Open access

Adoptive Cell Therapy of Melanoma: The Challenges of Targeting the Beating Heart

Written By

Jennifer Makalowski and Hinrich Abken

Submitted: 19 April 2012 Published: 30 January 2013

DOI: 10.5772/53619

From the Edited Volume

Melanoma - From Early Detection to Treatment

Edited by Guy Huynh Thien Duc

Chapter metrics overview

2,524 Chapter Downloads

View Full Metrics

1. Introduction

The identification of melanoma-associated antigens, the isolation of tumor infiltrating T cells from melanoma lesions, and the significant progress in engineering redirected T cells has favored the development of various strategies in the adoptive immunotherapy of melanoma. Recent trials in adoptive cell therapy (ACT) have achieved spectacular results in inducing remission in advanced stages of the disease, although produced on-target off-tumor toxicities, emphasizing the tremendous potential benefit of harnessing the immune system for fighting the disease. Moreover, the identification of so-called melanoma stem cells along with strategies for selectively eliminating subsets of melanoma cells implies that there is a need for redefining therapeutic targets in melanoma. This review discusses current challenges in the rational design of adoptive cell therapy to target “the beating heart” of melanoma.

1.1. Advanced stages of melanoma resist conventional therapeutic regimens

Surgical resection of tumor lesions in early stages of the disease is the curative option for combating melanoma; a 10-year-survival rate of 75 - 85% can be achieved for melanoma in stage I or II. However, melanoma in stage III or IV is still associated with low survival rates of less than 1 year upon diagnosis [1]. Despite the development of novel drugs and major improvements in therapeutic regimens, significant responses were only achieved in predefined groups and of short duration. Treatment with the chemotherapeutic drug dacarbazine (DTIC) and vemurafenib, an inhibitor of mutated BRAF, produced a median progression-free survival of 64% with dacarbazine, respectively 84% with vemurafenib of approximately 6 months [2-4]. The biology of melanoma and the heterogeneity of malignant cells are thought to be responsible for this unsatisfactory situation. First, melanoma cells can persist for long periods of time in a “dormant” stage without any progression in tumor formation [5]. Second, melanoma cells can disseminate early into distant organs including the brain forming micro-metastases, which are small in cell numbers and frequently beyond the detection limit of current imaging procedures [6, 7]. Third, many melanoma cells are notoriously resistant to chemo- and radiation therapy [8-10], making alternative strategies in tumor cell elimination necessary.

Therefore, in more progressed stages of the disease the recruitment of the cellular immune defense to eliminate cancer cells is thought to be an alternative. Administration of high dose interleukin-2 (IL-2) [11] and anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) antibody [12] as well as interferon (IFN) α-2b prolongs the disease-free survival although at a relatively low response rate and without being curative over time [13, 14]. However, these and other observations imply that activation or modulation of the patient’s immune response may be effective in the treatment of melanoma. A number of approaches for enhancing the immune cell response against melanoma are currently explored with some success. In particular, the adoptive transfer of autologous T cells isolated from melanoma lesions and expanded to large numbers ex vivo has produced encouraging phase II results [15, 16]. The administration of patient’s blood T cells engineered with defined specificity for melanoma-associated antigens are additionally being explored in a number of trials. In this review, we summarize evidence for the potency of adoptive T cell therapy in the treatment of melanoma and discuss current challenges in achieving long-term remission. Upcoming strategies in selective targeting cancer stem cells are also discussed.

Advertisement

2. Adoptive cell therapy can successfully fight melanoma

Melanoma can trigger a curative immune response; this conclusion is drawn from the clinical observation of spontaneous and complete melanoma regressions and of the higher frequency of melanomas among immune compromised patients [17, 18]. More direct evidence for the immune cell control of melanoma growth was obtained by the treatment with high dose IL-2, which produces an objective response rate of 16%. Indeed, some of the patients receiving thus treatment exhibit a long-term complete response for years [11, 19]. These observations are remarkable in light of the low and short-lived response rates after chemotherapy and currently drive the development of adoptive T cell therapy for treatment of late stage melanoma.

The development of adoptive cell therapy (ACT) was further strengthened by upcoming technologies in isolating tumor infiltrating lymphocytes (TIL's) from melanoma biopsies (Figure 1). First described in 1969 [20], TIL's from melanoma lesions consisted of both effector and helper T cell subsets and can be expanded ex vivo in the presence of IL-2. The expanded cells are then selected for melanoma reactivity. A strong rationale for using these T cells in adoptive therapy is provided by the observation that the infusion of high TIL numbers correlates with better clinical outcome [21, 22] although the prevalence of TIL's in primary melanoma lesions and metastases is not a prognostic factor itself.

Protocols according to GMP standards have been established in several centers to isolate and amplify TIL's to numbers appropriate for adoptive therapy. Melanoma reactive T cells are expanded in the presence of IL-2 by culture on feeder cells expressing melanoma antigens [23]. Subsequent to TIL re-infusions, metastases regressed in the majority of patients and a stable disease phase followed. However, only few patients remained in complete remission [21]. The disappointing therapeutic efficacy, despite high numbers of infused TIL's is thought to be due to low responsiveness of highly expanded T cells which are unable to execute a productive anti-melanoma attack after administration to the patient. Current TIL protocols therefore attempt to administer so-called “young TIL's” (Figure 1), i.e. melanoma infiltrating T cells which underwent short-term culture expansions and therefore passed through fewer cell division cycles prior to re-infusion and thereby exhibit a less differentiated phenotype [24]. Another change in protocols is that TIL's are not selected for melanoma reactivity; the rationale behind this is that re-infusion of ex vivo IFN-γ secreting TIL's exhibited no major benefit compared to non-responding TIL's [16]. Early phase I trials showed improved persistence of young TIL's [25] and 50% response rates in a cohort of 20 patients [26], which is just as effective as traditionally grown TIL's [27]. Different non-randomized phase II trials at the NCI and at Sheba Medical Center confirmed these early observations (Table 1) [28, 29]. A roadmap describing critical steps for comparative testing the TIL strategy in a randomized multi-center setting was recently published in a White Paper on adoptive cell therapy [30].

Figure 1.

Adoptive cell therapy for metastatic melanoma. Adoptive cell therapy with tumor infiltrating lymphocytes (TIL´s) makes use of melanoma-specific TIL´s which are isolated from a melanoma biopsy, amplified ex vivo by stimulation with melanoma biopsy cells and propagated to high numbers in the presence of IL-2. In more recent trials, TIL´s are propagated short-term ex vivo without stimulation by melanoma cells and administered as "young" TIL´s.

Target antigen Adoptively transferred T cells NCT ID / Reference Center
melanoma specific CD8+ T cells [118] FHCRC
melanoma specific T cells [119] LUMC
MART-1 MART-1 specific CD8+ T cells [113] DFCI
MART-1 MART-1 specific CD8+ T cells NCT00512889 DFCI
MART-1 MART-1 specific CD8+ T cells [87] UR
MART-1 MART-1 specific CD8+ T cells [33] UNH
MART-1 MART-1 specific CD8+ T cells NCT00324623 CHUV
MART-1 MART-1 specific CD8+ T cells NCT01106235 FHCRC
NY-ESO-1 NY-ESO-1 specific CD8+ T cells and anti-CTLA-4 antibody NCT00871481 FHCRC
TILs [114] NIH
TIL [120] NIH
TILs [27] NIH
TILs [29] NIH
TILs [115] NIH
TILs NCT00287131 SMC
TILs NCT000604136 HMO
TILs NCT01005745 MOFFITT
TILs and IFN-γ NCT01082887 NUH
“young“ TILs [116] NIH
“young“ TILs [28] SMC
“young“ TILs NCT01118091 NIH
“young“ TILs NCT01319565 NIH
“young“ TILs NCT01369888 MIH
“young“ TILs NCT01468818 NIH
“young“ TILs NCT00513604 NIH
MART-1 MART-1 specific TILs NCT00720031 NUH
MART-1 MART-1 specific TILs (DMF5) NCT00924001 CC
IL-2 engineered TILs [117] NIH
IL-2 engineered TIL NCT00062036 NIH
IL-12 engineered TIL NCT01236573 NIH
CXCR2 engineered TIL [86] MDACC
NY-ESO-1 anti-NY-ESO-1 TCR [121] NIH
NY-ESO-1 anti-NY-ESO-1 TCR NCT00670748 NIH
MART-1 anti-MART-1 TCR (low-affinity) [49] NIH
MART-1 anti-MART-1 TCR NCT00910650 UC
MART-1 anti-MART-1 TCR (high-affinity) [38] NIH
gp-100 anti-gp-100 TCR [38] NIH
MART-1 anti-MART-1 TCR [114] NIH
gp-100 anti-gp-100 TCR [114] NIH
MART-1 anti-MART-1 TCR NCT00612222 NIH
gp-100 anti-gp-100 TCR NCT00610311 NIH
MART-1 anti-MART-1 TCR plus MART-1 vaccination NCT00923195 NIH
gp-100 anti-gp-100 TCR plus gp-100 vaccination NCT00923195 NIH
p53 anti-p53 TCR NCT00393029 NIH
VEGFR2 anti-VEGFR2 CAR engineered CD8+ T cells NCT01218867 NIH
Ganglioside GD-3 anti-GD-3 CAR PI: M. Davies MDACC

Table 1.

