Cellular immune responses that protect against tumors typically have been attributed to CD8 T cells. However, CD4 T cells also play a central role. It was shown recently that, in a patient with metastatic cholangiocarcinoma, CD4 T cells specific for a peptide from a mutated region of ERBB2IP could arrest tumor progression. This and other recent findings highlight new opportunities for CD4 T cells in cancer immunotherapy. In this article, I discuss the role and regulation of CD4 T cells in response to tumor Ags. Emphasis is placed on the types of Ags and mechanisms that elicit tumor-protective responses. I discuss the advantages and drawbacks of cancer immunotherapy through personalized genomics. These considerations should help to guide the design of next-generation therapeutic cancer vaccines.

Antitumor immune defenses include Abs, T cells, and NK cells. Abs are effective against surface-exposed, tumor-specific Ags. The use of monoclonal, bifunctional, or multispecific Abs to treat cancer requires multiple injections and is expensive (14). T cells express polymorphic AgRs for specific Ag recognition, possess effector functions, and develop memory characteristics. T cells are at the core of “immune surveillance” theories and are the best candidates to exact a toll on cancer cells in an Ag-specific manner (5, 6). NK cells and related cell types (NKT cells and cytokine-induced killer cells) express nonpolymorphic cell surface receptors that target cells for destruction in an Ag-independent manner. These cells lack the ability to acquire functional memory characteristics (79).

Typically, cellular immune responses that protect against tumors have been attributed to CD8 T cells. One main reason is that, similar to most normal tissues, tumor cells express little, if any, MHC class II molecules (10, 11). When MHC class II molecules are expressed, the invariant chain is often highly expressed, resulting in the generation of class II–associated invariant chain peptides that prevent the presentation of endogenous peptides in tumor cells (12). Consequently, pursuits of T cell–based therapies have focused primarily on CD8 T cells. In experimental animals, tumor-specific CD8 T cells are highly protective (reviewed in Ref. 13). In humans, tumor-specific CD8 T cells are present in patients with hematologic malignancies and solid tumors and within the pool of tumor-infiltrating lymphocytes, but they express high levels of PD-1 or exhibit suppressive characteristics (1417). However, therapeutic vaccines designed to induce CD8 T cell responses have been largely disappointing (1820).

In recent years, there has been increased interest in adoptive T cell therapies (21). In one approach, a patient’s T cells are genetically engineered to express a chimeric TCR, which consists of an Ag-binding domain of an Ab fused to the signaling components of a TCR and other signaling domains (22). This targeted treatment has shown great promise for treating B cell malignancies and is poised for success in other types of cancer (23, 24). In a second approach, tumor-infiltrating lymphocytes are isolated, expanded, and, in some cases, selected for TCR specificity before being reinfused into the same patient. The goal of this approach is to kill cancer cells through the recognition of MHC/peptide complexes (reviewed in Refs. 25, 26).

Despite the clinical successes of adoptive T cell therapies and Abs, cancer vaccines could be a more effective and potentially less toxic approach (27). In this article, I discuss strategies to pursue this objective with a focus on CD4 T cells and their role in antitumor immunity and tumor protection. Particular emphasis is placed on the process of peptide selection for inclusion in cancer vaccines comparing peptides from unmutated self-tumor Ags and mutated gene products.

CD4 T cells typically recognize peptides 12–16 aa in length presented by MHC class II molecules. These cells play a central role in the beginning and maintenance of adaptive immune responses. Their contribution to antitumor immunity is complex and reflects the diverse functions of various types of CD4 T cells (reviewed in Ref. 28). Almost 50 y ago, it was observed that, during the generation of Ab responses against thymus-dependent Ags, T cells provide help to B cells, facilitating isotype switching and affinity maturation (29). In 1971, Mitchison (30) showed that these effects require that both T and B cells recognize and respond to two different regions of the same protein. It was later shown that activation of CD4 T cells requires processing and presentation of the T cell determinant by the B cell (31). Thus, the helper function of CD4 T cells requires Ag processing and presentation on MHC class II molecules, both of which can occur in the B cell serving as an APC (3234). A similar form of T cell cooperation was demonstrated for CD4 T cells helping the activation and expansion of CD8 T cells (35). CD4 T cells also play a pivotal role in the generation and maintenance of memory CD8 T cells (3639). In addition to their helper role for B cells and CD8 T cells, CD4 T cells can be distinguished on the basis of the cytokines that they produce (IFN-γ is produced by Th1 cells, IL-4 is produced by Th2 cells, and IL-17 is produced by Th17 cells) or by their ability to downregulate the function of other T cells (regulatory T cells) (40, 41). The final type of CD4 T cells, T follicular helper cells, selects high-affinity, Ab-producing B cells for clonal expansion in germinal centers (42). The complex array of functions by the different classes of CD4 T cells in relation to the antitumor immune response was reviewed recently (28).

Early studies using tumor-bearing mice treated with an adoptive transfer of tumor-reactive CD4 T cells or by selectively depleting CD4 T cells demonstrated that these cells were needed in the effector phase of a protective antitumor immune response against tumors lacking MHC class II (4349). These experiments also found that activated CD4 T cells induced delayed-type hypersensitivity–like reactions and attracted inflammatory cells (macrophages, granulocytes, eosinophils, and NK cells) in or around the tumor (47, 50). Protection was thought to be mediated by CD4 T cells secreting IFN-γ, which would mediate cytotoxicity of tumor cells synergistically with TNF-α while also inducing reactive oxygen species and NO, inhibiting angiogenesis and stimulating cytotoxic macrophages (48, 5157). More recently, protection from tumors lacking MHC class II was studied after a low-dose adoptive transfer of Th1-like CD4 T cells specific for the melanoma-associated Ag Trp1 (58, 59). Both studies found that these Th1-like CD4 T cells acquired cytotoxicity in vivo and secreted IFN-γ, leading to the upregulation and expression of MHC class II molecules on the surface of tumor cells. This mechanism enabled MHC class II–restricted killing and protection independent of B, NK, or other T cells in the host (58, 59). For tumors expressing MHC class II molecules, CD4 T cells kill target cells directly via conventional MHC-dependent recognition (46, 58, 60, 61). This includes CD4 T cell responses against carbohydrate epitopes (62). A recent report found that conventional Th1 CD4 T cells can be converted into cytolytic CD4 T cells by reducing the expression of the transcription factor ThPOK (63).

In cancer patients, MHC class II–restricted CD4 T cell responses against self-Ags have been detected in the circulation and at the tumor site (6473). Consistent with the finding that human CD4 T cells against pathogens are cytotoxic—lysing target cells—human CD4 T cells were reported to lyse tumor cells in a MHC class II–restricted manner by predominantly perforin- or granzyme-mediated killing (7480). Collectively, human CD4 T cells can suppress tumor growth through the secretion of IFN-γ or directly kill tumor cells expressing adequate levels of MHC class II and self-Ags on their surface (79, 81). Activated CD4 T cells also could lead to the expression of MHC class II molecules through the secretion of IFN-γ or by blocking the inhibitory receptor CTLA-4, as demonstrated in the mouse (59). Whichever scenario applies, if a comparison with MHC class I–restricted CD8 T cell activation and function is warranted, one would expect that few (<100) complexes would suffice to mediate both intratumor activation and tumor cell killing by CD4 T cells (82, 83).

The induction and maintenance of tumor-specific T cells are regulated by mechanisms that, alone or in combination, diminish the ability of the immune system to control tumor growth and spread. These mechanisms include the following: central and peripheral tolerance, ignorance, the size of the repertoire and the hierarchical order with which T cell determinants are used and become immunogenic, regulatory T cells, myeloid suppressor cells, immunosuppression generated in the tumor microenvironment through inflammation, and endoplasmic reticulum stress and its influence on phagocytic cells and Ag presentation (8491).

Without going into the details of each of these mechanisms, it is noteworthy that sporadic tumors in mice are immunogenic, yet tolerance is induced by the expansion of nonfunctional T cells (92). Likewise, CD8 T cells generated by vaccination (peptide-in-adjuvant) in melanoma patients predominantly have a quiescent phenotype (93). In mice, CD8 T cells against self-Ags become tolerant through epigenetic mechanisms that are independent of the tolerogen (94). Together, these examples suggest that at least one main reason for the inefficient control of cancer by T cells is the T cells themselves. One possibility is that this is the result of immunosuppressive signals within the tumor microenvironment (85). Not surprisingly, the reactivation of T cells with agonistic Abs against inhibitory receptors on T cells (immune checkpoints) has been associated with clinical remission in some forms of cancer (95, 96).

Although it is undisputed that CD4 T cells provide help to B and CD8 T lymphocytes, who helps them? Years ago, my colleagues and I proposed that cooperation between CD4 T cells enables the activation and expansion of CD4 T cells specific for poorly immunogenic determinants and/or tolerized CD4 T cells, which would otherwise be unable to expand or expand only to a limited extent (97). We named this process Th–Th cooperation or “help for helpers” (98). Th–Th cooperation enables the activation of CD4 T cells specific for a self-tumor Ag, providing complete, durable, and specific protection against s.c. tumor implants and tumor rechallenge (99). The mechanism of Th–Th cooperation is based on associative recognition of Ag, where self and nonself Th cell determinants are presented by the same APC (32, 34, 100, 101). It also was found that the activation of anti-nonself T cells precedes the activation of anti-self T cells by 48 h. This provides a cytokine environment to further activate APCs, enabling the activation of otherwise unresponsive anti-self CD4 T cells (97). Furthermore, the help received through this “immunological switch” is as effective as the signals imparted using agonistic Abs to CD40 and OX40, alone or in combination (97).

The Th–Th cooperation model postulates that the anti-nonself response precedes and drives the anti-self CD4 T cell response (Fig. 1, left panel) based on a sequential three-cell interaction (Fig. 1, right panel) where the same APC processes and presents the nonself determinant and activates the corresponding anti-self CD4 T cells. Upon activation, cytokines produced by anti-nonself CD4 T cells heighten the expression of costimulatory molecules on the APC, enabling the presentation of the self determinant to CD4 T cells specific for self (102). In this model, the anti-nonself response is anticipatory of the anti-self response. A similar three-cell model was proposed for a CD4 T cell–dependent activation of CD8 T cells in which CD4 T cell activation by the APC is crucial (103). The collective value of this model is 2-fold. It provides a mechanism for otherwise subimmunogenic CD4 T cell determinants of a self-tumor Ag to break self-tolerance (102). It also points to the possibility of directly immunizing against weak CD4 T cell determinants, such as self-Ags, needed to induce protective antitumor responses in vivo by exploiting the ability of CD4 T cells to activate APCs, which, in turn, primes other (anti-self) CD4 T cells through enhanced costimulation (104).

FIGURE 1.

Temporal dynamics in the activation of CD4 T cell responses against NS and S determinants through Th–Th cooperation. NS and S determinants presented in linked association by the APC (a B lymphocyte) instruct the response of the corresponding CD4 T cells through a biphasic, sequential process. The anti-NS response is anticipatory (left panel) of the activation of anti-S CD4 T cells and is a prerequisite for their subsequent expansion through a three-cell model of dynamic interactions (right panel). NS, nonself; S, self.

FIGURE 1.

Temporal dynamics in the activation of CD4 T cell responses against NS and S determinants through Th–Th cooperation. NS and S determinants presented in linked association by the APC (a B lymphocyte) instruct the response of the corresponding CD4 T cells through a biphasic, sequential process. The anti-NS response is anticipatory (left panel) of the activation of anti-S CD4 T cells and is a prerequisite for their subsequent expansion through a three-cell model of dynamic interactions (right panel). NS, nonself; S, self.

Close modal

It appears that the balance between tolerance and immunity depends on an inherent property of the immune system: the productive cooperation between two Th cells with different specificity, one for a nonself determinant and one for a self determinant, engaged by the same APC. One prediction of this model is that attempts to activate CD4 T cells against “weak” self-tumor antigenic peptides without enabling the “immunological switch” could lead either to their inactivation (34) or to immunity without clinical response (discussed in Ref. 105). At the turn of the twentieth century, German pathologist Georg Schone noted that “the degree of immunity which develops against tumor depends on the foreignness of the immunizing cell with respect to the organism into which it is introduced. The more foreign cells accordingly serve as the more effective and the more closely related cells as the less effective antigens” (106). Th–Th cooperation through associative recognition of Ag incorporates this idea and enables the generation of protective cellular responses against self-tumor Ags. This has been applied successfully in several systems in mice and humans (107110). Thus, Th–Th cooperation can be viewed as an archetypal form of immune regulation that provides a mechanistic solution to how class determination of effector CD4 T cells with anti-self specificity is generated in vivo.

Human CD4 T cells can recognize MHC class II–restricted self-tumor Ags, such as tissue-specific Ags, common tumor Ags (i.e., Ags present in the vast majority of tumors, irrespective of their histological origin), and viral Ags causative of tumor transformation (98). In some instances, peptides from unmutated tumor Ags bind different MHC class II alleles, so-called “promiscuous” peptides (79, 111115). These peptides are therapeutically useful because they could be used to immunize a large segment of the human population.

Of particular interest, however, are those instances in which CD4 T cells are specific for MHC class II–restricted peptides corresponding to mutated genes (nonsynonymous mutations) or gene fusion (translocation) regions (MaFs) (Fig. 2). Early examples include the triosephosphate isomerase, the LDPF fusion gene product between the low-density lipid receptor and the GDP-l-fucose:b-d-galactoside-2-a-l-fucosyltransferase, and CDC27, a component of the anaphase-promoting complex involved in cell cycle regulation (116118). A recent publication reported that scleroderma patients who also developed cancer and carry a mutation in the POLR3A gene spontaneously expand MHC class II–restricted CD4 T cells specific for peptides from the mutant POLR3A gene product (119). Various other peptides were described in the past decade (120). The peptide recently identified by Tran et al. (121) corresponds to a mutation in the ERBB2IP gene. Inoculation of the patient with autologous CD4 T cells reactive against this mutated peptide caused a dramatic decrease in target lesions and prolonged stabilization of disease, possibly through direct cytotoxic activity (121).

FIGURE 2.

Process for the identification of CD4 T cell determinants from MaF gene products. Chromosomal areas containing nonsynonymous mutations or translocations, as well as attendant genes, are identified by next-generation exome sequencing. From the corresponding amino acid sequences comprising MaF gene products, discrete length peptide (∼15 aa) sequences centered in or around MaFs are then identified and cataloged. These peptides are then subject to in silico prediction for MHC class II binding (∼2800 MHC class II alleles) using bioinformatics tools (e.g., IEDB) followed by patient haplotype matching. Next, one must determine whether the identified peptides are naturally processed and presented. Ideally, this should be done in a patient-specific manner using the patient’s own tumor cells. Because this may be not be feasible, established tumor cell lines transfected with a minigene coding for the selected MaF peptide(s) should be used. At this time, no predictive criteria exist for either in vivo immunogenicity or protective value. MHC binding affinity (percentile rank) correlates with immunogenicity but does not guarantee immunogenicity. Preclinical studies in HLA-DR/DQ/DP–transgenic mice remain the only possibility before human experimentation, but this again is no guarantee that these responses are relevant to protection in vivo. As discussed in the text, it is important to interrogate the specificities of tumor-infiltrating T cells, but this may be a difficult hurdle to overcome based on tissue availability and logistics. An alternative could be to interrogate circulating T cells using, for example, MaF-specific tetramers to determine whether such T cells exist in the patient. However, detection of T cells specific for selected MaF peptides in blood cannot be considered a proxy of tumor-infiltrating lymphocytes and/or protective CD4 T lymphocytes. AA, amino acid.

FIGURE 2.

Process for the identification of CD4 T cell determinants from MaF gene products. Chromosomal areas containing nonsynonymous mutations or translocations, as well as attendant genes, are identified by next-generation exome sequencing. From the corresponding amino acid sequences comprising MaF gene products, discrete length peptide (∼15 aa) sequences centered in or around MaFs are then identified and cataloged. These peptides are then subject to in silico prediction for MHC class II binding (∼2800 MHC class II alleles) using bioinformatics tools (e.g., IEDB) followed by patient haplotype matching. Next, one must determine whether the identified peptides are naturally processed and presented. Ideally, this should be done in a patient-specific manner using the patient’s own tumor cells. Because this may be not be feasible, established tumor cell lines transfected with a minigene coding for the selected MaF peptide(s) should be used. At this time, no predictive criteria exist for either in vivo immunogenicity or protective value. MHC binding affinity (percentile rank) correlates with immunogenicity but does not guarantee immunogenicity. Preclinical studies in HLA-DR/DQ/DP–transgenic mice remain the only possibility before human experimentation, but this again is no guarantee that these responses are relevant to protection in vivo. As discussed in the text, it is important to interrogate the specificities of tumor-infiltrating T cells, but this may be a difficult hurdle to overcome based on tissue availability and logistics. An alternative could be to interrogate circulating T cells using, for example, MaF-specific tetramers to determine whether such T cells exist in the patient. However, detection of T cells specific for selected MaF peptides in blood cannot be considered a proxy of tumor-infiltrating lymphocytes and/or protective CD4 T lymphocytes. AA, amino acid.

Close modal

Recognition of MaF-derived peptides is not unique to CD4 T cells; MHC class I–restricted CD8 T cells also were reported to target peptides from the BCR-ABL and TMPRSS2-ERG fusion gene products (122, 123). A complete listing of MHC class II–restricted peptides was published recently (124). Recent findings suggest that solid tumors have an average of 33–66 genes with somatic mutations that are expected to alter their protein products (i.e., synonymous mutations) (125). As more human tumors are analyzed by exon and whole-genome sequencing, it is likely that the number of MaF-derived peptides will increase.

Are there particular properties of Ags and peptides that one should use to induce anti-tumor CD4 T cell responses in a clinical setting? MaF gene products might initially appear advantageous because MaF-derived peptides are neoantigens with CD4 T cell precursors that exist in a nontolerized form within the repertoire of the nonimmunized individual. With exon and whole-genome sequencing becoming increasingly common, algorithms already exist to rapidly match MaF regions with MHC alleles, thereby facilitating the identification of peptides for personalized cancer medicine. Yet this approach is fraught with concerns based both on the biology of the immune system and of cancer, particularly intratumor heterogeneity (126).

