Abstract
Purpose
This review demonstrates the importance of immunobiology and immunotherapy research for understanding and treating neuroblastoma.
Principal results
The first suggestions of immune system-neuroblastoma interactions came from in vitro experiments showing that lymphocytes from patients were cytotoxic for their own tumor cells and from evaluations of tumors from patients that showed infiltrations of immune system cells. With the development of monoclonal antibody (mAb) technology, a number of mAbs were generated against neuroblastoma cells lines and were used to define tumor associated antigens. Disialoganglioside (GD2) is one such antigen that is highly expressed by virtually all neuroblastoma cells and so is a useful target for both identification and treatment of tumor cells with mAbs. Preclinical research using in vitro and transplantable tumor models of neuroblastoma has demonstrated that cytotoxic T lymphocytes (CTLs) can specifically recognize and kill tumor cells as a result of vaccination or of genetic engineering that endows them with chimeric antigen receptors. However, CTL based clinical trials have not progressed beyond pilot and phase I studies. In contrast, anti-GD2 mAbs have been extensively studied and modified in pre-clinical experiments and have progressed from phase I through phase III clinical trials. Thus, the one proven beneficial immunotherapy for patients with high-risk neuroblastoma uses a chimeric anti-GD2 mAb combined with IL-2 and GM-CSF to treat patients after they have received intensive cyto-reductive chemotherapy, irradiation, and surgery. Ongoing pre-clinical and clinical research emphasizes vaccine, adoptive cell therapy, and mAb strategies. Recently it was shown that the neuroblastoma microenvironment is immunosuppressive and tumor growth promoting, and strategies to overcome this are being developed to enhance anti-tumor immunotherapy.
Conclusions
Our understanding of the immunobiology of neuroblastoma has increased immensely over the past 40 years, and clinical translation has shown that mAb based immunotherapy can contribute to improving treatment for high-risk patients. Continued immunobiology and pre-clinical therapeutic research will be translated into even more effective immunotherapeutic strategies that will be integrated with new cytotoxic drug and irradiation therapies to improve survival and quality of life for patients with high-risk neuroblastoma.
Keywords: neuroblastoma, immunology, immunotherapy, microenvironment, anti-GD2
Introduction
Neuroblastoma is the most common extracranial solid tumor of childhood, and 45% of patients have high-risk tumors, nearly all of which are metastatic (stage 4) when diagnosed (1–4). Although outcome has steadily improved over the past 20 years, event-free survival (EFS) is still only 45% for patients with high-risk, metastatic disease (5–7). This review will focus on this group of patients since those with low- and intermediate-risk disease are currently effectively treated with surgery alone or surgery and modest-dose chemotherapy and so are not likely to receive immunotherapy in the future.
Initial evidence suggesting immune responses to neuroblastoma was provided in 1968 when blood leukocytes, which were 50–70% lymphocytes, from nine children with neuroblastoma were reported to inhibit colony formation by neuroblastoma cells that had been cultured for 10–30 days prior to testing (8). These lymphocytes inhibited colony formation by both autologous and allogeneic neuroblastoma cells but did not affect growth of fibroblasts from the same donors. Plasma from these patients also was reported to inhibit tumor cell colony formation in the presence of complement (8). In this same time, primary tumors were reported to contain leukocytes (9, 10), and some localized and metastatic neuroblastomas were reported to regress spontaneously (11–13). Together, these studies supported the hypothesis that the immune system could develop an anti-neuroblastoma response. However, the technology available at the time was not sufficient to define the cellular or molecular basis for the observations, and so definitive conclusions could not be reached.
Current “standard” therapy for high-risk patients, given in sequence, consists of 1) intensive induction chemotherapy and surgery; 2) myeloablative consolidation chemotherapy, autologous hematopoietic progenitor cell transplantation, and local irradiation; and 3) 13-cis-retinoic acid (cisRA) combined with IL-2, GM-CSF, and anti-disialoganglioside (GD2) mAb ch14.18, which targets tumor cells (6, 7). Although EFS was improved by adding ch14.18, IL-2 and GM-CSF immunotherapy to cisRA, approximately 40% of patients still relapse during or after this therapy (7). Development of new and more effective immunotherapy strategies for treating minimal residual disease and possibly for treating clinically measurable disease will be based upon improved understanding of interactions between tumor cells and the immune system and upon maximizing anti-tumor cell immune responses while minimizing or blocking pro-tumor and immunosuppressive immune responses.
This paper will review anti-tumor and pro-tumor (Yin and Yang) functions of the immune system in the context of neuroblastoma. The development and implementation of clinical immunotherapy with mAb ch14.18 has been successful as demonstrated in a phase III randomized study (7). However, anti-neuroblastoma T cell therapy trials, including those testing vaccines and adoptive cell therapy, are in early development and not yet proven to be beneficial. Pro-tumor functions (Yang) of the immune system in the tumor microenvironment and their negative impact upon immunotherapy of neuroblastoma are just beginning to be recognized, and further improvement of immunotherapy will need to address the tumor microenvironment.