Adoptive cell therapy trials in patients with metastatic melanoma

CHUV, Centre Hospitalier Universitaire Vaudois; DFCI, Dana-Farber Cancer Institute; FHCRC, Fred Hutchinson Cancer Research Center; HMO, Hadassah Medical Organization; LUMC, Leiden University Medical Center; MDACC, M.D. Anderson Cancer Center; MOFFITT, H. Lee Moffitt Cancer Center and Research Institute; NIH, National Institutes of Health; NUH, Nantes University Hospital; PI, principal investigator; SMC, Sheba Medical Center; UC, University of California; UR, University of Regensburg


Advertisement

3. Adoptive cell therapy with antigen-specific T cells

The rationale for using melanoma antigen-specific T cells is based on the observation that the success of TIL therapy in some patients correlates with the presence of melanoma-reactive T cells, in particular with those cells specific for Melan-A, MART-1 or gp100 [23, 31]. The median survival of patients treated with Melan-A specific TIL's was 53.5 months compared to 3.5 months for patients who received TIL's without Melan-A specificity [32]. These observations together with a number of technical obstacles in obtaining TIL's from biopsies strengthened efforts to derive melanoma-specific T cell clones from peripheral blood lymphocytes for the use in adoptive cell therapy. The strategy was corroborated by a 50% response rate obtained after transfer of MART-1 or gp100 specific T cell clones isolated and propagated ex vivo from peripheral blood lymphocytes (Table 1) [33]. Melanoma reactive T cell clones in peripheral blood are rare, TIL therapy increases the otherwise low magnitude of the tumor-reactive T cell compartment in vivo, which matches the reactivity in the TIL product [34]. Interestingly, individual TIL products from different patients contain unique patterns of reactivity against shared melanoma-associated antigens [34]. TIL isolation and expansion in vitro, however, is extremely laborious. This limit leads to attempts to engineer patient's blood T cells with pre-defined specificity for more specifically redirecting the cytotoxic response toward melanoma. It is therefore assumed that the clinical efficacy of TIL therapy can be improved by application of T cells with more defined tumor-reactivity.

To engineer specificity for melanoma, T cell receptors (TCR's) were cloned from TIL's of responding melanoma patients and transferred to peripheral blood T cells of the same patient (Figure 2) [35-38]. The gp100 specific TCR was one of the first TCR’s, cloned from melanoma TIL's and introduced ex vivo by retrovirus-mediated gene transfer into blood T cells, which thus obtained redirected specificity for gp100 positive cells. In contrast to their non-modified counterparts, TCR engineered T cells responded to gp100+ melanoma cells by secreting pro-inflammatory cytokines including IFN-γ and by lysing the target cells [45, 46]. Similarly, blood T cells were engineered with recombinant TCR’s with specificity for MART-1 or MAGE-A1. The functional avidity of cloned TCR's was improved and engineered T cells were successfully used in subsequent trials [47, 48]. About 30% of patients receiving ACT with MART-1 specific T cells responded with melanoma regression; 19% of patients treated with gp100 specific TCR T cells exhibited objective response, most responses were persistent [38]. TCR engineered T cells also showed efficacy towards brain metastases, which indicates that patients with otherwise incurable metastatic sites may benefit from ACT (Table 1) [115]. In patients with prolonged clinical remission, engineered T cells were present in the circulation for more than a year after initiation of treatment; this indicates that therapeutic efficacy and long-term anti-melanoma immunity may correlate with T cell persistence [49, 50].

Figure 2.

Adoptive cell therapy with redirected T cells. T cells from the peripheral blood of the patient are engineered ex vivo by retro- or lentiviral gene transfer with cDNA coding for a T cell receptor (TCR) with specificity for a melanoma-associated antigen. Alternatively, T cells are engineered with a chimeric antigen receptor (CAR) which recognizes a melanoma-associated antigen by an antibody-derived binding domain. Engineered T cells are expanded ex vivo prior to administration to the patient.

However, the enthusiasm for adoptive cell therapy with TCR modified T cells has been dampened by several limitations. Tumor cells including those of the melanoma undergo clonal evolution, and some of these evolved cells evade T cell recognition, for instance, as a result of repression of their MHC complex [51], of mutations in their β2 microglobulin chain [52], and of deficiencies in their antigen processing machinery [51, 53]. Each of these alterations renders the melanoma cell invisible to a TCR-mediated T cell attack. A possible safety hazard moreover became apparent when analyzing in more detail the transgenic TCR, which is co-expressed with the physiological TCR in the same T cell. The transgenic TCR turned out to create new but unpredictable specificities by forming hetero-dimers of the recombinant α and β TCR chains with the respective chains of the physiological TCR. Undesirable mispairing of TCR chains may result in loss of specificity and may induce severe auto-reactivity [54, 55]. Tremendous efforts were subsequently made to solve the problem including replacement of TCR constant moieties by the homologous murine domains [56] and creation of additional cysteine bridges [57] to enforce preferential pairing of the recombinant αβ TCR chains in the presence of the physiological TCR.

These and other technical difficulties promoted the development of an artificial “one-chain-receptor” molecule to redirect T cells in an antigen-restricted manner (Figure 3). In a seminal paper Zelig Eshhar of the Weizmann Institute of Science described a chimeric antigen receptor (CAR), also named immunoreceptor, which is composed in the extracellular part of a single chain antibody for antigen binding and in the intracellular part of the TCR/CD3ζ endodomain for provision of T cell activation [58]. The CAR modified T cell, also known as “T-body”, becomes activated by binding to antigen, and secretes pro-inflammatory cytokines, amplifies and lyses target cells expressing the respective antigen. By using an antibody for binding, the CAR recognizes the target in a MHC-independent fashion and is therefore not affected by loss of HLA molecules, which frequently occurs during neoplastic progression. An additional advantage over transgenic TCR's is that CAR's can be used independently of the individual HLA subtype. However, the T-body strategy is restricted to antigens expressed on the surface of the target cell; intracellular antigens are not visible to CAR's. Due to the broad variety of antibodies available, a nearly unlimited panel of antigens can be targeted with high affinity and specificity, including those which are not classical T cell antigens, e.g. carbohydrates. High affinity CAR's activate engineered T cells even after binding to low amounts of target antigen; this not only makes the approach highly sensitive, but also makes the choice of the appropriate melanoma-selective antigen difficult.

Figure 3.

Recombinant receptors to redirect T cells for use in antigen-specific cell therapy. The physiologic T cell receptor (TCR)/CD3 complex consists of the α and β TCR chains, which recognize major histocompatibility complex (MHC)-presented antigen by binding through both variable regions Vα Vβ, and of the CD3 chains. Antigen engagement induces clustering of the TCR complex and the primary signal for T cell activation is generated by the intracellular CD3ζ chain. Recombinant TCR α and β chains can be engineered to T cells in order to provide a new specificity. Alternatively, the V regions of the TCR chains can be combined and fused to the intracellular CD3ζ chain to produce a T cell activation signal upon binding to antigen. The chimeric antigen receptor (CAR) makes use of an antibody binding domain for antigen recognition which is enigneered by fusing the variable (V) regions of the immunoglobulin heavy (H) and light (L) chain. The VH-VL single chain antibody is linked via a spacer to the intracellular CD3ζ chain to produce the primary T cell activation signal upon antigen binding. Intracellular signaling domains of costimulatory molecules like CD28 can be added to provide appropriate costimulation in addition to the primary CD3ζ signal.

