer EA, Schuster SJ, Luger SM, Grupp S, et al. Adoptive transfer of costimulated T cells induces lymphocytosis in patients with relapsed/refractory non-Hodgkin lymphoma following CD34+-selected hematopoietic cell transplantation. Blood. 2003;102:2004–13. doi: 10.1182/blood-2003-01-0095. [DOI] [PubMed] [Google Scholar]
  • 103.Berdeja JG, Hess A, Lucas DM, O’Donnell P, Ambinder RF, Diehl LF, et al. Systemic interleukin-2 and adoptive transfer of lymphokine-activated killer cells improves antibody-dependent cellular cytotoxicity in patients with relapsed B-cell lymphoma treated with rituximab. Clin Cancer Res. 2007;13:2392–9. doi: 10.1158/1078-0432.CCR-06-1860. [DOI] [PubMed] [Google Scholar]
  • 104.Wallen H, Thompson JA, Reilly JZ, Rodmyre RM, Cao J, Yee C. Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma. PLoS One. 2009;4:e4749. doi: 10.1371/journal.pone.0004749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Fang HB, Cai L, et al. Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood. 2011;117:788–97. doi: 10.1182/blood-2010-08-299396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2011 doi: 10.1182/blood-2011-10-384388. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sangiolo D. Cytokine induced killer cells as promising immunotherapy for solid tumors. J Cancer. 2011;2:363–8. doi: 10.7150/jca.2.363. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES