Mutant cancer-driving oncogenes are the best therapeutic targets, both with drugs like small-molecule inhibitors (SMI) and adoptive T-cell therapy (ATT), the most effective form of immunotherapy. Cancer cell survival often depends on oncogenes, which implies that they are homogenously expressed by all cancer cells and are difficult to select against. Mutant oncogene-directed therapy is relatively selective, as it targets preferentially the oncogene-expressing cancer cells. Both SMI and ATT can be highly effective in relevant preclinical models as well as selected clinical situations, and both share the risk of therapy resistance, facilitated by the frequent genetic instability of cancer cells. Recently, both therapies were compared in the same experimental model targeting the same oncogene. It showed that the oncogene-inactivating drug selected resistant clones, leading eventually to tumor relapse, whereas ATT eradicated large established tumors completely. The mode of tumor destruction likely explained the different outcome with only ATT destroying the tumor vasculature. Elucidating the cellular and molecular mechanisms responsible for tumor regression and relapse will define optimal conditions for the clinic. We argue that the ideal conditions of ATT in the experimental cancer model can be translated to individuals with cancer. Clin Cancer Res; 19(2); 320–6. ©2012 AACR.

The survival of cancer cells often remains long-term dependent on a single oncogene, a phenomenon termed as “oncogene addiction” (1, 2). This was shown in mice transgenic for an oncogene controlled by the tetracycline (Tet) system, which allowed to switch the oncogene on and off in a cell type–specific fashion with doxycycline (3). Doxycycline mimicked small-molecule inhibitors (SMI) blocking oncogene activity. The downregulation of cancer-driving oncogenes, for example, RAS, MYC, BCR-ABL, HER-2, or SV40 large T antigen (Tag), led to sustained tumor regression in different models including lymphoma, leukemia, adenoma, glioblastoma, sarcoma, and several different carcinomas (4–13). Drug resistance and tumor recurrence was observed in most models, albeit to different extent (2). The phenomenon of oncogene addiction led to the first SMI, imatinib mesylate, which was designed to inactivate the BCR-ABL fusion protein. BCR-ABL is an overactive tyrosine kinase causing chronic myeloid leukemia (CML) by a chromosomal translocation. The clinical effectiveness of imatinib against BCR-ABL–associated CML (14) led to further successful SMIs, for example, in the treatment of patients with melanoma and non–small cell lung cancer (NSCLC; ref. 15). Yet, therapeutic effects of SMIs are transient in most patients, and drug resistance is common with tumor burden being one critical factor. For example, in most imatinib-treated CML patients in the blast crisis, tumor recurred after 3 to 6 months, whereas those in the chronic phase remained tumor free for years (16). Many reasons can account for drug failure. The effectiveness of SMI in inhibiting oncogene activity in clinical situations is less clear than that of the Tet system in the experimental model. Currently, it is also difficult to assess which cancers in the clinic are long-term dependent on a single oncogene.

ATT experiments have been carried out for almost 50 years. Improvements such as the grafting of T cells selectively in lymphodepleted hosts have improved its success (17, 18). However, outcomes were variable because experimental models varied with regard to T-cell phenotype, target antigen, host conditions, and tumor burden. Under optimal conditions, ATT can lead to eradication of large established tumors in preclinical models distinguishing ATT from other immunotherapies. Herein we define a tumor as established when it grew for several weeks and reached a volume of at least 500 mm3, which is equivalent to approximately 109 cells and the least clinically detectable tumor size (19). It is critical to use models of such tumor burden, because only then tumors have built up a stroma, which is essential for progression and a decisive target of ATT (20). Furthermore, genetic instability, a hallmark of human cancer and primary cause of therapy resistance, becomes apparent with increasing tumor burden. T cells transferred in the course of allogeneic bone marrow (stem cell) transplantation can cure patients with leukemia (21) and virus-associated cancer (22). With regard to solid tumors, ATT in the clinic was hampered by the difficulty in generating T cells in vitro. Nevertheless, tumor-infiltrating lymphocytes (TIL) from melanomas, expanded in vitro and reinfused, achieved regression in a high proportion of patients (23). As was the case with SMIs, tumor recurrence frequently occurred. Thus, both SMI and ATT have been successfully used in the clinic but are impeded by therapy resistance. Because both therapies compete with each other, with clinical efficacy being the only measure, we recently established cancer models that allowed for the first time direct comparison of SMI with ATT targeting the same oncogene to treat large established tumors (9). Here, the molecular and cellular mechanisms leading to tumor regression and, possibly, recurrence are reviewed. We argue that ATT, by newly developed techniques, has become feasible and offers opportunities for improvement that at least in some indications it will successfully compete with SMI.