T cells require two signals for full and lasting activation, one provided by the TCR and the other by costimulatory co-receptors; the prototype of which is CD28. The corresponding ligands are usually not present in the tumor micro-environment. Some effector functions including IL-2 secretion require CD28 costimulation along with the primary TCR/CD3ζ signal; this provides a rationale for combining the intracellular CD3ζ with the CD28 signaling domain in one polypeptide chain (Figure 3) [59]. Other costimulatory domains, such as 4-1BB (CD137) and OX40 (CD134), were also linked to CD3ζ; each domain has a different impact on T cell effector functions [60]. Costimulatory domains were furthermore combined in so-called 3rd generation CAR's, and a number of additional modifications have been introduced in the last years to improve T cell persistence and activation [61, 62]. CAR's with a costimulatory domain clearly demonstrated clinical benefit and improved T cell persistence compared to CAR's targeting the same antigen but with only the CD3ζ domain [63-65].

Various CARs were engineered for targeting melanoma-associated antigens, including HMW-MAA, also known as MCSP [67, 68], melanotransferrin [69], the ganglioside GD2 [70] and GD3 [71]. A clinical trial targeting melanoma cells with CAR engineered T cells is currently recruiting participants [66]. Recent phase I trials using CAR redirected T cells in the treatment of lymphoma/leukemia exhibited spectacular efficacy [72, 73]. However, the enthusiasm was dampened by reports on serious adverse events and even fatalities after CAR T cell therapy [74, 75]. Targeting ErbB2 produced a cytokine storm and respiratory failure in one case [76] which is thought to be due to low levels of antigen on a number of healthy cells which can trigger CAR T cell activation. On the one hand, this event points out that ACT with CAR modified T cells may be a powerful therapy; but, on the other hand, emphasizes the necessity for careful T cell dose escalation studies to balance anti-tumor efficacy and auto-immunity[61, 77, 78].

Advertisement

4. Challenges and premises in the adoptive cell therapy of melanoma

To date, approximately half of the melanoma patients treated with TIL ACT benefit from this therapy; genetic modification of T cells may further improve clinical response to melanoma, but this will have to be proven in upcoming trials. However, the strategy has potential challenges which need to be addressed.

A major challenge of redirected T cells is the tumor selectivity for the target antigen itself, which in most cases is not exclusively expressed on tumor cells but also on healthy cells [79], although almost always at lower levels: for instance MART-1, which is also expressed by melanocytes. When targeting these antigens, vitiligo and inner ear toxicity resulting in a certain degree of deafness are frequently observed side effects [38]. From this perspective it is reasonable to assume that off-target toxicities may be adverse reactions for clinical efficacy in an anti-melanoma response [80]. Since nearly all tumor-associated antigens are self-antigens, strategies will have to be developed to ensure that off-target toxicities are kept to a minimum. Whether T cells with low-avidity TCR or CAR are less prone to induce such undesirable side effects is currently under investigation.

Melanoma cells, like other cancer cells, down-regulate components of the MHC and become increasingly deficient in antigen processing. As a consequence, TCR engineered T cells can no longer bind to and destroy those melanoma cells. However, they may be visible to a CAR recognizing surface antigens in a MHC independent manner, because of the antibody-derived binding domain (Figure 3). TCR redirected T cells, on the one hand, may also recognize cross-presented targeted antigen, for instance by stroma cells, but this is not the case for CAR engineered T cells. Cross-presented antigen, on the other hand, may help to destroy stroma, which is required to eliminate large tumor lesions [39, 40].

To avoid mispairing of the recombinant TCR with the physiological TCR chains and the resulting unpredictable auto-immunity, TCR-like single chain antibodies were used as targeting domain in a CAR. Thus combining the MHC-restricted recognition of antigen with the T-body strategy. T cells with TCR-like CAR were redirected towards NY-ESO-1 and MAGE-A1, respectively [41, 42]. The possible advantages of these MHC restricted CAR's compared to the use of recombinant TCR's still has to be determined in trials.

The antibody-derived binding domain of a CAR displays extraordinary high affinity compared to a TCR. However, an increase in affinity, for instance, by affinity maturation, does not necessarily improve CAR redirected T cell activation above threshold [41, 43], which is not additionally modulated by CD28 costimulation [44]. A similar effect is also assumed for TCR mediated T cell activation. The TCR or CAR binding avidity probably affects the persistence of engineered T cells at the targeted tumor site. Strong binding to a target antigen may cause the T cells to be trapped and to become fully activated for a cytolytic attack, whereas low avidity interactions may not provide sufficiently long T cell – melanoma cell contacts. In addition to the binding avidity, the amount of target antigen on the cell surface also impacts on the selectivity of redirected T cell activation. In essence, low affinity binding directs the activity of engineered T cells preferentially toward target cells with abundant antigen levels; high affinity binding is likewise effective against low antigen levels on target cells. The optimized affinity to sustain a more selective T cell trafficking to the tumor and activation while avoiding targeting healthy cells that are expressing low quantities of the same antigen, however, still has to be determined.

A beneficial T cell-to-target cell ratio at the tumor site seems to be required for efficient tumor elimination. Higher numbers of engineered T cells applied per dose will probably increase clinical efficacy; the majority of recent trials have applied up to 1010 cells per dose [27]. These and higher numbers of engineered T cells can be generated by extended expansion protocols; however, cells with a "young" phenotype may not be generated for adoptive transfer under these conditions. Short-term amplification protocols are therefore envisioned for both TIL's and engineered blood T cells. However, the majority of recent trials targeting CD19+ leukemia provided evidence for therapeutic efficacy at numbers less than or equal to 105 engineered T cells [73]. This once again raises the question of whether high T cell doses are required for a therapeutic effect.

The clinical outcome of adoptive cell therapy correlates with the persistence of adoptively transferred T cells [81]. As long as T cells engage their cognate antigen, T cells will expand and persist in detectable numbers; but when the antigen is no longer present, the T cell population will contract to potentially undetectable levels and disappear from circulation. To improve survival of CAR T cells, Epstein-Barr virus (EBV)-specific T cells were engineered with a tumor-specific CAR based on the rationale that T cells recognizing the low amounts of EBV antigens by their physiological TCR will be maintained in a sizable population in circulation and in the process providing enough CAR T cells to recognize and kill melanoma cells in the surrounding tissues. A clinical trial with EBV-specific T cells engineered with an anti-GD2 CAR thus showed benefit over non-virus-specific, CAR engineered T cells in the treatment of neuroblastoma [81].

Adoptively transferred CD8+ T cell clones may be less persistent than CD4+ T cell clones due to T cell exhaustion after extensive ex vivo amplification and multiple rounds of activation. In addition, CD4+ T cell help is essential for CD8+ T cell persistence in vivo; adoptively transferred pure CD8+ T cell clones may fail to persist [82]. T cell therapy may be combined with antibody therapy to prolong the initiated immune response. For instance, CTLA-4 is upregulated on the surface of activated T cells, where it acts as negative regulator to return the T cell to a resting stage. Co-application of the anti-CTLA-4 blocking antibody, ipilimumab, may prolong the anti-tumor activation of transferred T cells, although it would also affect all the other T cells.

Besides maintaining a high number of T cells in circulation, another challenge is to accumulate significant numbers of effector T cells in the tumor lesion. A tightly controlled network of chemokines controls the migration of cells in the body; adoptively transferred T cells use these networks to accumulate at the tumor site. The expression of specific chemokine receptors controls how cells will migrate against the chemokine gradient into the targeted lesion. Melanoma cells secrete a number of chemokines including CXCL1. However, early imaging studies revealed that melanoma-specific T cells massively infiltrate the lungs, spleen and liver with some accumulation at the tumor site, which clearly represents a minority of the transferred cells, before the cells decline to undetectable levels in circulation [83-85]. Since those T cells do not express CXCR2, the receptor for melanoma secreted CXCL1, TIL's were engineered with CXCR2 which generated improved melanoma accumulation and anti-tumor activity in a mouse model [86]. The strategy is currently being explored in an early phase I trial (Table 1) [86].