The first concern is that not all mutations and gene fusion products may code for peptides that bind MHC class II alleles of the individual with sufficient affinity to be immunogenic. If one can identify peptides that do bind to MHC class II, it is not clear how to determine whether these peptides can be processed and presented by the patient’s own tumor cells. These concerns can only be answered experimentally (Fig. 2). Likewise, it also would be important to identify MaF peptides that bind to different alleles and demonstrate their immunogenicity in HLA-transgenic mice (Fig. 2) (127). Provided that these steps are satisfied, what criteria will be used to predict which MaF peptides are tumor protective? Tran et al. (121) demonstrated that CD4 T cell determinants of clinical value can be identified by characterizing tumor-infiltrating lymphocytes using a library of minigenes coding for all of the possible mutations in multiple genes found in the patient’s tumor. This challenging approach is hardly applicable on a large scale. Thus, verification that putatively selected MaF peptides are recognized by tumor-infiltrating lymphocytes appears to be an unavoidable step.

In addition to the difficulties in identifying immunologically and clinically relevant MaF-derived peptides, there are concerns about intratumor heterogeneity (126). Data from genome sequencing, single-cell analyses, and multiregion sequencing point to a surprising genetic heterogeneity, including subclonal differences in driver mutations (128132). Accordingly, mutations in one cell may not be representative of mutations present in another that has grown aggressively and spread (i.e., metastasis) or in a cell residing in spatially distinct areas of the tumor. The findings suggest that MaF peptide–based immunotherapy against the MaF peptide selected as the neoantigen and tumor target could be of limited value.

Other potential concerns when vaccinating using a CD4 MaF peptide are the expression level and penetrance of the mutated protein in the tumor. For instance, Sampson et al. (133) showed that vaccination against variant III of the epidermal growth factor receptor, which is expressed in glioblastoma cells, is associated with the elimination of EGFRvIII-expressing cells at recurrence, but it did not prevent recurrence. A recent study on preclinical vaccination in mice implanted with tumors, in which not all of the cells were transduced with the IDH1R132H target gene, also found evidence of immunological escape (127). This suggests that immunological escape constitutes both a conceptual and practical obstacle. It would be of interest to know whether MaF peptides recognized by CD4 T cells can initiate epitope spreading. This concept was described originally for autoimmune diseases, but there is little evidence for this phenomenon in response to mutated cancer Ags (134, 135).

One last concern about the usefulness of vaccinating with MaF peptides is that focusing exclusively on the genomic landscape of a cancer patient ignores emerging evidence that cell nonautonomous processes condition tumor growth, tumor progression, and clonal diversity. A recent study on the dynamics of clonal repopulation using colorectal cancer cell xenografts in SCID mice showed that genetically stable clones differ with regard to proliferation and response to chemotherapy (136). Similar conclusions were reached by two other studies, both favoring the idea that tumor growth and malignant phenotypes are driven by a subpopulation of cells that can stimulate the growth of other cells in a cell-nonautonomous way (137, 138). Thus, it is increasingly likely that tumor growth and acceleration during cancer progression are independent of genetic mutations, calling for careful assessment of the cost/benefit ratio of MaF-based CD4 T cell immunotherapy.

A reasonable alternative is to focus on MHC class II–binding peptides from unmutated sequences of already identified Ags, such as telomerase, survivin, MUC.1, and HER-2, which are widely overexpressed in human cancers. These frequently overexpressed proteins could lend themselves to inclusion in vaccines designed to exploit Th–Th cooperation (139143). Presentation of unmutated peptides by the APC, along with a suitably selected nonself CD4 T cell determinant, may prove sufficient for immunological and clinical effects. In addition, there is evidence for epitope spreading following immunization of cancer patients with unmutated tumor Ag peptides (144149). Although no single MHC class II determinant can cover the entire human population (there are 2870 MHC class II alleles in the Immuno Polymorphism Database-ImMunoGeneTics-Human Leukocyte Antigen release as of November 2014), epitopes can be selected on the basis of the frequency of the MHC class II alleles binding to MHC class II supertypes or binding multiple alleles (promiscuous peptides) (79, 150, 151). This represents a simpler and more economical approach compared with those centered around the use of unique peptides from MaF gene products. Advantages and disadvantages exist: one advantage is that the expansion of CD4 T cells that specifically recognize tumor self-Ags could be a source of help for antitumor CD8 T cells locally in the tumor microenvironment (152). The activation of self-reactive T cells must not pass a critical threshold to avoid the clinical manifestations of autoimmunity against normal tissues expressing the same Ag or tissues expressing other self-Ags via bystander help. However, it was argued and demonstrated that a tolerable degree of autoimmunity is a key aspect of successful cancer immunotherapy (153, 154).

As with all new therapeutic modalities, it is important to consider the regulatory process that lies ahead. Although the U.S. Food and Drug Administration (FDA) has approved new biologic therapeutics for cancer at an unprecedented and surprisingly expedited pace (e.g., the mAb pembrolizumab for melanoma), the future of therapeutic vaccines that vary from patient to patient as a result of the nature of the immunogens and the MHC polymorphism remains unclear. Designer personalized cancer vaccines may not be immediately embraced by the FDA but will require a slow process of adaptation and adoption of new measures as we transition into a new regulatory era. Currently, the most expedited approach might be to seek approval for the methods needed to prepare the delivery of MaF-based new therapies (i.e., approval could be granted to the process rather than to the end product). Furthermore, the approval of investigational new drug applications could be expedited if these were limited to one specific mutation, such as KRAS (which is relevant for colon, lung, and pancreatic cancers) or epidermal growth factor receptor (lung cancer and glioblastoma), because these are regarded as cancer-driving mutations (125). The identification of a promiscuous peptide within a mutated region, as recently demonstrated for IDH1 (127), also would simplify the process. Perhaps these and other considerations together with anecdotal successes, such as the one reported recently by Tran et al. (121), would encourage a dialogue between regulatory agencies and proponents of the new approach to find acceptable solutions, shape a new policy, and avoid tempering the current enthusiasm for genomic-based interventions that target the immune system. This appears to be the spirit of “Paving the Way for Personalized Medicine: FDA’s Role in a New Era of Medical Product Development” released by the FDA in October 2013 (155).

There exists little doubt that CD4 T cell–based therapies, and vaccination in particular, will play a relevant role in tumor control and patient management in the future. One key issue is whether to focus on MaF neoantigens on an individual basis at a cost that may not be affordable and without a guarantee of durable success or to focus on therapeutic vaccines using MHC class II–binding peptides from unmutated sequences of already-characterized tumor Ags along the principle of Th–Th cooperation. Focus on MaF gene products appears to be a logical solution to an immunological quandary with promise for clinical benefit. However, as discussed in this article, there exist many conceptual and practical hurdles. This approach may not be a viable option for all tumors; perhaps only tumors carrying specific translocations may be suited for this approach. In addition, the existing evidence is limited to mostly anecdotal reports, and the long-term success of this approach remains uncertain. There are also financial considerations, including high costs, uncertainties about FDA approval, and likely little return on investment given the small size of patient population, which may hinder the development of such personalized cancer vaccines. In contrast, peptides from the unmutated sequence of cancer-relevant Ags could simplify vaccine production, thereby benefitting a large fraction of cancer patients at a much lower cost. The answer to this timely question may influence the direction of future efforts for effective cancer immunotherapies.

I thank Drs. Alessandro Sette, Antonella Vitiello, Maurizio Sollazzo, Razelle Kurzrock, and Navin Mahadevan for comments and helpful suggestions.

Abbreviations used in this article:

FDA

U.S. Food and Drug Administration

MaF

mutated genes and gene fusion regions.