Depending on the cell type and the involved oncogene, various mechanisms have been associated with tumor regression following oncogene inactivation in mouse models. These include proliferative arrest, apoptosis, cell differentiation, autophagy, senescence, tumor dormancy, and modulation of the tumor microenvironment (Fig. 1). In the Tet-TagLuc model that uses sarcoma cells transformed by a Tet-controlled fusion protein of Tag and luciferase (TagLuc), noninvasive bioluminescence imaging visualized oncogene downregulation concomitant with proliferative arrest (loss of Ki-67 expression) as early as 1 day after doxycycline application (9). Tag inactivates tumor suppressors p53 and retinoblastoma (Rb), which are likely released upon its inactivation. In other models, for example, downregulation of BCR-ABL, RAS, MYC, and HER-2 in different cancers, apoptosis was observed (2). In the Tet-TagLuc model, we failed to see induction of apoptosis (cleaved caspase-3 expression). Instead, switching off TagLuc induced autophagy (lipidation of light chain 3 microtubule-associated protein). Autophagy, the degradation of proteins and damaged organelles in cytoplasmic vesicles, is induced under stress conditions such as starvation, hypoxia, and drug treatment. Autophagy was associated with tumor regression.

Figure 1.

Anticancer therapy against large established tumors using small-molecule inhibitor (SMI) therapy. Top, due to inherent genetic instability, cancer cells acquire additional mutation during progression from small malignant lesions to established tumors. Before starting therapy, the majority of clinically detectable tumors (≥500 mm3) contain cancer cell variants already that are resistant to the intended drug treatment (red cell). Several transgenic models with tetracycline controllable oncogenes (as indicated) proved the concept of “oncogene addiction.” Middle, treatment with oncogene inactivation drugs (SMI) can have multiple consequences on drug-responsive cancer cells, including apoptotic cell death, autophagic stress response that can culminate in cell death, differentiation, senescence, and tumor dormancy (as shown from left to right). These effects are thought not to be mutually exclusive. Bottom, left, SMI therapy targets drug-responsive cancer cell but leaves the tumor stroma, including tumor vasculature, largely intact. In addition, drug-responsive cells that do not respond with apoptotic cell death, but, for instance, undergo senescence, become part of the tumor microenvironment, which can suppress but also support survival and proliferation of escape variants, for example, by releasing growth factors. Bottom, right, drug resistance is caused by secondary mutation within the oncogene, for example, receptor tyrosine kinase (RTK), (blue star) that in most cases prevents drug binding (left) or by activation of alternative oncogenic pathways, for example, by growth factors released by stroma or drug-responsive tumor cells (right).

Figure 1.

Anticancer therapy against large established tumors using small-molecule inhibitor (SMI) therapy. Top, due to inherent genetic instability, cancer cells acquire additional mutation during progression from small malignant lesions to established tumors. Before starting therapy, the majority of clinically detectable tumors (≥500 mm3) contain cancer cell variants already that are resistant to the intended drug treatment (red cell). Several transgenic models with tetracycline controllable oncogenes (as indicated) proved the concept of “oncogene addiction.” Middle, treatment with oncogene inactivation drugs (SMI) can have multiple consequences on drug-responsive cancer cells, including apoptotic cell death, autophagic stress response that can culminate in cell death, differentiation, senescence, and tumor dormancy (as shown from left to right). These effects are thought not to be mutually exclusive. Bottom, left, SMI therapy targets drug-responsive cancer cell but leaves the tumor stroma, including tumor vasculature, largely intact. In addition, drug-responsive cells that do not respond with apoptotic cell death, but, for instance, undergo senescence, become part of the tumor microenvironment, which can suppress but also support survival and proliferation of escape variants, for example, by releasing growth factors. Bottom, right, drug resistance is caused by secondary mutation within the oncogene, for example, receptor tyrosine kinase (RTK), (blue star) that in most cases prevents drug binding (left) or by activation of alternative oncogenic pathways, for example, by growth factors released by stroma or drug-responsive tumor cells (right).

Close modal

Different mechanisms associated with oncogene inactivation–induced tumor regression are not mutually exclusive. TagLuc downregulation additionally led to rapid upregulation of fibronectin, a differentiation marker, which has been implicated in cellular senescence (24). In MYC-driven cancer models, oncogene inactivation similarly induced terminal differentiation (5, 10, 25), which ultimately may lead to senescence, characterized by irreversible cell-cycle arrest (26). Senescent cells remain metabolically active and produce such molecules as metalloproteinase, growth factors, proinflammatory cytokines, and chemokines, termed senescence-associated secretory phenotype (SASP), which may reinforce senescence in an autocrine manner and in a pINK4a/Rb-dependent fashion (24, 26–29). Conversely, growth factors secreted by senescent cells may support tumor growth and drug resistance by acting on neighboring cells, which allows selection of variants that activate alternative oncogenic pathways (28, 30). Recent clinical data with BRAF inhibitors support this assumption (31, 32). Whether tumor dormancy, observed upon MYC-inactivation (5), is related to cellular differentiation or senescence is not known.