One of the major hurdles of redirected immunotherapy of cancer in general is the tremendous heterogeneity of cancer cells with respect to the expression of the targeted antigen. Low or lack of antigen expression within the malignant lesions will negatively affect the long-term therapeutic efficacy of the approach. Several reports document relapse of antigen-loss tumor metastases after adoptive therapy with melanoma-reactive T cell clones [87-89] and argue for the use of polyclonal T cells with various melanoma specificities. Melanoma cells expressing the target antigen may successfully be eliminated by redirected T cells, whereas antigen-negative tumor cells will not be recognized. T cell populations modified with different CAR's recognizing different antigens expressed by the same tumor may be able to overcome these limitations. However, pro-inflammatory cytokines secreted by redirected T cells into the tumor micro-environment upon activation may attract a second wave of non-antigen restricted effector cells, which in turn may eradiate antigen-negative tumor cells. At least in an animal model, antigen-negative melanoma cells are indeed eliminated when co-inoculated with antibody-targeted cytokines [90]. Moreover, T cells engineered with induced expression of transgenic IL-12 attract innate immune cells including macrophages into the tumor tissue; they eliminate antigen-negative tumor cells in the same lesion [91].

Highly expanded T cells, such as TIL's, become hypo-responsive to CD28 costimulation and rapidly enter activation induced cell death, in particular upon IL-2 driven expansion [92]. This may be counteracted by expansion in the presence of IL-15 and IL-21 and/or by co-stimulation via 4-1BB by an agonistic antibody [93].

Metastatic melanoma patients with the B-raf activating mutation V600E transiently benefit from a small molecule drug, PLX4032 or vemurafenib, which inhibits the mitogen-activated protein kinase (MAPK) pathway. Treatment with vemurafenib is accompanied by increased T cell infiltrations in the melanoma lesions [94, 95]. Combination of B-raf inhibition with melanoma-specific ACT may provide an option to prolong the clinical response.

Although the TCR downstream signaling machinery is used by the prototype CAR, monocytes, macrophages as well as NK cells can also be redirected by CAR's in an antigen-specific fashion [96, 97]. Whether redirected non-T cells are advantageous in tumor elimination to cancer patients in general and to melanoma patients in particular has to be explored in clinical trials.

Advertisement

5. Does targeting "melanoma stem cells" provide hope for long-term remission from melanoma?

Observations that a number of malignant lesions display a tremendous cellular and phenotypic heterogeneity and contain pluripotent stem cells led to the hypothesis that cancer is initiated and maintained by so-called cancer stem cells (CSC’s). Low abundance, induction of tumors upon transplantation under limiting conditions, radiation and chemo-resistance, self-renewal and a-symmetric differentiation into a variety of cell types are properties postulated for CSC’s. The concept was sustained by deciphering the hierarchical organization in hematological malignancies [98], and subsequently in solid cancers including mammary, prostate, pancreatic, colon carcinoma and glioma [99-103]. Transplantation of melanoma cell subsets under limiting dilution conditions showed that a subset of cancer cells can induce tumors of the same histological phenotype as the parental tumor [99, 104, 105]. A first study using the limiting dilution transplantation assay identified a melanoma cell subset which exhibits stem-like capacities and expresses CD20 [106]. A conclusion drawn from these and other experiments was that melanoma is organized in a hierarchical manner originating from an initiator cell. In this context, several phenomena in melanoma biology which have been clinically observed but not well understood are described by the CSC model, for instance, metastatic relapse more than a decade after surgical treatment of the primary lesion. Residual CSC’s are thought to drive cancer relapse even after years of “dormancy” [107]. Moreover, melanoma initiating cells were identified as expressing either the transporter protein ABCB5 [104] or the nerve growth factor receptor CD271; the latter occurs in melanoma in a frequency of approximately 1/2000 cells [108].

However, transplantation under more rigorous conditions, i.e., ideally of one isolated melanoma cell, revealed that nearly every fourth randomly taken melanoma cell (1/2 - 1/15) can induce tumors and raising the question of the validity the stem cell paradigm for melanoma [109, 110]. From these and subsequent studies, it has been concluded that the potential of melanoma induction is not closely associated with a particular phenotype and that the number of potential CSC’s in melanoma may not necessarily be low. This resulted in a further conclusion that nearly every melanoma cell is capable to re-program to a tumor initiating cell under certain experimental conditions of xeno-transplantation irrespectively which particular marker phenotype the cell expressed at the time of isolation from a melanoma lesion.

Once the tumor is established, a minor subset seems to take over control of melanoma progression. Evidence is provided by recent observations from a pre-clinical model [69], which addressed the question of whether specific elimination of defined melanoma cells from an established xeno-transplanted lesion causes tumor regression by adoptive transfer of antigen-specific cytotoxic T cell. The rationale is that, if there is a clearly defined hierarchy of cancer cells in an established tumor, specific ablation of the melanoma sustaining cells from the established tumor tissue must inevitably lead to a decay of the tumor lesion independently of targeting the cancer cell mass. However, the melanoma sustaining cell may, but must not, be identical to CSC’s identified by the transplantation assay. Targeted elimination of a minor subset of CD20+ melanoma cells completely eradicated transplanted melanoma lesions, whereas targeted elimination of any random melanoma cell population in the same lesion did not. CD20+ melanoma cells are rare, i.e. approximately 1-2%, in melanoma, independently of the histological type and the transplanted tumor tissue. A caveat is that in approximately 20% of melanoma samples, no CD20+ melanoma cells could be detected by histological screening. When these tumors were transplanted, adoptive transfer of CD20-specific CAR T cells did not induce tumor regression. Interestingly, CD20 re-expression in a random subpopulation of those tumor cells did not render the tumor lesion sensitive for complete eradication with CD20-specific T cells. This indicates that CD20 expression per se is not dominant in maintaining melanoma progression. However, the phenotype of CD20+ melanoma cells may be flexible and associated with additional capabilities which mediate the dominant effect.

The first clinical evidence confirming this concept was recently provided by a case report [111]. A patient with stage III/IV metastatic melanoma, which harbored CD20+ melanoma cells at a frequency of 2%, received intra-lesional injections of the anti-CD20 therapeutic antibody rituximab and concomitant dacarbazine treatment. Dacarbazine as mono-therapy had already proved to be ineffective. This treatment produced lasting complete and partial remission accompanied by a decline of the melanoma serum marker S-100 to physiological levels, a switch of a T helper-2 to a more pro-inflammatory T helper-1 response, all without treatment related grade 3/4 toxicity. Although anecdotic, this data provides the first clinical evidence that targeting the subset of CD20+ melanoma sustaining cells can produce regression of chemotherapy-refractory melanoma. Moreover, the report highlights the potency of selective cancer cell targeting in the treatment of melanoma.

These observations although so far based on a pre-clinical model and a clinical observation which will have to be reproduced in larger cohorts have major impact on the future development of melanoma therapy.

First, the melanoma maintaining cells may be more resistant to current therapy regimens than the bulk of melanoma cells. Standard therapy strategies attempt to eliminated all cancer cells in a tumor lesion; elimination of any other cancer cells than the tumor progressing cells will rapidly de-bulk the tumor lesion. The melanoma will inevitably relapse, driven by the remaining melanoma sustaining cells, which are extraordinary resistant to chemotherapeutics. This resistance is probably due to transporter molecules like ABCB5, which are highly expressed by a number of CSC’s including melanoma [104] and therefore efficiently counteract chemotherapy. Melanoma maintaining cells like other CSC’s are merely in a "dormant" state and replicate less frequently than the majority of cancer cells in the same lesion, which reduces the efficacy of anti-proliferative drugs. Low proliferative capacities together with the efficient export of chemotherapeutics contribute to CSC resistance toward a variety of therapeutic drugs. As a consequence, alternative strategies that specifically induce cell death of those cells are required. Moreover, the situation is exacerbated by the fact that the melanoma maintaining cells in the lesion are rare and unlikely to be eliminated by the random targeting provided by most therapeutic agents. Specific targeting by cytotoxic T cells redirected towards CD20 or by CD20-specific therapeutic antibodies like Rituxan (rituximab) or Arzerra (ofatumumab), probably as adjunct to a tumor de-bulking strategy, may improve the situation.

Second, whether the prevalence of CD20+ melanoma maintaining cells in a tumor lesion may correlate with clinical progression or relapse has to be addressed. If so, the frequency of CD20+ melanoma cells may serve as a surrogate marker for therapeutic efficacy and/or prognosis. Chemotherapy and/or radiation may induce amplification of these cells thus contributing to their accumulation during tumor progression and metastasis.