1
Waldmann
T. A.
2003
.
Immunotherapy: past, present and future.
Nat. Med.
9
:
269
277
.
2
Baeuerle
P. A.
,
Reinhardt
C.
.
2009
.
Bispecific T-cell engaging antibodies for cancer therapy.
Cancer Res.
69
:
4941
4944
.
3
Pillay
V.
,
Gan
H. K.
,
Scott
A. M.
.
2011
.
Antibodies in oncology.
New Biotechnol.
28
:
518
529
.
4
Jachimowicz
R. D.
,
Borchmann
S.
,
Rothe
A.
.
2014
.
Multi-specific antibodies for cancer immunotherapy.
BioDrugs
28
:
331
343
.
5
Burnet
F. M.
1971
.
Immunological surveillance in neoplasia.
Transplant. Rev.
7
:
3
25
.
6
Schreiber
R. D.
,
Old
L. J.
,
Smyth
M. J.
.
2011
.
Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion.
Science
331
:
1565
1570
.
7
Bottino
C.
,
Moretta
L.
,
Moretta
A.
.
2006
.
NK cell activating receptors and tumor recognition in humans.
Curr. Top. Microbiol. Immunol.
298
:
175
182
.
8
Faveeuw
C.
,
Trottein
F.
.
2014
.
Optimization of natural killer T cell-mediated immunotherapy in cancer using cell-based and nanovector vaccines.
Cancer Res.
74
:
1632
1638
.
9
Jiang
J.
,
Wu
C.
,
Lu
B.
.
2013
.
Cytokine-induced killer cells promote antitumor immunity.
J. Transl. Med.
11
:
83
.
10
Koretz
K.
,
Moldenhauer
G.
,
Majdic
O.
,
Möller
P.
.
1989
.
Correlation of HLA-D/Ii antigen expression in breast carcinoma with local lymphohistiocytic infiltration reveals considerable dysregulation in a subset of tumors.
Int. J. Cancer
44
:
816
822
.
11
Marincola
F. M.
,
Jaffee
E. M.
,
Hicklin
D. J.
,
Ferrone
S.
.
2000
.
Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance.
Adv. Immunol.
74
:
181
273
.
12
Möller
P.
,
Mattfeldt
T.
,
Gross
C.
,
Schlosshauer
P.
,
Koch
A.
,
Koretz
K.
,
Moldenhauer
G.
,
Kaufmann
M.
,
Otto
H. F.
.
1989
.
Expression of HLA-A, -B, -C, -DR, -DP, -DQ, and of HLA-D-associated invariant chain (Ii) in non-neoplastic mammary epithelium, fibroadenoma, adenoma, and carcinoma of the breast.
Am. J. Pathol.
135
:
73
83
.
13
Offringa
R.
,
van der Burg
S. H.
,
Ossendorp
F.
,
Toes
R. E.
,
Melief
C. J.
.
2000
.
Design and evaluation of antigen-specific vaccination strategies against cancer.
Curr. Opin. Immunol.
12
:
576
582
.
14
Brossart
P.
,
Stuhler
G.
,
Flad
T.
,
Stevanovic
S.
,
Rammensee
H. G.
,
Kanz
L.
,
Brugger
W.
.
1998
.
Her-2/neu-derived peptides are tumor-associated antigens expressed by human renal cell and colon carcinoma lines and are recognized by in vitro induced specific cytotoxic T lymphocytes.
Cancer Res.
58
:
732
736
.
15
Molldrem
J. J.
,
Lee
P. P.
,
Wang
C.
,
Felio
K.
,
Kantarjian
H. M.
,
Champlin
R. E.
,
Davis
M. M.
.
2000
.
Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia.
Nat. Med.
6
:
1018
1023
.
16
Filaci
G.
,
Fravega
M.
,
Setti
M.
,
Traverso
P.
,
Millo
E.
,
Fenoglio
D.
,
Negrini
S.
,
Ferrera
F.
,
Romagnoli
A.
,
Basso
M.
, et al
.
2006
.
Frequency of telomerase-specific CD8+ T lymphocytes in patients with cancer.
Blood
107
:
1505
1512
.
17
Ahmadzadeh
M.
,
Johnson
L. A.
,
Heemskerk
B.
,
Wunderlich
J. R.
,
Dudley
M. E.
,
White
D. E.
,
Rosenberg
S. A.
.
2009
.
Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired.
Blood
114
:
1537
1544
.
18
Rosenberg
S. A.
,
Yang
J. C.
,
Schwartzentruber
D. J.
,
Hwu
P.
,
Marincola
F. M.
,
Topalian
S. L.
,
Restifo
N. P.
,
Dudley
M. E.
,
Schwarz
S. L.
,
Spiess
P. J.
, et al
.
1998
.
Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma.
Nat. Med.
4
:
321
327
.
19
Rosenberg
S. A.
,
Yang
J. C.
,
Restifo
N. P.
.
2004
.
Cancer immunotherapy: moving beyond current vaccines.
Nat. Med.
10
:
909
915
.
20
Klebanoff
C. A.
,
Acquavella
N.
,
Yu
Z.
,
Restifo
N. P.
.
2011
.
Therapeutic cancer vaccines: are we there yet?
Immunol. Rev.
239
:
27
44
.
21
Flemming
A.
2014
.
Deal watch: Pfizer and GSK join race for T cell cancer therapies.
Nat. Rev. Drug Discov.
13
:
568
569
.
22
June
C.
,
Rosenberg
S. A.
,
Sadelain
M.
,
Weber
J. S.
.
2012
.
T-cell therapy at the threshold.
Nat. Biotechnol.
30
:
611
614
.
23
Porter
D. L.
,
Levine
B. L.
,
Kalos
M.
,
Bagg
A.
,
June
C. H.
.
2011
.
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia.
N. Engl. J. Med.
365
:
725
733
.
24
Kochenderfer
J. N.
,
Dudley
M. E.
,
Feldman
S. A.
,
Wilson
W. H.
,
Spaner
D. E.
,
Maric
I.
,
Stetler-Stevenson
M.
,
Phan
G. Q.
,
Hughes
M. S.
,
Sherry
R. M.
, et al
.
2012
.
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
119
:
2709
2720
.
25
Vacchelli
E.
,
Eggermont
A.
,
Fridman
W. H.
,
Galon
J.
,
Tartour
E.
,
Zitvogel
L.
,
Kroemer
G.
,
Galluzzi
L.
.
2013
.
Trial Watch: Adoptive cell transfer for anticancer immunotherapy.
OncoImmunology
2
:
e24238
.
26
Ruella
M.
,
Kalos
M.
.
2014
.
Adoptive immunotherapy for cancer.
Immunol. Rev.
257
:
14
38
.
27
Couzin-Frankel
J.
2013
.
Breakthrough of the year 2013. Cancer immunotherapy.
Science
342
:
1432
1433
.
28
Kim
H. J.
,
Cantor
H.
.
2014
.
CD4 T-cell subsets and tumor immunity: the helpful and the not-so-helpful.
Cancer Immunol Res
2
:
91
98
.
29
Claman
H. N.
,
Chaperon
E. A.
,
Triplett
R. F.
.
1966
.
Thymus-marrow cell combinations. Synergism in antibody production.
Proc. Soc. Exp. Biol. Med.
122
:
1167
1171
.
30
Mitchison
N. A.
1971
.
The carrier effect in the secondary response to hapten-protein conjugates. II. Cellular cooperation.
Eur. J. Immunol.
1
:
18
27
.
31
Lanzavecchia
A.
1985
.
Antigen-specific interaction between T and B cells.
Nature
314
:
537
539
.
32
Bretscher
P.
,
Cohn
M.
.
1970
.
A theory of self-nonself discrimination.
Science
169
:
1042
1049
.
33
Lake
P.
,
Mitchison
N. A.
.
1977
.
Regulatory mechanisms in the immune response to cell-surface antigens.
Cold Spring Harb. Symp. Quant. Biol.
41
:
589
595
.
34
Cohn
M.
2005
.
The common sense of the self-nonself discrimination.
Springer Semin. Immunopathol.
27
:
3
17
.
35
Cassell
D.
,
Forman
J.
.
1988
.
Linked recognition of helper and cytotoxic antigenic determinants for the generation of cytotoxic T lymphocytes.
Ann. N. Y. Acad. Sci.
532
:
51
60
.
36
Janssen
E. M.
,
Lemmens
E. E.
,
Wolfe
T.
,
Christen
U.
,
von Herrath
M. G.
,
Schoenberger
S. P.
.
2003
.
CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes.
Nature
421
:
852
856
.
37
Shedlock
D. J.
,
Shen
H.
.
2003
.
Requirement for CD4 T cell help in generating functional CD8 T cell memory.
Science
300
:
337
339
.
38
Sun
J. C.
,
Bevan
M. J.
.
2003
.
Defective CD8 T cell memory following acute infection without CD4 T cell help.
Science
300
:
339
342
.
39
Langlade-Demoyen
P.
,
Garcia-Pons
F.
,
Castiglioni
P.
,
Garcia
Z.
,
Cardinaud
S.
,
Xiong
S.
,
Gerloni
M.
,
Zanetti
M.
.
2003
.
Role of T cell help and endoplasmic reticulum targeting in protective CTL response against influenza virus.
Eur. J. Immunol.
33
:
720
728
.
40
Mosmann
T. R.
,
Coffman
R. L.
.
1989
.
TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties.
Annu. Rev. Immunol.
7
:
145
173
.
41
Sakaguchi
S.
,
Sakaguchi