The role of the tumor microenvironment for tumor regression upon oncogene inactivation is poorly understood. Tumor regression by MYC inactivation in experimental models was accompanied by regression of blood vessels, compatible with the activity of MYC to repress the angiogenesis inhibitor thrombospondin-1 and induce VEGF (25, 33–35). Reduced numbers of endothelial cells were also found within RAS-deprived melanomas (11) and TagLuc-deprived sarcomas (9), even though in the latter model most endothelial cells persisted in regressing tumors. It is currently not clear whether reduction of the tumor vasculature is a cause of tumor regression or a consequence of cancer cell death and subsequent remodeling of the tumor microenvironment. Together, a variety of mechanisms are associated with oncogene inactivation–induced tumor regression. They may occur simultaneously, they may differ depending on the transformed cell type, and they may occur, at least partially, in a similar fashion regardless of the inactivated oncogene.

The critical question is which mechanisms confer drug resistance. Tumor burden is of pivotal importance. If comparably small Tet-TagLuc tumors were treated by oncogene inactivation, most completely regressed without recurrence (9). If large tumors were treated, drug-resistant clones emerged without exception. Each tumor had a different genetically acquired mutation in the transactivator gene preventing doxycycline binding and TagLuc downregulation, resembling secondary mutations in the target oncogenes selected by SMI in the clinic (36–38). These data suggest that genetic instability, likely caused by the p53/Rb-inactivating activity of TagLuc, together with tumor burden, caused drug resistance. Because TagLuc inactivation resulted in rapid growth arrest but acquisition of mutations requires cell division, resistant clones likely preexisted in vivo before therapy. Similarly, recurrent tumors in patients may often be derived from preexisting variants insensitive to the drug due to secondary mutations, oncogene amplification, activation of upstream receptor tyrosine kinases, or pursuing alternative survival pathways (37, 39, 40), and hence, were never amenable to respond to the drug. However, provided that the majority of cancer cells are drug responsive, the various mechanisms outlined above likely create a microenvironment that influences the resistant clone. Which mechanism (apoptosis, differentiation, autophagy, senescence, dormancy, or microenvironment) positively or negatively affects the resistant clone is still poorly understood.

In the same model of large established Tet-TagLuc tumors, in which drug resistance was the rule, ATT was used with CD8+ effector T (TE) cells, recognizing a single peptide epitope of Tag. Escape variants appeared likely, as in vitro T cells could select variants with mutation in the peptide epitope (41). However, without exception, the TE cells eradicated large established tumors and no escape variants were ever selected (9), which raises 2 questions. First, how did the rejection response by the TE cells differ from the regression/recurrence response induced by the drug, and second, what distinguishes the experimental cancer model from the clinical ATT trials, mainly in melanoma? Here, objective responses were frequent, but long-term responses rare (23). In most patients, it remained unclear whether tumor recurrence was due to immune escape, for example, loss of the target antigen or the peptide presenting MHC I molecules (42, 43), or loss of T-cell function.

Drug therapy quite selectively eliminated cancer cells. Endothelial cells and probably the whole microenvironment remained largely alive. Therefore, we argue that a vital stroma facilitated the survival and outgrowth of resistant clones. In sharp contrast, during ATT, cancer cells and the tumor vasculature were simultaneously destroyed, so that the entire tumor tissue became necrotic (9). Under such hostile conditions, potential escape variants are unlikely to survive, a phenomenon termed bystander elimination (20). TE cell–mediated rejection of large established tumors depended on such cytokines as IFN-γ and TNF-α and such cytotoxic molecules as perforin (refs. 20, 44; Fig. 2). In one model of established tumors, tumor stroma cells needed to respond to both cytokines, which prevented selection of antigen loss variants (20, 44). In addition, tumor antigen cross-presenting stroma cells were lysed by TE cells in vitro. On the basis of the Tet-TagLuc model, we argue that antigen recognition on the cancer cells was sufficient for TE cells to produce IFN-γ, which acted on and destroyed the tumor vasculature without being directly recognized by the TE cells (9, 45, 46). The efficacy of ATT to reject large established Tet-TagLuc tumors can be explained by the optimal conditions of the experimental setting that relied on factors such as the phenotype of the TE cells, the cancer cells, the target antigen, and the host. T cells were derived from immunized T-cell receptor (TCR) transgenic mice, whose T cells were specific for Tag (9). They had not been cultured in vitro for prolonged time, which may render T cells ineffective (47), and they may have contained central memory T (TCM) cells or T cells with stem cell–like characteristics (TSCM) that showed improved efficacy in vivo (48, 49). The T cells were derived from the nontolerant repertoire, which excludes skewing toward low-avidity T cells by tolerance mechanisms, and recognized the target antigen as foreign. The cancer cells presumably upregulated MHC I molecules in response to IFN-γ, comparable with “soft” lesions in patients (50). The hosts, Rag-1−/− mice, were lymphopenic. Most importantly, the target antigen was the cancer-driving oncogene, expressed by all cancer cells, but not normal cells. How can these features be translated into the clinic?