Third, melanoma maintaining cells may exhibit an extraordinary functional and phenotypic plasticity. As a consequence, continuous presence of targeting therapeutic agents will be required to eliminate those cells, which exhibit newly acquired melanoma initiating and/or maintaining capacities. In their pre-clinical model, Schmidt and colleagues [69] used CAR engineered T cells which penetrate tissues, scan for targets and persist for long-term acting as an antigen-specific guardian. These T cells are present in the targeted lesion as long as cells expressing the target antigen appear. Repetitive restimulation of these T cells, for instance by engaging their TCR with EBV-specific antigens [63, 81], may sustain persistence of CAR T cells in sufficient numbers over long periods of time. In this constellation, cellular therapy has a major advantage compared to pharmaceutical drugs, which are present in therapeutic levels for short periods; in the case of melanoma the required period for screening for re-appearance of such melanoma initiating cells may be many years. The development of an antigen-specific memory by adoptively transferred CAR T cells, as recently shown in a pre-clinical model [112], may be of benefit to patients in preventing a melanoma relapse.

Advertisement

Acknowledgements

Work in the author's laboratory was supported by the Deutsche Krebshilfe, Bonn and Ziel 2. NRW Programm of the Ministerium für Innovation, Wissenschaft, Forschung und Technologie des Landes Nordrhein-Westfalen and of the European Union.

Advertisement

Abbreviations

ACT, adoptive cell therapy; CAR, chimeric antigen receptor; CTLA-4, anti-cytotoxic T-lymphocyte-associated antigen-4; CSC, cancer stem cell; EBV, Epstein-Barr virus; GMP, Good Manufacturing Practice; IFN, interferon; IL, interleukin; TCR, T cell receptor; TIL, tumor infiltrating lymphocyte