N.
,
Shimizu
J.
,
Yamazaki
S.
,
Sakihama
T.
,
Itoh
M.
,
Kuniyasu
Y.
,
Nomura
T.
,
Toda
M.
,
Takahashi
T.
.
2001
.
Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance.
Immunol. Rev.
182
:
18
32
.
42
Crotty
S.
2011
.
Follicular helper CD4 T cells (TFH).
Annu. Rev. Immunol.
29
:
621
663
.
43
Greenberg
P. D.
,
Cheever
M. A.
,
Fefer
A.
.
1981
.
Eradication of disseminated murine leukemia by chemoimmunotherapy with cyclophosphamide and adoptively transferred immune syngeneic Lyt-1+2- lymphocytes.
J. Exp. Med.
154
:
952
963
.
44
Fujiwara
H.
,
Fukuzawa
M.
,
Yoshioka
T.
,
Nakajima
H.
,
Hamaoka
T.
.
1984
.
The role of tumor-specific Lyt-1+2− T cells in eradicating tumor cells in vivo. I. Lyt-1+2− T cells do not necessarily require recruitment of host’s cytotoxic T cell precursors for implementation of in vivo immunity.
J. Immunol.
133
:
1671
1676
.
45
Hock
H.
,
Dorsch
M.
,
Diamantstein
T.
,
Blankenstein
T.
.
1991
.
Interleukin 7 induces CD4+ T cell-dependent tumor rejection.
J. Exp. Med.
174
:
1291
1298
.
46
Lauritzsen
G. F.
,
Weiss
S.
,
Dembic
Z.
,
Bogen
B.
.
1994
.
Naive idiotype-specific CD4+ T cells and immunosurveillance of B-cell tumors.
Proc. Natl. Acad. Sci. USA
91
:
5700
5704
.
47
Hung
K.
,
Hayashi
R.
,
Lafond-Walker
A.
,
Lowenstein
C.
,
Pardoll
D.
,
Levitsky
H.
.
1998
.
The central role of CD4(+) T cells in the antitumor immune response.
J. Exp. Med.
188
:
2357
2368
.
48
Mumberg
D.
,
Monach
P. A.
,
Wanderling
S.
,
Philip
M.
,
Toledano
A. Y.
,
Schreiber
R. D.
,
Schreiber
H.
.
1999
.
CD4(+) T cells eliminate MHC class II-negative cancer cells in vivo by indirect effects of IFN-gamma.
Proc. Natl. Acad. Sci. USA
96
:
8633
8638
.
49
Tempero
R. M.
,
VanLith
M. L.
,
Morikane
K.
,
Rowse
G. J.
,
Gendler
S. J.
,
Hollingsworth
M. A.
.
1998
.
CD4+ lymphocytes provide MUC1-specific tumor immunity in vivo that is undetectable in vitro and is absent in MUC1 transgenic mice.
J. Immunol.
161
:
5500
5506
.
50
Greenberg
P. D.
1991
.
Adoptive T cell therapy of tumors: mechanisms operative in the recognition and elimination of tumor cells.
Adv. Immunol.
49
:
281
355
.
51
Dighe
A. S.
,
Richards
E.
,
Old
L. J.
,
Schreiber
R. D.
.
1994
.
Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors.
Immunity
1
:
447
456
.
52
Williamson
B. D.
,
Carswell
E. A.
,
Rubin
B. Y.
,
Prendergast
J. S.
,
Old
L. J.
.
1983
.
Human tumor necrosis factor produced by human B-cell lines: synergistic cytotoxic interaction with human interferon.
Proc. Natl. Acad. Sci. USA
80
:
5397
5401
.
53
Fransen
L.
,
Van der Heyden
J.
,
Ruysschaert
R.
,
Fiers
W.
.
1986
.
Recombinant tumor necrosis factor: its effect and its synergism with interferon-gamma on a variety of normal and transformed human cell lines.
Eur. J. Cancer Clin. Oncol.
22
:
419
426
.
54
Coughlin
C. M.
,
Salhany
K. E.
,
Gee
M. S.
,
LaTemple
D. C.
,
Kotenko
S.
,
Ma
X.
,
Gri
G.
,
Wysocka
M.
,
Kim
J. E.
,
Liu
L.
, et al
.
1998
.
Tumor cell responses to IFNgamma affect tumorigenicity and response to IL-12 therapy and antiangiogenesis.
Immunity
9
:
25
34
.
55
Qin
Z.
,
Blankenstein
T.
.
2000
.
CD4+ T cell–mediated tumor rejection involves inhibition of angiogenesis that is dependent on IFN gamma receptor expression by nonhematopoietic cells.
Immunity
12
:
677
686
.
56
Corthay
A.
,
Skovseth
D. K.
,
Lundin
K. U.
,
Røsjø
E.
,
Omholt
H.
,
Hofgaard
P. O.
,
Haraldsen
G.
,
Bogen
B.
.
2005
.
Primary antitumor immune response mediated by CD4+ T cells.
Immunity
22
:
371
383
.
57
Haabeth
O. A.
,
Lorvik
K. B.
,
Hammarström
C.
,
Donaldson
I. M.
,
Haraldsen
G.
,
Bogen
B.
,
Corthay
A.
.
2011
.
Inflammation driven by tumour-specific Th1 cells protects against B-cell cancer.
Nat. Commun.
2
:
240
.
58
Xie
Y.
,
Akpinarli
A.
,
Maris
C.
,
Hipkiss
E. L.
,
Lane
M.
,
Kwon
E. K.
,
Muranski
P.
,
Restifo
N. P.
,
Antony
P. A.
.
2010
.
Naive tumor-specific CD4(+) T cells differentiated in vivo eradicate established melanoma.
J. Exp. Med.
207
:
651
667
.
59
Quezada
S. A.
,
Simpson
T. R.
,
Peggs
K. S.
,
Merghoub
T.
,
Vider
J.
,
Fan
X.
,
Blasberg
R.
,
Yagita
H.
,
Muranski
P.
,
Antony
P. A.
, et al
.
2010
.
Tumor-reactive CD4(+) T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts.
J. Exp. Med.
207
:
637
650
.
60
Horna
P.
,
Cuenca
A.
,
Cheng
F.
,
Brayer
J.
,
Wang
H. W.
,
Borrello
I.
,
Levitsky
H.
,
Sotomayor
E. M.
.
2006
.
In vivo disruption of tolerogenic cross-presentation mechanisms uncovers an effective T-cell activation by B-cell lymphomas leading to antitumor immunity.
Blood
107
:
2871
2878
.
61
Perez-Diez
A.
,
Joncker
N. T.
,
Choi
K.
,
Chan
W. F.
,
Anderson
C. C.
,
Lantz
O.
,
Matzinger
P.
.
2007
.
CD4 cells can be more efficient at tumor rejection than CD8 cells.
Blood
109
:
5346
5354
.
62
Vlad
A. M.
,
Muller
S.
,
Cudic
M.
,
Paulsen
H.
,
Otvos
L.
 Jr.
,
Hanisch
F. G.
,
Finn
O. J.
.
2002
.
Complex carbohydrates are not removed during processing of glycoproteins by dendritic cells: processing of tumor antigen MUC1 glycopeptides for presentation to major histocompatibility complex class II-restricted T cells.
J. Exp. Med.
196
:
1435
1446
.
63
Mucida
D.
,
Husain
M. M.
,
Muroi
S.
,
van Wijk
F.
,
Shinnakasu
R.
,
Naoe
Y.
,
Reis
B. S.
,
Huang
Y.
,
Lambolez
F.
,
Docherty
M.
, et al
.
2013
.
Transcriptional reprogramming of mature CD4+ helper T cells generates distinct MHC class II-restricted cytotoxic T lymphocytes.
Nat. Immunol.
14
:
281
289
.
64
Zeng
G.
,
Touloukian
C. E.
,
Wang
X.
,
Restifo
N. P.
,
Rosenberg
S. A.
,
Wang
R. F.
.
2000
.
Identification of CD4+ T cell epitopes from NY-ESO-1 presented by HLA-DR molecules.
J. Immunol.
165
:
1153
1159
.
65
Zeng
G.
,
Wang
X.
,
Robbins
P. F.
,
Rosenberg
S. A.
,
Wang
R. F.
.
2001
.
CD4(+) T cell recognition of MHC class II-restricted epitopes from NY-ESO-1 presented by a prevalent HLA DP4 allele: association with NY-ESO-1 antibody production.
Proc. Natl. Acad. Sci. USA
98
:
3964
3969
.
66
Campi
G.
,
Crosti
M.
,
Consogno
G.
,
Facchinetti
V.
,
Conti-Fine
B. M.
,
Longhi
R.
,
Casorati
G.
,
Dellabona
P.
,
Protti
M. P.
.
2003
.
CD4(+) T cells from healthy subjects and colon cancer patients recognize a carcinoembryonic antigen-specific immunodominant epitope.
Cancer Res.
63
:
8481
8486
.
67
Kudela
P.
,
Sun
Z.
,
Fourcade
J.
,
Janjic
B.
,
Kirkwood
J. M.
,
Maillere
B.
,
Zarour
H. M.
.
2011
.
Epitope hierarchy of spontaneous CD4+ T cell responses to LAGE-1.
J. Immunol.
186
:
312
322
.
68
Ohue
Y.
,
Eikawa
S.
,
Okazaki
N.
,
Mizote
Y.
,
Isobe
M.
,
Uenaka
A.
,
Fukuda
M.
,
Old
L. J.
,
Oka
M.
,
Nakayama
E.
.
2012
.
Spontaneous antibody, and CD4 and CD8 T-cell responses against XAGE-1b (GAGED2a) in non-small cell lung cancer patients.
Int. J. Cancer
131
:
E649
E658
.
69
Tsuji
T.
,
Matsuzaki
J.
,
Ritter
E.
,
Miliotto
A.
,
Ritter
G.
,
Odunsi
K.
,
Old
L. J.
,
Gnjatic
S.
.
2011
