Redirecting T-cell specificity through the introduction of a chimeric antigen receptor (CAR) is emerging as a clinically feasible approach for adoptive immunotherapy. In this issue of Blood, Till and colleagues now demonstrate that autologous T cells genetically modified to express a CD20-specific CAR can be safely infused in patients with B-lineage lymphomas.

Approximately 15 years after Zelig Eshhar demonstrated how to redirect the specificity of T cells,1  investigators are reporting their early clinical experiences with infusing T cells genetically modified to express chimeric antigen receptors (CARs). By combining T-cell therapy with gene therapy in compliance with current good manufacturing practice (cGMP) for phase 1 and 2 trials, investigators have bypassed tolerance to enable clinical-grade T cells to recognize desired cell-surface antigens independent of MHC. T cells are rendered tumor-specific through introduction of a CAR, which is typically composed of the scFv from a monoclonal antibody (mAb) that forms part of the CAR ectodomain and which, upon binding antigen, activates T cells by phosphorylation of conserved immunoglobulin tyrosine activation motifs within a chimeric CD3-ζ or FcϵRI endodomain. To limit potential deleterious off-target effects, the first human trials for hematopoietic malignancies have infused genetically modified CAR+ T cells that target lineage-restricted antigens, such as CD19 and CD20 expressed on malignant (and healthy) B cells,2,3  and generally have used first-generation CARs (activating T cells solely through CD3-ζ4 ). Building on clinical experiences of therapeutic mAbs targeting CD20, Till and colleagues demonstrate that an intrapatient dose-escalation study infusing autologous clinical-grade T cells expressing a CD20-specific CAR with or without low-dose IL-2 can be undertaken in patients with non-Hodgkin lymphomas. In the current financial and regulatory climate, publishing the results of a gene-therapy trial is a singular accomplishment. However, like many initial human experiences, the results raise more questions than they answer.

Sustaining the survival of adoptively transferred CAR+ T cells is one of the major impediments to achieving significant therapeutic responses. One way to enhance persistence is to infuse a heterogeneous population of T cells so that subpopulations can participate in lymphopenia-induced proliferation. This has been championed by Rosenberg and colleagues5  at the National Institutes of Health who demonstrated that clinical responses can be achieved when melanoma-specific T cells are infused after lymphodepleting chemotherapy and when bulk populations of T cells are given rather than T-cell clones. Thus, most infusions of CD20-specific T cells were given after chemotherapy and the trial was altered to infuse populations of genetically modified T cells rather than clones. Cumulatively, up to 4.4 × 109/m2 cells were infused within 10 days, and while a limited number of infused T cells could be detected in the peripheral blood of some patients for up to 3 months, the question remains as to why the infused T cells did not persist longer and at increased levels leading to loss of normal CD20+ B cells. Perhaps it was due to the cells entering replicative senescence after nonviral gene transfer and ex vivo expansion to clinically meaningful numbers. Perhaps it was due to insufficient T-cell help, despite the use of low-dose IL-2. Perhaps it was due to the level of CAR expression or competency of CAR-dependent signaling, which might have been insufficient to sustain a proliferative T-cell signal. Perhaps it was due to a lack of CAR+ central-memory T cells in the inoculum that were capable of long-term in vivo persistence.6  Or perhaps it was due to incomplete lymphodepletion resulting from the choice of preinfusion chemotherapies used.

The trial described by Till et al drives home the observation that multiple infusions of autologous CAR+ T cells targeting a B-lineage antigen are both safe and feasible. This clinical experience can now serve as a platform for future endeavors to answer questions concerning the improvement of persistence and the resolving of issues regarding homing to tumor deposits, thereby improving the therapeutic potential of their CAR+ T cells.

The clinical data in this issue of Blood are among the first reports on the potential of T cells manufactured under cGMP that have been genetically modified to redirect specificity.7-9  With this publication, the authors have advanced the promising technology of CARs, which combine the specificity of mAbs with the replicative and homing potentials of T cells.

Conflict-of-interest disclosure: The author declares no competing financial interests. ■

1
Eshhar
 
Z
Waks
 
T
Gross
 
G
Schindler
 
DG
Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors.
Proc Natl Acad Sci U S A
1993
, vol. 
90
 (pg. 
720
-
724
)
2
Cooper
 
LJ
Topp
 
MS
Serrano
 
LM
, et al. 
T-cell clones can be rendered specific for CD19: toward the selective augmentation of the graft-versus-B-lineage leukemia effect.
Blood
2003
, vol. 
101
 (pg. 
1637
-
1644
)
3
Brentjens
 
RJ
Latouche
 
JB
Santos
 
E
, et al. 
Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15.
Nat Med
2003
, vol. 
9
 (pg. 
279
-
286
)
4
Wang
 
J
Press
 
OW
Lindgren
 
CG
, et al. 
Cellular immunotherapy for follicular lymphoma using genetically modified CD20-specific CD8+ cytotoxic T lymphocytes.
Mol Ther
2004
, vol. 
9
 (pg. 
577
-
586
)
5
Dudley
 
ME
Wunderlich
 
JR
Yang
 
JC
, 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
, vol. 
23
 (pg. 
2346
-
2357
)
6
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
, vol. 
118
 (pg. 
294
-
305
)
7
Park
 
JR
DiGiusto
 
DL
Slovak
 
M
, et al. 
Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma.
Mol Ther
2007
, vol. 
15
 (pg. 
825
-
833
)
8
Lamers
 
CH
Sleijfer
 
S
Vulto
 
AG
, et al. 
Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience.
J Clin Oncol
2006
, vol. 
24
 (pg. 
220
-
222
)
9
Kershaw
 
MH
Westwood
 
JA
Parker
 
LL
, et al. 
A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer.
Clin Cancer Res
2006
, vol. 
12
 (pg. 
6106
-
6115
)
Sign in via your Institution