References

  1. 1. Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Spatz A, Grob J-J, Malvehy J, Newton-Bishop J, Stratigos A, Pehamberger H, Eggermont A. Diagnosis and treatment of melanoma: European consensus-based interdisciplinary guideline. European Journal of Cancer 2010;46(2) 270-283.
  2. 2. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O'Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. The New England Journal of Medicine 2011;364 (26) 2507-2516.
  3. 3. Carter RD, Krementz ET, Hill GJ 2nd, Metter GE, Fletcher WS, Golomb FM, Grage TB, Minton JP, Sparks FC. DTIC (nsc-45388) and combination therapy for melanoma. I. Studies with DTIC, BCNU (NSC-409962), CCNU (NSC-79037), vincristine (NSC-67574), and hydroxyurea (NSC-32065). Cancer Treat Rep. 1976;60(5) 601-609.
  4. 4. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O'Dwyer PJ, Lee RJ, Grippo JF, Nolop K, Chapman PB. Inhibition of mutated, activated BRAF in metastatic melanoma. The New England Journal of Medicine 2010;363(9) 809-819.
  5. 5. Leiter U, Eigentler TK, Forschner A, Pflugfelder A, Weide B, Held L, Meier F, Garbe C. Excision guidelines and follow-up strategies in cutaneous melanoma: Facts and controversies. Clinics in Dermatology 2010;28(3) 311-315.
  6. 6. Denninghoff VC, Kahn AG, Falco J, Curutchet HP, Elsner B. Sentinel lymph node: detection of micrometastases of melanoma in a molecular study. Molecular Diagnosis 2004;8(4) 253-258.
  7. 7. Bedikian AY, Wei C, Detry M, Kim KB, Papadopoulos NE, Hwu WJ, Homsi J, Davies M, McIntyre S, Hwu P. Predictive Factors for the Development of Brain Metastasis in Advanced Unresectable Metastatic Melanoma. American Journal of Clinical Oncology 2010;34(6) 603-610.
  8. 8. Bradbury PA, Middleton MR. DNA repair pathways in drug resistance in melanoma. Anti-cancer Drugs 2004;15(5) 421-426.
  9. 9. Pak BJ, Chu W, Lu SJ, Kerbel RS, Ben-David Y. Lineage-specific mechanism of drug and radiation resistance in melanoma mediated by tyrosinase-related protein 2. Cancer Metastasis Reviews 2001;20(1-2) 27-32.
  10. 10. Pak BJ, Lee J, Thai BL, Fuchs SY, Shaked Y, Ronai Z, Kerbel RS, Ben-David Y. Radiation resistance of human melanoma analysed by retroviral insertional mutagenesis reveals a possible role for dopachrome tautomerase. Oncogene 2004;23(1) 30-38.
  11. 11. Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, Abrams J, Sznol M, Parkinson D, Hawkins M, Paradise C, Kunkel L, Rosenberg SA. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. Journal of Clinical Oncology 1999;17( 7) 2105-2116.
  12. 12. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. The New England Journal of Medicine 2010;363(8) 711-723.
  13. 13. Kirkwood JM, Ibrahim JG, Sondak VK, Richards J, Flaherty LE, Ernstoff MS, Smith TJ, Rao U, Steele M, Blum RH. High- and low-dose interferon alfa-2b in high-risk melanoma: first analysis of intergroup trial E1690/S9111/C9190. Journal of Clinical Oncology 2000;18(12) 2444-2458.
  14. 14. Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC, Blum RH. Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST 1684. Journal of Clinical Oncology 1996;14(1) 7-17.
  15. 15. Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology 2012;1(3) 306-315.
  16. 16. Bernatchez C, Radvanyi LG, Hwu P. Advances in the treatment of metastatic melanoma: adoptive T-cell therapy. Seminars in Oncology 2012;39(2) 215-226.
  17. 17. Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM. Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet 2007;370(9581) 59-67.
  18. 18. Nathanson. Spontaneous regression of malignant melanoma: a review of the literature on incidence, clinical features, and possible mechanisms. National Cancer Institute Monograph 1976;44 67-76.
  19. 19. Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. The journal of the American Medical Association 1994;271(12) 907-913.
  20. 20. Clark WH Jr, From L, Bernardino EA, Mihm MC. The histogenesis and biologic behavior of primary human malignant melanomas of the skin. Cancer Research 1969;29(3) 705-727.
  21. 21. Clemente CG, Mihm MC Jr, Bufalino R, Zurrida S, Collini P, Cascinelli N. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 1996;77(7) 1303-1310.
  22. 22. Burton AL, Roach BA, Mays MP, Chen AF, Ginter BA, Vierling AM, Scoggins CR, Martin RC, Stromberg AJ, Hagendoorn L, McMasters KM. Prognostic significance of tumor infiltrating lymphocytes in melanoma. The American Surgeon 2011;77(2) 188-192.
  23. 23. Vignard V, Lemercier B, Lim A, Pandolfino MC, Guilloux Y, Khammari A, Rabu C, Echasserieau K, Lang F, Gougeon ML, Dreno B, Jotereau F, Labarriere N. Adoptive transfer of tumor-reactive Melan-A-specific CTL clones in melanoma patients is followed by increased frequencies of additional Melan-A-specific T cells. Journal of Immunology 2005;175(7) 4797-4805.
  24. 24. Itzhaki O, Hovav E, Ziporen Y, Levy D, Kubi A, Zikich D, Hershkovitz L, Treves AJ, Shalmon B, Zippel D, Markel G, Shapira-Frommer R, Schachter J, Besser MJ. Establishment and large-scale expansion of minimally cultured "young" tumor infiltrating lymphocytes for adoptive transfer therapy. Journal of Immunology 2011;34(2) 212-220.
  25. 25. Shen X, Zhou J, Hathcock KS, Robbins P, Powell DJ Jr, Rosenberg SA, Hodes RJ. Persistence of tumor infiltrating lymphocytes in adoptive immunotherapy correlates with telomere length. Journal of Immunology 2007;30(1) 123-129.
  26. 26. Besser MJ, Shapira-Frommer R, Treves AJ, Zippel D, Itzhaki O, Schallmach E, Kubi A, Shalmon B, Hardan I, Catane R, Segal E, Markel G, Apter S, Nun AB, Kuchuk I, Shimoni A, Nagler A, Schachter J. Minimally cultured or selected autologous tumor-infiltrating lymphocytes after a lympho-depleting chemotherapy regimen in metastatic melanoma patients. Journal of Immunotherapy 2009;32(4) 415-423.
  27. 27. Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, Rogers LJ, Gracia GJ, Jones SA, Mangiameli DP, Pelletier MM, Gea-Banacloche J, Robinson MR, Berman DM, Filie AC, Abati A, Rosenberg SA. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. Journal of Clinical Oncology 2005;23(10) 2346-2357.
  28. 28. Besser MJ, Shapira-Frommer R, Treves AJ, Zippel D, Itzhaki O, Hershkovitz L, Levy D, Kubi A, Hovav E, Chermoshniuk N, Shalmon B, Hardan I, Catane R, Markel G, Apter S, Ben-Nun A, Kuchuk I, Shimoni A, Nagler A, Schachter J. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clinical Cancer Research 2010;16(9) 2646-2655.
  29. 29. Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, Robbins PF, Huang J, Citrin DE, Leitman SF, Wunderlich J, Restifo NP, Thomasian A, Downey SG, Smith FO, Klapper J, Morton K, Laurencot C, White DE, Rosenberg SA. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. Journal of Clinical Oncology 2008;26(32) 5233-5239.
  30. 30. Weber J, Atkins M, Hwu P, Radvanyi L, Sznol M, Yee C; Immunotherapy Task Force of the NCI Investigational Drug Steering Committee.
  31. 31. Kawakami Y, Eliyahu S, Jennings C, Sakaguchi K, Kang X, Southwood S, Robbins PF, Sette A, Appella E, Rosenberg SA. Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor-infiltrating T lymphocytes associated with in vivo tumor regression. Journal of Immunology 1995;154(8) 3961-39618.
  32. 32. Benlalam H, Vignard V, Khammari A, Bonnin A, Godet Y, Pandolfino MC, Jotereau F, Dreno B, Labarrière N. Infusion of Melan-A/Mart-1 specific tumor-infiltrating lymphocytes enhanced relapse-free survival of melanoma patients. Cancer Immunology Immunotherapy 2007;56(4) 515-526.
  33. 33. Khammari A, Labarrière N, Vignard V, Nguyen JM, Pandolfino MC, Knol AC, Quéreux G, Saiagh S, Brocard A, Jotereau F, Dreno B. Treatment of metastatic melanoma with autologous Melan-A/MART-1-specific cytotoxic T lymphocyte clones. The Journal of Investigative Dermatology 2009;129(12) 2835-2842.
  34. 34. Kvistborg P, Shu CJ, Heemskerk B, Fankhauser M, Thrue CA, Toebes M, van Rooij N, Linnemann C, van Buuren MM, Urbanus JH, Beltman JB, Thor Straten P, Li YF, Robbins PF, Besser MJ, Schachter J, Kenter GG, Dudley ME, Rosenberg SA, Haanen JB, Hadrup SR, Schumacher TN. TIL therapy broadens the tumor-reactive CD8(+) T cell compartment in melanoma patients. Oncoimmunology 2012;1(4) 409-418.
  35. 35. Johnson LA, Heemskerk B, Powell DJ Jr, Cohen CJ, Morgan RA, Dudley ME, Robbins PF, Rosenberg SA. Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes. Journal of Immunology 2006;177(9) 6548-6559.
  36. 36. Zhao Y, Zheng Z, Khong HT, Rosenberg SA, Morgan RA. Transduction of an HLA-DP4-restricted NY-ESO-1-specific TCR into primary human CD4+ lymphocytes. Journal of Immunology 2006;29(4) 398-406.
  37. 37. Frankel TL, Burns WR, Peng PD, Yu Z, Chinnasamy D, Wargo JA, Zheng Z, Restifo NP, Rosenberg SA, Morgan RA. Both CD4 and CD8 T cells mediate equally effective in vivo tumor treatment when engineered with a highly avid TCR targeting tyrosinase. Journal of Immunology 2010;184(11) 5988-5998.
  38. 38. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, Kammula US, Royal RE, Sherry RM, Wunderlich JR, Lee CC, Restifo NP, Schwarz SL, Cogdill AP, Bishop RJ, Kim H, Brewer CC, Rudy SF, VanWaes C, Davis JL, Mathur A, Ripley RT, Nathan DA, Laurencot CM, Rosenberg SA. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009;114(3) 535-546.
  39. 39. Spiotto MT, Rowley DA, and Schreiber H. “Bystander” elimination of antigen loss variants in established tumors. Nature Medicine 2004; 10(3) 294-298.