Figure 2.

Anticancer therapy against large established tumors using adoptive T-cell therapy (ATT). Top left, at least 4 reasons may account for successful TCR gene therapy: 1, cancer cells respond to IFN-γ by MHC I upregulation. 2, TCR is isolated from the nontolerant repertoire of humanized TCR gene loci/MHC I transgenic mice. 3, TCR is introduced into patients' TCM/TSCM cells. 4, TCR is specific for mutant immunogenic epitope of cancer-driving oncogene. Top, right, tumor destruction by ATT involves 3 main mechanisms: First, direct killing of cancer cells by TE cells occurs upon recognition of antigenic epitopes presented on MHC I molecules by cancer cells. TCR engagement leads to expression of cytotoxic molecules by TE cells that lyse the antigen-presenting cancer cells. Second, under certain conditions, tumor antigens are transferred to noncancer cells, for example, via vesicle transport, and are presented by MHC I molecules on stroma cells. Those cross-presenting stroma cells become targets for TE cells and will be killed upon recognition. Finally, molecules (TNF-α and IFN-γ) released upon cancer cell recognition by TE cells can have destructive effects on neighboring cells, for example, endothelial cells, that do not present antigenic epitopes on MHC I molecules. Those cells are killed by bystander elimination. Bottom, ideally, T cells invade tumors in large numbers, leaving dead tumor cells, including stroma cells, behind. Potential antigen loss variants that escaped T-cell destruction fail to survive in the harsh tumor microenvironment created by those destructive TE cells. huTCR, human T-cell receptor; TSCM, T cells with stem cell-like characteristics.

Figure 2.

Anticancer therapy against large established tumors using adoptive T-cell therapy (ATT). Top left, at least 4 reasons may account for successful TCR gene therapy: 1, cancer cells respond to IFN-γ by MHC I upregulation. 2, TCR is isolated from the nontolerant repertoire of humanized TCR gene loci/MHC I transgenic mice. 3, TCR is introduced into patients' TCM/TSCM cells. 4, TCR is specific for mutant immunogenic epitope of cancer-driving oncogene. Top, right, tumor destruction by ATT involves 3 main mechanisms: First, direct killing of cancer cells by TE cells occurs upon recognition of antigenic epitopes presented on MHC I molecules by cancer cells. TCR engagement leads to expression of cytotoxic molecules by TE cells that lyse the antigen-presenting cancer cells. Second, under certain conditions, tumor antigens are transferred to noncancer cells, for example, via vesicle transport, and are presented by MHC I molecules on stroma cells. Those cross-presenting stroma cells become targets for TE cells and will be killed upon recognition. Finally, molecules (TNF-α and IFN-γ) released upon cancer cell recognition by TE cells can have destructive effects on neighboring cells, for example, endothelial cells, that do not present antigenic epitopes on MHC I molecules. Those cells are killed by bystander elimination. Bottom, ideally, T cells invade tumors in large numbers, leaving dead tumor cells, including stroma cells, behind. Potential antigen loss variants that escaped T-cell destruction fail to survive in the harsh tumor microenvironment created by those destructive TE cells. huTCR, human T-cell receptor; TSCM, T cells with stem cell-like characteristics.

Close modal

For ATT with the best possible outcome, we envisage the following scenario. The use of TCR gene therapy overcomes a number of current hurdles (51). It allows equipping patients' own T cells with desired specificities and generation of sufficient numbers of T cells in a short period of time, avoiding their exhaustion (47). The TCR will be transduced into TCM or TSCM cells, which may ensure better persistence and function upon transfer (48, 49). TCR-engineered T cells will be infused into patients with cancer rendered lymphopenic by chemotherapy or irradiation, allowing efficient engraftment but inhibiting immune suppression (17, 18, 23, 52). Transgenic mice expressing human MHC molecules and a diverse human TCR repertoire serve as a tool to rapidly analyze whether peptide antigens are immunogenic, that is, if they are processed and presented by MHC molecules and if they efficiently induce T-cell responses following immunization (53). With the use of the human TCR transgenic mouse, any human peptide sequence not encoded by the mouse genome is suitable for immunization and might yield TCRs with optimal affinity. Optimal affinity means that the T cells are restricted to human self-MHC molecules and recognize the peptide antigen as foreign, for example, as representatives of the nontolerant repertoire. By using peptide/MHC multimers, specific T cells of the transgenic mice can be sorted, then, human TCRs isolated, for example, by single cell PCR, the TCRs optimized for efficient expression while avoiding mispairing with endogenous TCR and are used for transduction of patients' T cells with viral vectors (54, 55). We anticipate that such engineered T cells, comparable with the Tet-TagLuc tumor model, will exert optimal effector function within the tumor microenvironment and not only eliminate the cancer cells but also destroy the tumor vasculature by release of large amounts of IFN-γ and TNF-α, thereby indirectly eliminating potential escape variants.