.
Split T cell tolerance against a self/tumor antigen: spontaneous CD4+ but not CD8+ T cell responses against p53 in cancer patients and healthy donors.
PLoS ONE
6
:
e23651
.
70
Munir
S.
,
Larsen
S. K.
,
Iversen
T. Z.
,
Donia
M.
,
Klausen
T. W.
,
Svane
I. M.
,
Straten
P. T.
,
Andersen
M. H.
.
2012
.
Natural CD4+ T-cell responses against indoleamine 2,3-dioxygenase.
PLoS ONE
7
:
e34568
.
71
Galon
J.
,
Costes
A.
,
Sanchez-Cabo
F.
,
Kirilovsky
A.
,
Mlecnik
B.
,
Lagorce-Pagès
C.
,
Tosolini
M.
,
Camus
M.
,
Berger
A.
,
Wind
P.
, et al
.
2006
.
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome.
Science
313
:
1960
1964
.
72
Yoshida
N.
,
Abe
H.
,
Ohkuri
T.
,
Wakita
D.
,
Sato
M.
,
Noguchi
D.
,
Miyamoto
M.
,
Morikawa
T.
,
Kondo
S.
,
Ikeda
H.
,
Nishimura
T.
.
2006
.
Expression of the MAGE-A4 and NY-ESO-1 cancer-testis antigens and T cell infiltration in non-small cell lung carcinoma and their prognostic significance.
Int. J. Oncol.
28
:
1089
1098
.
73
Ayyoub
M.
,
Pignon
P.
,
Classe
J. M.
,
Odunsi
K.
,
Valmori
D.
.
2013
.
CD4+ T effectors specific for the tumor antigen NY-ESO-1 are highly enriched at ovarian cancer sites and coexist with, but are distinct from, tumor-associated Treg.
Cancer Immunol Res
1
:
303
308
.
74
Moreno
A.
,
Clavijo
P.
,
Edelman
R.
,
Davis
J.
,
Sztein
M.
,
Herrington
D.
,
Nardin
E.
.
1991
.
Cytotoxic CD4+ T cells from a sporozoite-immunized volunteer recognize the Plasmodium falciparum CS protein.
Int. Immunol.
3
:
997
1003
.
75
Barnaba
V.
,
Franco
A.
,
Paroli
M.
,
Benvenuto
R.
,
De Petrillo
G.
,
Burgio
V. L.
,
Santilio
I.
,
Balsano
C.
,
Bonavita
M. S.
,
Cappelli
G.
, et al
.
1994
.
Selective expansion of cytotoxic T lymphocytes with a CD4+CD56+ surface phenotype and a T helper type 1 profile of cytokine secretion in the liver of patients chronically infected with Hepatitis B virus.
J. Immunol.
152
:
3074
3087
.
76
Nisini
R.
,
Paroli
M.
,
Accapezzato
D.
,
Bonino
F.
,
Rosina
F.
,
Santantonio
T.
,
Sallusto
F.
,
Amoroso
A.
,
Houghton
M.
,
Barnaba
V.
.
1997
.
Human CD4+ T-cell response to hepatitis delta virus: identification of multiple epitopes and characterization of T-helper cytokine profiles.
J. Virol.
71
:
2241
2251
.
77
Thomas
W. D.
,
Hersey
P.
.
1998
.
CD4 T cells kill melanoma cells by mechanisms that are independent of Fas (CD95).
Int. J. Cancer
75
:
384
390
.
78
Soghoian
D. Z.
,
Jessen
H.
,
Flanders
M.
,
Sierra-Davidson
K.
,
Cutler
S.
,
Pertel
T.
,
Ranasinghe
S.
,
Lindqvist
M.
,
Davis
I.
,
Lane
K.
, et al
.
2012
.
HIV-specific cytolytic CD4 T cell responses during acute HIV infection predict disease outcome.
Sci. Transl. Med.
4
:
23ra25
.
79
Manici
S.
,
Sturniolo
T.
,
Imro
M. A.
,
Hammer
J.
,
Sinigaglia
F.
,
Noppen
C.
,
Spagnoli
G.
,
Mazzi
B.
,
Bellone
M.
,
Dellabona
P.
,
Protti
M. P.
.
1999
.
Melanoma cells present a MAGE-3 epitope to CD4(+) cytotoxic T cells in association with histocompatibility leukocyte antigen DR11.
J. Exp. Med.
189
:
871
876
.
80
Hombach
A.
,
Köhler
H.
,
Rappl
G.
,
Abken
H.
.
2006
.
Human CD4+ T cells lyse target cells via granzyme/perforin upon circumvention of MHC class II restriction by an antibody-like immunoreceptor.
J. Immunol.
177
:
5668
5675
.
81
Hess
S. D.
,
Egilmez
N. K.
,
Bailey
N.
,
Anderson
T. M.
,
Mathiowitz
E.
,
Bernstein
S. H.
,
Bankert
R. B.
.
2003
.
Human CD4+ T cells present within the microenvironment of human lung tumors are mobilized by the local and sustained release of IL-12 to kill tumors in situ by indirect effects of IFN-gamma.
J. Immunol.
170
:
400
412
.
82
Viola
A.
,
Lanzavecchia
A.
.
1996
.
T cell activation determined by T cell receptor number and tunable thresholds.
Science
273
:
104
106
.
83
Sykulev
Y.
,
Joo
M.
,
Vturina
I.
,
Tsomides
T. J.
,
Eisen
H. N.
.
1996
.
Evidence that a single peptide-MHC complex on a target cell can elicit a cytolytic T cell response.
Immunity
4
:
565
571
.
84
Sercarz
E. E.
,
Lehmann
P. V.
,
Ametani
A.
,
Benichou
G.
,
Miller
A.
,
Moudgil
K.
.
1993
.
Dominance and crypticity of T cell antigenic determinants.
Annu. Rev. Immunol.
11
:
729
766
.
85
Gabrilovich
D. I.
,
Ostrand-Rosenberg
S.
,
Bronte
V.
.
2012
.
Coordinated regulation of myeloid cells by tumours.
Nat. Rev. Immunol.
12
:
253
268
.
86
Kappler
J. W.
,
Roehm
N.
,
Marrack
P.
.
1987
.
T cell tolerance by clonal elimination in the thymus.
Cell
49
:
273
280
.
87
Cobbold
S. P.
,
Adams
E.
,
Marshall
S. E.
,
Davies
J. D.
,
Waldmann
H.
.
1996
.
Mechanisms of peripheral tolerance and suppression induced by monoclonal antibodies to CD4 and CD8.
Immunol. Rev.
149
:
5
33
.
88
Ochsenbein
A. F.
,
Klenerman
P.
,
Karrer
U.
,
Ludewig
B.
,
Pericin
M.
,
Hengartner
H.
,
Zinkernagel
R. M.
.
1999
.
Immune surveillance against a solid tumor fails because of immunological ignorance.
Proc. Natl. Acad. Sci. USA
96
:
2233
2238
.
89
Mahadevan
N. R.
,
Zanetti
M.
.
2011
.
Tumor stress inside out: cell-extrinsic effects of the unfolded protein response in tumor cells modulate the immunological landscape of the tumor microenvironment.
J. Immunol.
187
:
4403
4409
.
90
Wing
K.
,
Sakaguchi
S.
.
2010
.
Regulatory T cells exert checks and balances on self tolerance and autoimmunity.
Nat. Immunol.
11
:
7
13
.
91
Grivennikov
S. I.
,
Greten
F. R.
,
Karin
M.
.
2010
.
Immunity, inflammation, and cancer.
Cell
140
:
883
899
.
92
Willimsky
G.
,
Blankenstein
T.
.
2005
.
Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance.
Nature
437
:
141
146
.
93
Monsurrò
V.
,
Wang
E.
,
Yamano
Y.
,
Migueles
S. A.
,
Panelli
M. C.
,
Smith
K.
,
Nagorsen
D.
,
Connors
M.
,
Jacobson
S.
,
Marincola
F. M.
.
2004
.
Quiescent phenotype of tumor-specific CD8+ T cells following immunization.
Blood
104
:
1970
1978
.
94
Schietinger
A.
,
Delrow
J. J.
,
Basom
R. S.
,
Blattman
J. N.
,
Greenberg
P. D.
.
2012
.
Rescued tolerant CD8 T cells are preprogrammed to reestablish the tolerant state.
Science
335
:
723
727
.
95
Brahmer
J. R.
,
Tykodi
S. S.
,
Chow
L. Q.
,
Hwu
W. J.
,
Topalian
S. L.
,
Hwu
P.
,
Drake
C. G.
,
Camacho
L. H.
,
Kauh
J.
,
Odunsi
K.
, et al
.
2012
.
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer.
N. Engl. J. Med.
366
:
2455
2465
.
96
Topalian
S. L.
,
Hodi
F. S.
,
Brahmer
J. R.
,
Gettinger
S. N.
,
Smith
D. C.
,
McDermott
D. F.
,
Powderly
J. D.
,
Carvajal
R. D.
,
Sosman
J. A.
,
Atkins
M. B.
, et al
.
2012
.
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer.
N. Engl. J. Med.
366
:
2443
2454
.
97
Gerloni
M.
,
Xiong
S.
,
Mukerjee
S.
,
Schoenberger
S. P.
,
Croft
M.
,
Zanetti
M.
.
2000
.
Functional cooperation between T helper cell determinants.
Proc. Natl. Acad. Sci. USA
97
:
13269
13274
.
98
Gerloni
M.
,
Zanetti
M.
.
2005
.
CD4 T cells in tumor immunity.
Springer Semin. Immunopathol.
27
:
37
48
.
99
Gerloni
M.
,
Castiglioni
P.
,
Zanetti
M.
.
2005
.