  40. 40. Schüler T, Blankenstein T. Cutting edge: CD8+ effector T cells reject tumors by direct antigen recognition but indirect action on host cells. Journal of Immunology 2003;170(9) 4427-4431.
  41. 41. Stewart-Jones G, Wadle A, Hombach A, Shenderov E, Held G, Fischer E, Kleber S, Nuber N, Stenner-Liewen F, Bauer S, McMichael A, Knuth A, Abken H, Hombach AA, Cerundolo V, Jones EY, Renner C. Rational development of high-affinity T-cell receptor-like antibodies. Proceedings of the National Academy of Sciences of the United States of America 2009;106(14) 5784-5788.
  42. 42. Willemsen RA, Debets R, Hart E, Hoogenboom HR, Bolhuis RL, Chames P. A phage display selected fab fragment with MHC class I-restricted specificity for MAGE-A1 allows for retargeting of primary human T lymphocytes. Gene Therapy 2001;8(21) 1601-1608.
  43. 43. Chmielewski M, Hombach A, Heuser C, Adams GP, Abken H.T cell activation by antibody-like immunoreceptors: increase in affinity of the single-chain fragment domain above threshold does not increase T cell activation against antigen-positive target cells but decreases selectivity. Journal of Immunology 2004; 173( 12) 7647-7653.
  44. 44. Chmielewski M, Hombach AA, Abken H. CD28 cosignalling does not affect the activation threshold in a chimeric antigen receptor-redirected T-cell attack. Gene Therapy 2011;18(1) 62-72.
  45. 45. Schaft N, Willemsen RA, de Vries J, Lankiewicz B, Essers BW, Gratama JW, Figdor CG, Bolhuis RL, Debets R, Adema GJ. Peptide fine specificity of anti-glycoprotein 100 CTL is preserved following transfer of engineered TCR alpha beta genes into primary human T lymphocytes. Journal of Immunology 2003; 170( 4) 2186-2194.
  46. 46. Morgan RA, Dudley ME, Yu YY, Zheng Z, Robbins PF, Theoret MR, Wunderlich JR, Hughes MS, Restifo NP, Rosenberg SA. High efficiency TCR gene transfer into primary human lymphocytes affords avid recognition of melanoma tumor antigen glycoprotein 100 and does not alter the recognition of autologous melanoma antigens,” Journal of Immunology 2003 ;171(6) 3287-3295.
  47. 47. Hughes MS, Yu YY, Dudley ME, Zheng Z, Robbins PF, Li Y, Wunderlich J, Hawley RG, Moayeri M, Rosenberg SA, Morgan RA. Transfer of a TCR gene derived from a patient with a marked antitumor response conveys highly active T-cell effector functions. Human Gene Therapy 2005;16(4) 457-472.
  48. 48. Willemsen R, Ronteltap C, Heuveling M, Debets R, Bolhuis R. Redirecting human CD4+ T lymphocytes to the MHC class I-restricted melanoma antigen MAGE-A1 by TCR alphabeta gene transfer requires CD8alpha. Gene Therapy 2005; 12(2) 140-146.
  49. 49. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA, Rosenberg SA. Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes. Science. 2006;314(5796) 126-129.
  50. 50. Coccoris M, Swart E, de Witte MA, van Heijst JW, Haanen JB, Schepers K, Schumacher TN. Long-term functionality of TCR-transduced T cells in vivo. Journal of Immunology 2008; 180(10) 6536-6543.
  51. 51. Seliger B. Molecular mechanisms of MHC class I abnormalities and APM components in human tumors. Cancer Immunology Immunotherapy 2008;57(11) 1719-1726.
  52. 52. Sigalotti L, Fratta E, Coral S, Tanzarella S, Danielli R, Colizzi F, Fonsatti E, Traversari C, Altomonte M, Maio M. Intratumor heterogeneity of cancer/testis antigens expression in human cutaneous melanoma is methylation-regulated and functionally reverted by 5-aza-2'-deoxycytidine. Cancer Research 2004;64(24) 9167-9171.
  53. 53. Vitale M, Pelusi G, Taroni B, Gobbi G, Micheloni C, Rezzani R, Donato F, Wang X, Ferrone S. HLA class I antigen down-regulation in primary ovary carcinoma lesions: association with disease stage. Clinical Cancer Research 2005;11(1) 67-72.
  54. 54. Coccoris M, Straetemans T, Govers C, Lamers C, Sleijfer S, Debets R. T cell receptor (TCR) gene therapy to treat melanoma: lessons from clinical and preclinical studies. Expert Opinion on Biological Therapy 2010;10(4) 547-562.
  55. 55. Bendle GM, Linnemann C, Hooijkaas AI, Bies L, de Witte MA, Jorritsma A, Kaiser AD, Pouw N, Debets R, Kieback E, Uckert W, Song JY, Haanen JB, Schumacher TN. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nature Medicine 2010; 16(5) 565-570.
  56. 56. Cohen CJ, Zhao Y, Zheng Z, Rosenberg SA, Morgan RA. Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Research 2006;66(17) 8878-8886.
  57. 57. Kuball J, Dossett ML, Wolfl M, Ho WY, Voss RH, Fowler C, Greenberg PD. Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood 2007;109(6) 2331-2338.
  58. 58. Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proceedings of the National Academy of Sciences of the United States of America 1993;90(2) 720-724.
  59. 59. Hombach A, Abken H. Costimulation tunes tumor-specific activation of redirected T cells in adoptive immunotherapy,” Cancer Immunology Immunotherapy 2007;56(5) 731-737.
  60. 60. Hombach AA, Abken H. Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling. International Journal of Cancer 2011;129(12) 2935-2944.
  61. 61. Bridgeman JS, Hawkins RE, Hombach AA, Abken H, Gilham DE.Building better chimeric antigen receptors for adoptive T cell therapy. Current Gene Therapy 2010; 10( 2) 77-90.
  62. 62. Gilham DE, Debets R, Pule M, Hawkins RE, Abken H. CAR-T cells and solid tumors: tuning T cells to challenge an inveterate foe. Trends in Molecular Medicine 2012;18(7) 377-384.
  63. 63. Savoldo B, Rooney CM, Di Stasi A, Abken H, Hombach A, Foster AE, Zhang L, Heslop HE, Brenner MK, Dotti G. Epstein Barr virus specific cytotoxic T lymphocytes expressing the anti-CD30zeta artificial chimeric T-cell receptor for immunotherapy of Hodgkin disease. Blood. 2007;110(7) 2620-2630.
  64. 64. ClinicalTrials.gov A service of the U.S. National Institutes of Health. Trial ID: NCT00586391 http://clinicaltrials.gov/ct2/results?term=NCT+00586391
  65. 65. ClinicalTrials.gov A service of the U.S. National Institutes of Health. Trial ID: NCT00709033 http://clinicaltrials.gov/ct2/results?term=NCT+00709033
  66. 66. ClinicalTrials.gov A service of the U.S. National Institutes of Health. Trial ID: NCT01218867 http://clinicaltrials.gov/ct2/results?term=NCT01218867
  67. 67. Reinhold U, Liu L, Lüdtke-Handjery HC, Heuser C, Hombach A, Wang X, Tilgen W, Ferrone S, Abken H. Specific lysis of melanoma cells by receptor grafted T cells is enhanced by anti-idiotypic monoclonal antibodies directed to the scFv domain of the receptor. The Journal of Investigative Dermatology 1999;112(5) 744-750.
  68. 68. Burns WR, Zhao Y, Frankel TL, Hinrichs CS, Zheng Z, Xu H, Feldman SA, Ferrone S, Rosenberg SA, Morgan RA. A high molecular weight melanoma-associated antigen-specific chimeric antigen receptor redirects lymphocytes to target human melanomas. Cancer Research 2010;70(8) 3027-3033.
  69. 69. Schmidt P, Kopecky C, Hombach A, Zigrino P, Mauch C, Abken H. Eradication of melanomas by targeted elimination of a minor subset of tumor cells. Proceedings of the National Academy of Sciences of the United States of America 2011;108(6) 2474-2479.
  70. 70. Yvon E, Del Vecchio M, Savoldo B, Hoyos V, Dutour A, Anichini A, Dotti G, Brenner MK. Immunotherapy of metastatic melanoma using genetically engineered GD2-specific T cells. Clinical Cancer Research 2009;15(18) 5852-5860.
  71. 71. Lo AS, Ma Q, Liu DL, Junghans RP. Anti-GD3 chimeric sFv-CD28/T-cell receptor zeta designer T cells for treatment of metastatic melanoma and other neuroectodermal tumors. Clinical Cancer Research 2010;16(10) 2769-2780.
  72. 72. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science Translational Medicine 2011;3(95) 95ra73.
  73. 73. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. The New England Journal of Medicine 2011;365(8) 725-733.
  74. 74. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. Journal of Clinical Oncology 2006; 24(3) 20-22.
  75. 75. Brentjens R, Yeh R, Bernal Y, Riviere I, Sadelain M. Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial. Molecular Therapy 2010;18(4) 666-668.
  76. 76. 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. Molecular Therapy 2010;8(4) 843-851.
  77. 77. Hawkins RE, Gilham DE, Debets R, Eshhar Z, Taylor N, Abken H, Schumacher TN, ATTACK Consortium. Development of adoptive cell therapy for cancer: a clinical perspective. Human Gene Therapy 2010;21(6) 665-672.
  78. 78. Büning H, Uckert W, Cichutek K, Hawkins RE, Abken H. Do CARs need a driver's license? Adoptive cell therapy with chimeric antigen receptor-redirected T cells has caused serious adverse events. Human Gene Therapy 2010;21( 9) 1039-1042.
  79. 79. Offringa R. Antigen choice in adoptive T-cell therapy of cancer. Current Opinion in Immunology 2009;21(2) 190-199.
  80. 80. Overwijk WW, Theoret MR, Finkelstein SE, Surman DR, de Jong LA, Vyth-Dreese FA, Dellemijn TA, Antony PA, Spiess PJ, Palmer DC, Heimann DM, Klebanoff CA, Yu Z, Hwang LN, Feigenbaum L, Kruisbeek AM, Rosenberg SA, Restifo NP. Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. The Journal of Experimental Medicine 2003;198(4) 569-580.
  81. 81. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, Huls MH, Liu E, Gee AP, Mei Z, Yvon E, Weiss HL, Liu H, Rooney CM, Heslop HE, Brenner MK. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma Nature Medicine 2008;14(11) 1264-1270.