The key problem of ATT is to target the right antigen to prevent tumor recurrence and toxic side effects (56). This sounds simple, given the large number of putative tumor antigens. However, most are tumor-associated (self) antigens (TAA). TAAs are also expressed by normal cells. Expression by rare but vital cells has usually not been analyzed. Moreover, TAA expression may be heterogeneous within the tumor/metastases of a given individual. Thus, targeting TAAs bears the risk of ineffective long-term responses and destruction of normal tissues. Clinical trials with TCR [or chimeric antibody receptor (CAR)]–engineered T cells, for example, directed against Melan-A/MART-1, gp100, HER-2 and carcinoembryonic antigen, support this assumption (57–59). Some differentiation antigens, such as CD19, expressed by B cells and B-cell lymphomas, represent exceptions. CAR-engineered T cells, specific for CD19, effectively eliminated B-cell lymphomas and normal B cells as an acceptable side effect (60). However, for many, if not most, TAA's substantial toxicity by effective ATT is predictable.

If, in contrast, mutant cancer–driving oncogenes are targeted by ATT, many of the problems with TAAs will resolve. Except for antigens encoded by cancer viruses, mutated antigens are the only tumor-specific antigens (TSA). Mutant cancer–driving antigens are expressed by all cancer cells, exclusively by cancer cells (with the possible exception of cross-presenting stroma cells; ref. 20), and difficult to select against. Of note, target antigen expression on normal cells can prevent tumor rejection by ATT (61). Some TSAs are shared between tumors, for example, RAS, BRAF, p53, or fusion regions of chromosomal translocations. However, despite continuous claims of their immunogenicity, often analyzed by reverse immunology, firm evidence that T cells raised against shared TSAs reject tumors is lacking. For example, the TEL–AML chromosomal breakpoint region was claimed to generate an immunogenic HLA-A0201–resticted epitope (62), but subsequently, it was shown that it is not processed and presented (63). Thus, raising T cells against shared TSAs is difficult and likely possible only for a few HLA restriction molecules. The solution to this problem is whole cancer genome analysis and identification of patient-specific somatic mutations, leading to immunogenic epitopes and being putatively cancer driving. Some cancers, such as melanoma, acquired more than 33,000 mutations, of which around 0.9% were point mutations in coding regions (64). In fact, patient-specific immunogenic TSAs have been detected in melanomas (65).

In conclusion, under ideal conditions ATT eradicates large tumors, whereas SMI treatment eventually leads to drug resistance. We think ideal ATT can be pursued clinically through the following process: sequence the cancer genome, identify recurrent somatic mutations in coding regions, use computer algorithms to predict immunogenic epitopes, analyze peptide/MHC stability, verify immunogenicity in and isolate optimal affinity TCRs from MHC/TCR humanized mice, construct TCR retroviral vectors, transduce patients' T cells, and transfer TCR-redirected T cells back into lymphopenic patients. We are aware that clinically additional problems have to be resolved, which could not be discussed here.

No potential conflicts of interest were disclosed.

Conception and design: T. Blankenstein, K. Anders

Writing, review, and/or revision of the manuscript: T. Blankenstein, K. Anders

This work was supported by grants from the Deutsche Forschungsgemeinschaft (Sonderforschungsbereich TR36) and the “Alliance” program of the Helmholtz-Gemeinschaft Deutscher Forschungszentren (HA-202).