The cooperation between two CD4 T cells induces tumor protective immunity in MUC.1 transgenic mice.
J. Immunol.
175
:
6551
6559
.
100
Bretscher
P. A.
1986
.
A cascade of T-T interactions, mediated by the linked recognition of antigen, in the induction of T cells able to help delayed-type hypersensitivity responses.
J. Immunol.
137
:
3726
3733
.
101
Bretscher
P. A.
1999
.
A two-step, two-signal model for the primary activation of precursor helper T cells.
Proc. Natl. Acad. Sci. USA
96
:
185
190
.
102
Zanetti
M.
2005
.
T for two: when helpers need help.
Autoimmun. Rev.
4
:
571
578
.
103
Schoenberger
S. P.
,
Toes
R. E.
,
van der Voort
E. I.
,
Offringa
R.
,
Melief
C. J.
.
1998
.
T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions.
Nature
393
:
480
483
.
104
den Haan
J. M.
,
Bevan
M. J.
.
2000
.
A novel helper role for CD4 T cells.
Proc. Natl. Acad. Sci. USA
97
:
12950
12952
.
105
Kyte
J. A.
2009
.
Cancer vaccination with telomerase peptide GV1001.
Expert Opin. Investig. Drugs
18
:
687
694
.
106
Silverstein, A. M. 1989. A History of Immunology. Academic Press, San Diego, CA
.
107
Savelyeva
N.
,
Munday
R.
,
Spellerberg
M. B.
,
Lomonossoff
G. P.
,
Stevenson
F. K.
.
2001
.
Plant viral genes in DNA idiotypic vaccines activate linked CD4+ T-cell mediated immunity against B-cell malignancies.
Nat. Biotechnol.
19
:
760
764
.
108
Stevenson
F. K.
,
Ottensmeier
C. H.
,
Johnson
P.
,
Zhu
D.
,
Buchan
S. L.
,
McCann
K. J.
,
Roddick
J. S.
,
King
A. T.
,
McNicholl
F.
,
Savelyeva
N.
,
Rice
J.
.
2004
.
DNA vaccines to attack cancer.
Proc. Natl. Acad. Sci. USA
101
(
Suppl. 2
):
14646
14652
.
109
Facciabene
A.
,
Aurisicchio
L.
,
Elia
L.
,
Palombo
F.
,
Mennuni
C.
,
Ciliberto
G.
,
La Monica
N.
.
2006
.
DNA and adenoviral vectors encoding carcinoembryonic antigen fused to immunoenhancing sequences augment antigen-specific immune response and confer tumor protection.
Hum. Gene Ther.
17
:
81
92
.
110
Snook
A. E.
,
Magee
M. S.
,
Schulz
S.
,
Waldman
S. A.
.
2014
.
Selective antigen-specific CD4(+) T-cell, but not CD8(+) T- or B-cell, tolerance corrupts cancer immunotherapy.
Eur. J. Immunol.
44
:
1956
1966
.
111
Kobayashi
H.
,
Wood
M.
,
Song
Y.
,
Appella
E.
,
Celis
E.
.
2000
.
Defining promiscuous MHC class II helper T-cell epitopes for the HER2/neu tumor antigen.
Cancer Res.
60
:
5228
5236
.
112
Zarour
H. M.
,
Maillere
B.
,
Brusic
V.
,
Coval
K.
,
Williams
E.
,
Pouvelle-Moratille
S.
,
Castelli
F.
,
Land
S.
,
Bennouna
J.
,
Logan
T.
,
Kirkwood
J. M.
.
2002
.
NY-ESO-1 119-143 is a promiscuous major histocompatibility complex class II T-helper epitope recognized by Th1- and Th2-type tumor-reactive CD4+ T cells.
Cancer Res.
62
:
213
218
.
113
Consogno
G.
,
Manici
S.
,
Facchinetti
V.
,
Bachi
A.
,
Hammer
J.
,
Conti-Fine
B. M.
,
Rugarli
C.
,
Traversari
C.
,
Protti
M. P.
.
2003
.
Identification of immunodominant regions among promiscuous HLA-DR-restricted CD4+ T-cell epitopes on the tumor antigen MAGE-3.
Blood
101
:
1038
1044
.
114
Neumann
F.
,
Wagner
C.
,
Stevanovic
S.
,
Kubuschok
B.
,
Schormann
C.
,
Mischo
A.
,
Ertan
K.
,
Schmidt
W.
,
Pfreundschuh
M.
.
2004
.
Identification of an HLA-DR-restricted peptide epitope with a promiscuous binding pattern derived from the cancer testis antigen HOM-MEL-40/SSX2.
Int. J. Cancer
112
:
661
668
.
115
Wang
X. F.
,
Kerzerho
J.
,
Adotevi
O.
,
Nuyttens
H.
,
Badoual
C.
,
Munier
G.
,
Oudard
S.
,
Tu
S.
,
Tartour
E.
,
Maillère
B.
.
2008
.
Comprehensive analysis of HLA-DR- and HLA-DP4-restricted CD4+ T cell response specific for the tumor-shared antigen survivin in healthy donors and cancer patients.
J. Immunol.
181
:
431
439
.
116
Pieper
R.
,
Christian
R. E.
,
Gonzales
M. I.
,
Nishimura
M. I.
,
Gupta
G.
,
Settlage
R. E.
,
Shabanowitz
J.
,
Rosenberg
S. A.
,
Hunt
D. F.
,
Topalian
S. L.
.
1999
.
Biochemical identification of a mutated human melanoma antigen recognized by CD4(+) T cells.
J. Exp. Med.
189
:
757
766
.
117
Wang
R. F.
,
Wang
X.
,
Rosenberg
S. A.
.
1999
.
Identification of a novel major histocompatibility complex class II-restricted tumor antigen resulting from a chromosomal rearrangement recognized by CD4(+) T cells.
J. Exp. Med.
189
:
1659
1668
.
118
Wang
R. F.
,
Wang
X.
,
Atwood
A. C.
,
Topalian
S. L.
,
Rosenberg
S. A.
.
1999
.
Cloning genes encoding MHC class II-restricted antigens: mutated CDC27 as a tumor antigen.
Science
284
:
1351
1354
.
119
Joseph
C. G.
,
Darrah
E.
,
Shah
A. A.
,
Skora
A. D.
,
Casciola-Rosen
L. A.
,
Wigley
F. M.
,
Boin
F.
,
Fava
A.
,
Thoburn
C.
,
Kinde
I.
, et al
.
2014
.
Association of the autoimmune disease scleroderma with an immunologic response to cancer.
Science
343
:
152
157
.
120
Vigneron
N.
,
Stroobant
V.
,
Van den Eynde
B. J.
,
van der Bruggen
P.
.
2013
.
Database of T cell-defined human tumor antigens: the 2013 update.
Cancer Immun.
13
:
15
.
121
Tran
E.
,
Turcotte
S.
,
Gros
A.
,
Robbins
P. F.
,
Lu
Y. C.
,
Dudley
M. E.
,
Wunderlich
J. R.
,
Somerville
R. P.
,
Hogan
K.
,
Hinrichs
C. S.
, et al
.
2014
.
Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer.
Science
344
:
641
645
.
122
Yotnda
P.
,
Firat
H.
,
Garcia-Pons
F.
,
Garcia
Z.
,
Gourru
G.
,
Vernant
J. P.
,
Lemonnier
F. A.
,
Leblond
V.
,
Langlade-Demoyen
P.
.
1998
.
Cytotoxic T cell response against the chimeric p210 BCR-ABL protein in patients with chronic myelogenous leukemia.
J. Clin. Invest.
101
:
2290
2296
.
123
Kissick
H. T.
,
Sanda
M. G.
,
Dunn
L. K.
,
Pellegrini
K. L.
,
On
S. T.
,
Noel
J. K.
,
Arredouani
M. S.
.
2014
.
Androgens alter T-cell immunity by inhibiting T-helper 1 differentiation.
Proc. Natl. Acad. Sci. USA
111
:
9887
9892
.
124
van der Bruggen, P., V. Stroobant, N. Vigneron, and B. Van den Eynde. 2013. Peptide Database: T Cell-Defined Tumor Antigens. Available at: http://www.cancerimmunity.org/peptide/. Accessed: November 2014
.
125
Vogelstein
B.
,
Papadopoulos
N.
,
Velculescu
V. E.
,
Zhou
S.
,
Diaz
L. A.
 Jr.
,
Kinzler
K. W.
.
2013
.
Cancer genome landscapes.
Science
339
:
1546
1558
.
126
Marusyk
A.
,
Almendro
V.
,
Polyak
K.
.
2012
.
Intra-tumour heterogeneity: a looking glass for cancer?
Nat. Rev. Cancer
12
:
323
334
.
127
Schumacher
T.
,
Bunse
L.
,
Pusch
S.
,
Sahm
F.
,
Wiestler
B.
,
Quandt
J.
,
Menn
O.
,
Osswald
M.
,
Oezen
I.
,
Ott
M.
, et al
.
2014
.
A vaccine targeting mutant IDH1 induces antitumour immunity.
Nature
512
:
324
327
.
128
Ding
L.
,
Ellis
M. J.
,
Li
S.
,
Larson
D. E.
,
Chen
K.
,
Wallis
J. W.
,
Harris
C. C.
,
McLellan
M. D.
,
Fulton
R. S.
,
Fulton
L. L.
, et al
.
2010
.
Genome remodelling in a basal-like breast cancer metastasis and xenograft.
Nature
464
:
999
1005
.
129
Gerlinger
M.
,
Rowan
A. J.
,
Horswell
S.
,
Larkin
J.
,
Endesfelder
D.
,
Gronroos
E.
,
Martinez
P.
,
Matthews
N.
,
Stewart
A.
,
Tarpey
P.
, et al
.
2012
.
Intratumor heterogeneity and branched evolution revealed by multiregion sequencing.