  82. 82. Antony PA, Piccirillo CA, Akpinarli A, Finkelstein SE, Speiss PJ, Surman DR, Palmer DC, Chan CC, Klebanoff CA, Overwijk WW, Rosenberg SA, Restifo NP. CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. Journal of Immunology 2005;174(5) 2591-2601.
  83. 83. Meidenbauer N, Marienhagen J, Laumer M, Vogl S, Heymann J, Andreesen R, Mackensen A. Survival and tumor localization of adoptively transferred Melan-A-specific T cells in melanoma patients. Journal of Immunology 2003;170(4) 2161-2169.
  84. 84. Griffith KD, Read EJ, Carrasquillo JA, Carter CS, Yang JC, Fisher B, Aebersold P, Packard BS, Yu MY, Rosenberg SA. In vivo distribution of adoptively transferred indium-111-labeled tumor infiltrating lymphocytes and peripheral blood lymphocytes in patients with metastatic melanoma. Journal of the National Cancer Institute 1989;81(22) 1709-1717.
  85. 85. Fisher B, Packard BS, Read EJ, Carrasquillo JA, Carter CS, Topalian SL, Yang JC, Yolles P, Larson SM, Rosenberg SA. Tumor localization of adoptively transferred indium-111 labeled tumor infiltrating lymphocytes in patients with metastatic melanoma. Journal of Clinical Oncology 1989;7(2) 250-261.
  86. 86. Peng W, Ye Y, Rabinovich BA, Liu C, Lou Y, Zhang M, Whittington M, Yang Y, Overwijk WW, Lizée G, Hwu P. Transduction of tumor-specific T cells with CXCR2 chemokine receptor improves migration to tumor and antitumor immune responses. Clinical Cancer Research 2010;16(22) 5458-5468.
  87. 87. Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. Journal of Clinical Oncology 2006;24(31) 5060-5069.
  88. 88. Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, Celis E, Greenberg PD. 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. Proceedings of the National Academy of Science of the United States of America 2002;99(25) 16168-16173.
  89. 89. Lozupone F, Rivoltini L, Luciani F, Venditti M, Lugini L, Cova A, Squarcina P, Parmiani G, Belardelli F, Fais S. Adoptive transfer of an anti-MART-1(27-35)-specific CD8+ T cell clone leads to immunoselection of human melanoma antigen-loss variants in SCID mice. European Journal of Immunology 2003;33(2) 556-566.
  90. 90. Becker JC, Varki N, Gillies SD, Furukawa K, Reisfeld RA. An antibody-interleukin 2 fusion protein overcomes tumor heterogeneity by induction of a cellular immune response,” Proceedings of the National Academy of Sciences of the United States of America 1996;93(15) 7826-7831.
  91. 91. Chmielewski M, Kopecky C, Hombach AA, Abken H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Research 2011;71(17) 5697-5706.
  92. 92. Li Y, Liu S, Hernandez J, Vence L, Hwu P, Radvanyi L.MART-1-specific melanoma tumor-infiltrating lymphocytes maintaining CD28 expression have improved survival and expansion capability following antigenic restimulation in vitro. Journal of Immunology 2010;184(1) 452-465.
  93. 93. Hernandez-Chacon JA, Li Y, Wu RC, Bernatchez C, Wang Y, Weber JS, Hwu P, Radvanyi LG. Costimulation through the CD137/4-1BB pathway protects human melanoma tumor-infiltrating lymphocytes from activation-induced cell death and enhances antitumor effector function. Journal of Immunology 2011;34(3) 236-250.
  94. 94. Boni A, Cogdill AP, Dang P, Udayakumar D, Njauw CN, Sloss CM, Ferrone CR, Flaherty KT, Lawrence DP, Fisher DE, Tsao H, Wargo JA. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Research 2010;70(13) 5213-5219.
  95. 95. Wilmott JS, Long GV, Howle JR, Haydu LE, Sharma RN, Thompson JF, Kefford RF, Hersey P, Scolyer RA. Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma. Clinical Cancer Research 2012;18(5) 1386-1394.
  96. 96. Pegram HJ, Jackson JT, Smyth MJ, Kershaw MH, Darcy PK. Adoptive transfer of gene-modified primary NK cells can specifically inhibit tumor progression in vivo. Journal of Immunology 2008;181( 5) 3449-3455.
  97. 97. Kruschinski A, Moosmann A, Poschke I, Norell H, Chmielewski M, Seliger B, Kiessling R, Blankenstein T, Abken H, Charo J. Engineering antigen-specific primary human NK cells against HER-2 positive carcinomas,” Proceedings of the National Academy of Sciences of the United States of America 2008;105(45) 17481-17486.
  98. 98. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nature Medicine 1997;3(7) 730-737.
  99. 99. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proceedings of the National Academy of Sciences of the United States of America 2003;100(7) 3983-3988.
  100. 100. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, Hoey T, Gurney A, Huang EH, Simeone DM, Shelton AA, Parmiani G, Castelli C, Clarke MF. Phenotypic characterization of human colorectal cancer stem cells. Proceedings of the National Academy of Sciences of the United States of America 2007;104(24) 10158-10163.
  101. 101. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Research 2003;63(18) 5821-5828.
  102. 102. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of human colon-cancer-initiating cells. Nature 2007;445(7123) 111-115.
  103. 103. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM. Identification of pancreatic cancer stem cells. Cancer Research 2007;67(3) 1030-1037.
  104. 104. Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, Fuhlbrigge RC, Kupper TS, Sayegh MH, Frank MH. Identification of cells initiating human melanomas. Nature. 2008;451(7176) 345-349.
  105. 105. Zabierowski SE, Herlyn M. Melanoma stem cells: the dark seed of melanoma. Journal of Clinical Oncology 2008;26(17) 2890-2894.
  106. 106. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, Van Belle PA, Xu X, Elder DE, Herlyn M. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Research 2005;65(20) 9328-9337.
  107. 107. Zhou BB, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nature Reviews Drug Discovery 2009;8(10) 806-823.
  108. 108. Boiko AD, Razorenova OV, van de Rijn M, Swetter SM, Johnson DL, Ly DP, Butler PD, Yang GP, Joshua B, Kaplan MJ, Longaker MT, Weissman IL. Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature 2010;466(7302) 133-137.
  109. 109. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature 2008;456(7222) 593-598.
  110. 110. Quintana E, Shackleton M, Foster HR, Fullen DR, Sabel MS, Johnson TM, Morrison SJ. Phenotypic Heterogeneity among Tumorigenic Melanoma Cells from Patients that Is Reversible and Not Hierarchically Organized. Cancer Cell 2010;18(5) 510-523.
  111. 111. Schlaak M, Schmidt P, Bangard C, Kurschat P, Mauch C, Abken H. Regression of metastatic melanoma in a patient by antibody targeting of cancer stem cells. Oncotarget 2012;3(1) 22-30.
  112. 112. Chmielewski M, Rappl G, Hombach AA, Abken H. T cells redirected by a CD3ζ chimeric antigen receptor can establish self-antigen-specific tumour protection in the long term. Gene therapy 2012;doi: 10.1038/gt.2012.21. http://www.nature.com/gt/journal/vaop/ncurrent/full/gt201221a.html
  113. 113. Butler MO, Friedlander P, Milstein MI, Mooney MM, Metzler G, Murray AP, Tanaka M, Berezovskaya A, Imataki O, Drury L, Brennan L, Flavin M, Neuberg D, Stevenson K, Lawrence D, Hodi FS, Velazquez EF, Jaklitsch MT, Russell SE, Mihm M, Nadler LM, Hirano N. Establishment of antitumor memory in humans using in vitro-educated CD8+ T cells. Science Translational Medicine 2011;3(80) 80ra34.
  114. 114. Hong JJ, Rosenberg SA, Dudley ME, Yang JC, White DE, Butman JA, Sherry RM. Successful treatment of melanoma brain metastases with adoptive cell therapy. Clinical Cancer Research 2010;16(19) 4892–4898.
  115. 115. Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. Journal of the National Cancer Institute 1994;86(15) 1159–1166.
  116. 116. Dudley ME, Gross CA, Langhan MM, Garcia MR, Sherry RM, Yang JC, Phan GQ, Kammula US, Hughes MS, Citrin DE, Restifo NP, Wunderlich JR, Prieto PA, Hong JJ, Langan RC, Zlott DA, Morton KE, White DE, Laurencot CM, Rosenberg SA. CD8+ enriched "young" tumor infiltrating lymphocytes can mediate regression of metastatic melanoma. Clinical Cancer Research 2010;16(24) 6122-6131.
  117. 117. Heemskerk B, Liu K, Dudley ME, Johnson LA, Kaiser A, Downey S, Zheng Z, Shelton TE, Matsuda K, Robbins PF, Morgan RA, Rosenberg SA. Adoptive cell therapy for patients with melanoma, using tumor-infiltrating lymphocytes genetically engineered to secrete interleukin-2. Human Gene Therapy 2008;19(5) 496-510.
  118. 118. Wallen H, Thompson JA, Reilly JZ, Rodmyre RM, Cao J, Yee C. Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma. PloS One 2009;4(3) e4749. http://www.plosone.org/article/info%3Adoi%2F10.1371%2Fjournal.pone.0004749
  119. 119. Verdegaal EM, Visser M, Ramwadhdoebé TH, van der Minne CE, van Steijn JA, Kapiteijn E, Haanen JB, van der Burg SH, Nortier JW, Osanto S. Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ T cells in combination with low-dose interferon-alpha. Cancer Immunology Immunotherapy 2011;60(7) 953-963.
  120. 120. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, Dudley ME. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clinical Cancer Research 2011;17(13) 4550-4557.
  121. 121. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, Nahvi AV, Helman LJ, Mackall CL, Kammula US, Hughes MS, Restifo NP, Raffeld M, Lee CC, Levy CL, Li YF, El-Gamil M, Schwarz SL, Laurencot C, Rosenberg SA. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. Journal of Clinical Oncology 2011;29(7) 917-924.

Written By

Jennifer Makalowski and Hinrich Abken

Submitted: 19 April 2012 Published: 30 January 2013