1.
Weinstein
IB
. 
Cancer. Addiction to oncogenes–the Achilles heal of cancer
.
Science
2002
;
297
:
63
4
.
2.
Felsher
DW
. 
Tumor dormancy and oncogene addiction
.
APMIS
2008
;
116
:
629
37
.
3.
Gossen
M
,
Bujard
H
. 
Studying gene function in eukaryotes by conditional gene inactivation
.
Annu Rev Genet
2002
;
36
:
153
73
.
4.
Felsher
DW
,
Bishop
JM
. 
Reversible tumorigenesis by MYC in hematopoietic lineages
.
Mol Cell
1999
;
4
:
199
207
.
5.
Jain
M
,
Arvanitis
C
,
Chu
K
,
Dewey
W
,
Leonhardt
E
,
Trinh
M
, et al
Sustained loss of a neoplastic phenotype by brief inactivation of MYC
.
Science
2002
;
297
:
102
4
.
6.
D'Cruz
CM
,
Gunther
EJ
,
Boxer
RB
,
Hartman
JL
,
Sintasath
L
,
Moody
SE
, et al
c-MYC induces mammary tumorigenesis by means of a preferred pathway involving spontaneous Kras2 mutations
.
Nat Med
2001
;
7
:
235
9
.
7.
Moody
SE
,
Sarkisian
CJ
,
Hahn
KT
,
Gunther
EJ
,
Pickup
S
,
Dugan
KD
, et al
Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis
.
Cancer Cell
2002
;
2
:
451
61
.
8.
Huettner
CS
,
Zhang
P
,
Van Etten
RA
,
Tenen
DG
. 
Reversibility of acute B-cell leukaemia induced by BCR-ABL1
.
Nat Genet
2000
;
24
:
57
60
.
9.
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
.
10.
Shachaf
CM
,
Kopelman
AM
,
Arvanitis
C
,
Karlsson
A
,
Beer
S
,
Mandl
S
, et al
MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer
.
Nature
2004
;
431
:
1112
7
.
11.
Chin
L
,
Tam
A
,
Pomerantz
J
,
Wong
M
,
Holash
J
,
Bardeesy
N
, et al
Essential role for oncogenic Ras in tumour maintenance
.
Nature
1999
;
400
:
468
72
.
12.
Fisher
GH
,
Wellen
SL
,
Klimstra
D
,
Lenczowski
JM
,
Tichelaar
JW
,
Lizak
MJ
, et al
Induction and apoptotic regression of lung adenocarcinomas by regulation of a K-Ras transgene in the presence and absence of tumor suppressor genes
.
Genes Dev
2001
;
15
:
3249
62
.
13.
Holmen
SL
,
Williams
BO
. 
Essential role for Ras signaling in glioblastoma maintenance
.
Cancer Res
2005
;
65
:
8250
5
.
14.
Jabbour
E
,
Cortes
J
,
Kantarjian
H
. 
Long-term outcomes in the second-line treatment of chronic myeloid leukemia: a review of tyrosine kinase inhibitors
.
Cancer
2011
;
117
:
897
906
.
15.
Flaherty
KT
,
Puzanov
I
,
Kim
KB
,
Ribas
A
,
McArthur
GA
,
Sosman
JA
, et al
Inhibition of mutated, activated BRAF in metastatic melanoma
.
N Engl J Med
2010
;
363
:
809
19
.
16.
Druker
BJ
,
Sawyers
CL
,
Kantarjian
H
,
Resta
DJ
,
Reese
SF
,
Ford
JM
, et al
Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome
.
N Engl J Med
2001
;
344
:
1038
42
.
17.
Glynn
JP
,
Halpern
BL
,
Fefer
A
. 
An immunochemotherapeutic system for the treatment of a transplanted Moloney virus-induced lymphoma in mice
.
Cancer Res
1969
;
29
:
515
20
.
18.
Greenberg
PD
,
Cheever
MA
,
Fefer
A
. 
Eradication of disseminated murine leukemia by chemoimmunotherapy with cyclophosphamide and adoptively transferred immune syngeneic Lyt-1+2- lymphocytes
.
J Exp Med
1981
;
154
:
952
63
.
19.
Schreiber
K
,
Rowley
DA
,
Riethmuller
G
,
Schreiber
H
. 
Cancer immunotherapy and preclinical studies: why we are not wasting our time with animal experiments
.
Hematol Oncol Clin North Am
2006
;
20
:
567
84
.
20.
Spiotto
MT
,
Rowley
DA
,
Schreiber
H
. 
Bystander elimination of antigen loss variants in established tumors
.
Nat Med
2004
;
10
:
294
8
.
21.
Kolb
HJ
,
Mittermuller
J
,
Clemm
C
,
Holler
E
,
Ledderose
G
,
Brehm
G
, et al
Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients
.
Blood
1990
;
76
:
2462
5
.
22.
Bollard
CM
,
Rooney
CM
,
Heslop
HE
. 
T-cell therapy in the treatment of post-transplant lymphoproliferative disease