N. Engl. J. Med.
366
:
883
892
.
130
Burrell
R. A.
,
McGranahan
N.
,
Bartek
J.
,
Swanton
C.
.
2013
.
The causes and consequences of genetic heterogeneity in cancer evolution.
Nature
501
:
338
345
.
131
Nik-Zainal
S.
,
Alexandrov
L. B.
,
Wedge
D. C.
,
Van Loo
P.
,
Greenman
C. D.
,
Raine
K.
,
Jones
D.
,
Hinton
J.
,
Marshall
J.
,
Stebbings
L. A.
, et al
Breast Cancer Working Group of the International Cancer Genome Consortium
.
2012
.
Mutational processes molding the genomes of 21 breast cancers.
Cell
149
:
979
993
.
132
Zhang
J.
,
Fujimoto
J.
,
Zhang
J.
,
Wedge
D. C.
,
Song
X.
,
Zhang
J.
,
Seth
S.
,
Chow
C. W.
,
Cao
Y.
,
Gumbs
C.
, et al
.
2014
.
Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing.
Science
346
:
256
259
.
133
Sampson
J. H.
,
Heimberger
A. B.
,
Archer
G. E.
,
Aldape
K. D.
,
Friedman
A. H.
,
Friedman
H. S.
,
Gilbert
M. R.
,
Herndon
J. E.
 II
,
McLendon
R. E.
,
Mitchell
D. A.
, et al
.
2010
.
Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma.
J. Clin. Oncol.
28
:
4722
4729
.
134
Lehmann
P. V.
,
Forsthuber
T.
,
Miller
A.
,
Sercarz
E. E.
.
1992
.
Spreading of T-cell autoimmunity to cryptic determinants of an autoantigen.
Nature
358
:
155
157
.
135
Vanderlugt
C. L.
,
Miller
S. D.
.
2002
.
Epitope spreading in immune-mediated diseases: implications for immunotherapy.
Nat. Rev. Immunol.
2
:
85
95
.
136
Kreso
A.
,
O’Brien
C. A.
,
van Galen
P.
,
Gan
O. I.
,
Notta
F.
,
Brown
A. M.
,
Ng
K.
,
Ma
J.
,
Wienholds
E.
,
Dunant
C.
, et al
.
2013
.
Variable clonal repopulation dynamics influence chemotherapy response in colorectal cancer.
Science
339
:
543
548
.
137
Marusyk
A.
,
Tabassum
D. P.
,
Altrock
P. M.
,
Almendro
V.
,
Michor
F.
,
Polyak
K.
.
2014
.
Non-cell-autonomous driving of tumour growth supports sub-clonal heterogeneity.
Nature
514
:
54
58
.
138
van Galen
P.
,
Kreso
A.
,
Mbong
N.
,
Kent
D. G.
,
Fitzmaurice
T.
,
Chambers
J. E.
,
Xie
S.
,
Laurenti
E.
,
Hermans
K.
,
Eppert
K.
, et al
.
2014
.
The unfolded protein response governs integrity of the haematopoietic stem-cell pool during stress.
Nature
510
:
268
272
.
139
Schroers
R.
,
Shen
L.
,
Rollins
L.
,
Rooney
C. M.
,
Slawin
K.
,
Sonderstrup
G.
,
Huang
X. F.
,
Chen
S. Y.
.
2003
.
Human telomerase reverse transcriptase-specific T-helper responses induced by promiscuous major histocompatibility complex class II-restricted epitopes.
Clin. Cancer Res.
9
:
4743
4755
.
140
Brunsvig
P. F.
,
Kyte
J. A.
,
Kersten
C.
,
Sundstrøm
S.
,
Møller
M.
,
Nyakas
M.
,
Hansen
G. L.
,
Gaudernack
G.
,
Aamdal
S.
.
2011
.
Telomerase peptide vaccination in NSCLC: a phase II trial in stage III patients vaccinated after chemoradiotherapy and an 8-year update on a phase I/II trial.
Clin. Cancer Res.
17
:
6847
6857
.
141
Dosset
M.
,
Godet
Y.
,
Vauchy
C.
,
Beziaud
L.
,
Lone
Y. C.
,
Sedlik
C.
,
Liard
C.
,
Levionnois
E.
,
Clerc
B.
,
Sandoval
F.
, et al
.
2012
.
Universal cancer peptide-based therapeutic vaccine breaks tolerance against telomerase and eradicates established tumor.
Clin. Cancer Res.
18
:
6284
6295
.
142
Widenmeyer
M.
,
Griesemann
H.
,
Stevanović
S.
,
Feyerabend
S.
,
Klein
R.
,
Attig
S.
,
Hennenlotter
J.
,
Wernet
D.
,
Kuprash
D. V.
,
Sazykin
A. Y.
, et al
.
2012
.
Promiscuous survivin peptide induces robust CD4+ T-cell responses in the majority of vaccinated cancer patients.
Int. J. Cancer
131
:
140
149
.
143
Kimura
T.
,
McKolanis
J. R.
,
Dzubinski
L. A.
,
Islam
K.
,
Potter
D. M.
,
Salazar
A. M.
,
Schoen
R. E.
,
Finn
O. J.
.
2013
.
MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility study.
Cancer Prev. Res. (Phila.)
6
:
18
26
.
144
Wierecky
J.
,
Müller
M. R.
,
Wirths
S.
,
Halder-Oehler
E.
,
Dörfel
D.
,
Schmidt
S. M.
,
Häntschel
M.
,
Brugger
W.
,
Schröder
S.
,
Horger
M. S.
, et al
.
2006
.
Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients.
Cancer Res.
66
:
5910
5918
.
145
Nishikawa
H.
,
Sato
E.
,
Briones
G.
,
Chen
L. M.
,
Matsuo
M.
,
Nagata
Y.
,
Ritter
G.
,
Jäger
E.
,
Nomura
H.
,
Kondo
S.
, et al
.
2006
.
In vivo antigen delivery by a Salmonella typhimurium type III secretion system for therapeutic cancer vaccines.
J. Clin. Invest.
116
:
1946
1954
.
146
Hunder
N. N.
,
Wallen
H.
,
Cao
J.
,
Hendricks
D. W.
,
Reilly
J. Z.
,
Rodmyre
R.
,
Jungbluth
A.
,
Gnjatic
S.
,
Thompson
J. A.
,
Yee
C.
.
2008
.
Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1.
N. Engl. J. Med.
358
:
2698
2703
.
147
Carmichael
M. G.
,
Benavides
L. C.
,
Holmes
J. P.
,
Gates
J. D.
,
Mittendorf
E. A.
,
Ponniah
S.
,
Peoples
G. E.
.
2010
.
Results of the first phase 1 clinical trial of the HER-2/neu peptide (GP2) vaccine in disease-free breast cancer patients: United States Military Cancer Institute Clinical Trials Group Study I-04.
Cancer
116
:
292
301
.
148
Friedman
K. M.
,
Prieto
P. A.
,
Devillier
L. E.
,
Gross
C. A.
,
Yang
J. C.
,
Wunderlich
J. R.
,
Rosenberg
S. A.
,
Dudley
M. E.
.
2012
.
Tumor-specific CD4+ melanoma tumor-infiltrating lymphocytes.
J. Immunother.
35
:
400
408
.
149
Inderberg-Suso
E. M.
,
Trachsel
S.
,
Lislerud
K.
,
Rasmussen
A. M.
,
Gaudernack
G.
.
2012
.
Widespread CD4+ T-cell reactivity to novel hTERT epitopes following vaccination of cancer patients with a single hTERT peptide GV1001.
OncoImmunology
1
:
670
686
.
150
McKinney
D. M.
,
Southwood
S.
,
Hinz
D.
,
Oseroff
C.
,
Arlehamn
C. S.
,
Schulten
V.
,
Taplitz
R.
,
Broide
D.
,
Hanekom
W. A.
,
Scriba
T. J.
, et al
.
2013
.
A strategy to determine HLA class II restriction broadly covering the DR, DP, and DQ allelic variants most commonly expressed in the general population.
Immunogenetics
65
:
357
370
.
151
Greenbaum
J.
,
Sidney
J.
,
Chung
J.
,
Brander
C.
,
Peters
B.
,
Sette
A.
.
2011
.
Functional classification of class II human leukocyte antigen (HLA) molecules reveals seven different supertypes and a surprising degree of repertoire sharing across supertypes.
Immunogenetics
63
:
325
335
.
152
Bos
R.
,
Sherman
L. A.
.
2010
.
CD4+ T-cell help in the tumor milieu is required for recruitment and cytolytic function of CD8+ T lymphocytes.
Cancer Res.
70
:
8368
8377
.
153
Pardoll
D. M.
1999
.
Inducing autoimmune disease to treat cancer.
Proc. Natl. Acad. Sci. USA
96
:
5340
5342
.
154
Dudley
M. E.
,
Wunderlich
J. R.
,
Robbins
P. F.
,
Yang
J. C.
,
Hwu
P.
,
Schwartzentruber
D. J.
,
Topalian
S. L.
,
Sherry
R.
,
Restifo
N. P.
,
Hubicki
A. M.
, et al
.
2002
.
Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes.
Science
298
:
850
854
.
155
U.S. Food and Drug Administration. Paving the Way for Personalized Medicine: FDA’s Role in a New Era of Medical Product Development. Available at: http://www.fda.gov/downloads/ScienceResearch/SpecialTopics/PersonalizedMedicine/UCM372421.pdf. Accessed: November 2014
.

The author has no financial conflicts of interest.