.
Nat Rev Clin Oncol
2012
;
9
:
510
9
.
23.
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
.
24.
Shelton
DN
,
Chang
E
,
Whittier
PS
,
Choi
D
,
Funk
WD
. 
Microarray analysis of replicative senescence
.
Curr Biol
1999
;
9
:
939
45
.
25.
Pelengaris
S
,
Littlewood
T
,
Khan
M
,
Elia
G
,
Evan
G
. 
Reversible activation of c-Myc in skin: induction of a complex neoplastic phenotype by a single oncogenic lesion
.
Mol Cell
1999
;
3
:
565
77
.
26.
Wu
CH
,
van Riggelen
J
,
Yetil
A
,
Fan
AC
,
Bachireddy
P
,
Felsher
DW
. 
Cellular senescence is an important mechanism of tumor regression upon c-Myc inactivation
.
Proc Natl Acad Sci U S A
2007
;
104
:
13028
33
.
27.
Liu
D
,
Hornsby
PJ
. 
Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion
.
Cancer Res
2007
;
67
:
3117
26
.
28.
Coppe
JP
,
Patil
CK
,
Rodier
F
,
Sun
Y
,
Munoz
DP
,
Goldstein
J
, et al
Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor
.
PLoS Biol
2008
;
6
:
2853
68
.
29.
Acosta
JC
,
O'Loghlen
A
,
Banito
A
,
Guijarro
MV
,
Augert
A
,
Raguz
S
, et al
Chemokine signaling via the CXCR2 receptor reinforces senescence
.
Cell
2008
;
133
:
1006
18
.
30.
Krtolica
A
,
Parrinello
S
,
Lockett
S
,
Desprez
PY
,
Campisi
J
. 
Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging
.
Proc Natl Acad Sci U S A
2001
;
98
:
12072
7
.
31.
Straussman
R
,
Morikawa
T
,
Shee
K
,
Barzily-Rokni
M
,
Qian
ZR
,
Du
J
, et al
Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion
.
Nature
2012
;
487
:
500
4
.
32.
Wilson
TR
,
Fridlyand
J
,
Yan
Y
,
Penuel
E
,
Burton
L
,
Chan
E
, et al
Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors
.
Nature
2012
;
487
:
505
9
.
33.
Pelengaris
S
,
Khan
M
,
Evan
GI
. 
Suppression of Myc-induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression
.
Cell
2002
;
109
:
321
34
.
34.
Janz
A
,
Sevignani
C
,
Kenyon
K
,
Ngo
CV
,
Thomas-Tikhonenko
A
. 
Activation of the myc oncoprotein leads to increased turnover of thrombospondin-1 mRNA
.
Nucleic Acids Res
2000
;
28
:
2268
75
.
35.
Ngo
CV
,
Gee
M
,
Akhtar
N
,
Yu
D
,
Volpert
O
,
Auerbach
R
, et al
An in vivo function for the transforming Myc protein: elicitation of the angiogenic phenotype
.
Cell Growth Differ
2000
;
11
:
201
10
.
36.
Kobayashi
S
,
Boggon
TJ
,
Dayaram
T
,
Janne
PA
,
Kocher
O
,
Meyerson
M
, et al
EGFR mutation and resistance of non-small-cell lung cancer to gefitinib
.
N Engl J Med
2005
;
352
:
786
92
.
37.
Nazarian
R
,
Shi
H
,
Wang
Q
,
Kong
X
,
Koya
RC
,
Lee
H
, et al
Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation
.
Nature
2010
;
468
:
973
7
.
38.
Gorre
ME
,
Mohammed
M
,
Ellwood
K
,
Hsu
N
,
Paquette
R
,
Rao
PN
, et al
Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification
.
Science
2001
;
293
:
876
80
.
39.
Diaz
LA
 Jr
,
Williams
RT
,
Wu
J
,
Kinde
I
,
Hecht
JR
,
Berlin
J
, et al
The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers
.
Nature
2012
;
486
:
537
40
.
40.
Engelman
JA
,
Zejnullahu
K
,
Mitsudomi
T
,
Song
Y
,
Hyland
C
,
Park
JO
, et al
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling
.
Science
2007
;
316
:
1039
43
.
41.
Mylin
LM
,
Schell
TD
,
Epler
M
,
Kusuma
C
,
Assis
D
,
Matsko
C
, et al
Diversity of escape variant mutations in Simian virus 40 large tumor antigen (SV40 Tag) epitopes selected by cytotoxic T lymphocyte (CTL) clones
.
Virology
2007
;
364
:
155
68
.
42.
Yee
C
,
Thompson
JA
,
Byrd
D
,
Riddell
SR
,
Roche
P
,
Celis
E
, et al
Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells
.
Proc Natl Acad Sci U S A
2002
;
99
:
16168
73
.
43.
Restifo
NP
,
Marincola
FM
,
Kawakami
Y
,
Taubenberger
J
,
Yannelli
JR
,
Rosenberg
SA
. 
Loss of functional beta 2-microglobulin in metastatic melanomas from five patients receiving immunotherapy
.
J Natl Cancer Inst
1996
;
88
:
100
8
.
44.
Zhang
B
,
Karrison
T
,
Rowley
DA
,
Schreiber
H
. 
IFN-gamma- and TNF-dependent bystander eradication of antigen-loss variants in established mouse cancers
.
J Clin Invest
2008
;
118
:
1398
404
.
45.
Schuler
T
,
Blankenstein
T
. 
Cutting edge: CD8+ effector T cells reject tumors by direct antigen recognition but indirect action on host cells
.
J Immunol
2003
;
170
:
4427
31
.
46.
Blankenstein
T
. 
The role of tumor stroma in the interaction between tumor and immune system
.
Curr Opin Immunol
2005
;
17
:
180
6
.
47.
Gattinoni
L
,
Klebanoff
CA
,
Palmer
DC
,
Wrzesinski
C
,
Kerstann
K
,
Yu
Z
, et al
Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells
.
J Clin Invest
2005
;
115
:
1616
26
.
48.
Gattinoni
L
,
Lugli
E
,
Ji
Y
,
Pos
Z
,
Paulos
CM
,
Quigley
MF
, et al
A human memory T cell subset with stem cell-like properties
.
Nat Med
2011
;
17
:
1290
7
.
49.
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
.
50.
Garrido
F
,
Cabrera
T
,
Aptsiauri
N
. 
“Hard” and “soft” lesions underlying the HLA class I alterations in cancer cells: implications for immunotherapy
.
Int J Cancer
2010
;
127
:
249
56
.
51.
Schumacher
TN
. 
T-cell-receptor gene therapy
.
Nat Rev Immunol
2002
;
2
:
512
9
.
52.
Gabrilovich
DI
,
Ostrand-Rosenberg
S
,
Bronte
V
. 
Coordinated regulation of myeloid cells by tumours
.
Nat Rev Immunol
2012
;
12
:
253
68
.
53.
Li
LP
,
Lampert
JC
,
Chen
X
,
Leitao
C
,
Popovic
J
,
Muller
W
, et al
Transgenic mice with a diverse human T cell antigen receptor repertoire
.
Nat Med
2010
;
16
:
1029
34
.
54.
Uckert
W
,
Schumacher
TN
. 
TCR transgenes and transgene cassettes for TCR gene therapy: status in 2008
.
Cancer Immunol Immunother
2009
;
58
:
809
22
.
55.
Kammertoens
T
,
Blankenstein
T
. 
Making and circumventing tolerance to cancer
.
Eur J Immunol
2009
;
39
:
2345
53
.
56.
Offringa
R
. 
Antigen choice in adoptive T-cell therapy of cancer
.
Curr Opin Immunol
2009
;
21
:
190
9
.
57.
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
.
58.
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
.
59.
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
.
60.
Kalos
M
,
Levine
BL
,
Porter
DL
,
Katz
S
,
Grupp
SA
,
Bagg
A
, et al
T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia
.
Sci Transl Med
2011
;
3
:
95ra73
.
61.
Buschow
C
,
Charo
J
,
Anders
K
,
Loddenkemper
C
,
Jukica
A
,
Alsamah
W
, et al
In vivo imaging of an inducible oncogenic tumor antigen visualizes tumor progression and predicts CTL tolerance
.
J Immunol
2010
;
184
:
2930
8
.
62.
Yotnda
P
,
Garcia
F
,
Peuchmaur
M
,
Grandchamp
B
,
Duval
M
,
Lemonnier
F
, et al
Cytotoxic T cell response against the chimeric ETV6-AML1 protein in childhood acute lymphoblastic leukemia
.
J Clin Invest
1998
;
102
:
455
62
.
63.
Popovic
J
,
Li
LP
,
Kloetzel
PM
,
Leisegang
M
,
Uckert
W
,
Blankenstein
T
. 
The only proposed T-cell epitope derived from the TEL-AML1 translocation is not naturally processed
.
Blood
2011
;
118
:
946
54
.
64.
Pleasance
ED
,
Cheetham
RK
,
Stephens
PJ
,
McBride
DJ
,
Humphray
SJ
,
Greenman
CD
, et al
A comprehensive catalogue of somatic mutations from a human cancer genome
.
Nature
2010
;
463
:
191
6
.
65.
Lennerz
V
,
Fatho
M
,
Gentilini
C
,
Frye
RA
,
Lifke
A
,
Ferel
D
, et al
The response of autologous T cells to a human melanoma is dominated by mutated neoantigens
.
Proc Natl Acad Sci U S A
2005
;
102
:
16013
8
.