Cytotoxic T Lymphocytes
The discovery that human cytotoxic T lymphocytes (CTLs) recognize a peptide presented by MHC class I molecules on autologous melanoma cells (14–16) opened the way for defining T cell targets on tumor cells. The peptide recognized in these experiments was derived from a protein encoded by the melanoma antigen-1 gene (MAGE-1, which was subsequently renamed MAGEA1). This gene, which is expressed by cancer cells and but not by normal cells with the exception of testis, was found to be a member of a large multi-gene family that has been termed the cancer/testis antigen family (16–18). Neuroblastoma cell lines and tumors express MAGE-1/MAGEA1, MAGE-3/MAGEA3, MAGE-6/MAGEA6, and NY-ESO-1/CTAG1B (19) (20–22). Although these genes are expressed by human neuroblastoma cells, natural immune responses to their peptides have not been reported, and clinical vaccine trials have not been performed. There is one report that CTLs against peptides from MAGEA1, MAGEA3, and CTAG1B can be generated in vitro using T cells from normal donors and that these CTLs are cytotoxic against HLA class I compatible human neuroblastoma cells (23).
Clinical and pre-clinical studies have identified peptides from survivin, which is an inhibitor of apoptosis protein, as targets for CTLs. Survivin is expressed by many malignancies, including neuroblastoma (24, 25). In a study of nine patients with high-risk neuroblastoma, T cells specific for survivin were detected in blood at diagnosis by tetramer analysis, and circulating survivin-specific CTLs, after stimulation with survivin in vitro, were cytotoxic for autologous and HLA-compatible neuroblastoma cells. However, by immunohistochemistry, tumor-infiltrating T cells were few or absent in 26 of 26 tumors (25). Survivin specific CTLs from patients have been induced in vitro by monocyte-derived dendritic cells transfected with neuroblastoma cell line mRNA (26). Using a syngeneic mouse neuroblastoma model, protective tumor immunity that at least in part was due to CTLs that recognized survivin peptides was induced by immunization with Neuro-2A cells transfected with IL-21 (27). With the NXS2 murine neuroblastoma model, oral vaccination with a survivin DNA minigene was associated with increased target cell lysis, increased presence of CD8(+) T-cells at the primary tumor site, and enhanced production of pro-inflammatory cytokines. Therapeutic vaccination led to complete neuroblastoma eradication in over 50% of immunized mice and surviving mice showed an over 80% reduction in primary tumor growth upon re-challenge (28).
Pre-clinical studies have demonstrated that tyrosine hydroxylase and MYCN proteins, which are relatively specific for neuroblastoma cells compared to normal cells, include peptides that can be targets for CTL. Vaccination of mice with tyrosine hydroxylase DNA minigenes can induce CTLs, eradicate established primary NXS2 neuroblastoma tumors, and inhibit spontaneous metastases without induction of autoimmunity (29, 30). MYCN derived peptides have been reported to generate MHC class I restricted human CTL in vitro that lyse MYCN amplified human neuroblastoma cells (31).
Pre-clinical and clinical immunotherapy studies have used neuroblastoma cells that have been transduced with cytokine genes to provide multi-antigen vaccines without identifying the specific target antigens. Murine Neuro-2A neuroblastoma cells that were transfected with IL-2 were less tumorigenic than unmodified parent cells, induced protective immunity against parent cells, and prolonged survival of mice with established Neuro-2A tumors. These functions were dependent upon CD8+ T cells (32). The Neuro-2A model also was used to demonstrate that transfection of tumor cells with GM-CSF and IFNγ genes significantly improved their ability to induce protective immunity against liver tumors (33).
Early phase clinical trials have used cytokine transfected autologous or allogeneic neuroblastoma cells for immunization. IL-2 transfected autologous neuroblastoma cells induced antitumor immune responses in patients with neuroblastoma manifested by IgG antitumor antibody and increased CTL killing of autologous tumor cells. Clinically, five of ten patients had tumor responses (one complete and one partial response and three stable disease), and four of these five were shown to have coexisting antitumor cytotoxic activity (34). In a second study, 12 patients were immunized with an IL-2 transfected allogeneic neuroblastoma cell line, and although none showed any increase in direct cytotoxicity against the immunizing cell line, one had a partial response, 7 had stable disease, and 4 had progressive disease (35). Another phase I study used the same allogeneic neuroblastoma cell line transfected to secrete both IL-2 and lymphotactin/XCL1, a chemokine for T cells. Among 21 patients with relapsed or refractory neuroblastoma, the vaccine increased CD4+ T cells, NK cells, eosinophils, and serum IL-5. Six patients had increased NK cytolytic activity, and 15 made IgG antibodies that bound to the immunizing cell line. Measurable tumor responses included complete remission in two patients and partial response in one patient (36). In a phase I study, seven patients received lymphotactin/XCL1 and IL-2-secreting autologous neuroblastoma cells, and this was associated with increased tumor recognition as measured by enzyme-linked immunosorbent spot (ELISPOT) assays (two patients had IFNγ and three had IL-5 responses). Clinical responses did not occur (37). In the most recent study, 13 patients who were presumed to have minimal residual disease (8 in first remission and 5 after treatment for recurrent NB) received an autologous neuroblastoma cell-IL-2 vaccine. ELISPOT assays for IFNγ and IL-5 demonstrated that vaccination produced a rise in circulating CD4 and CD8 T cells responsive to stimulation by autologous tumor cells. Median EFS was 22 months for patients in first remission, and three months for all others. Four patients treated in first remission remain alive and three are without disease recurrence (29+, 33+ and 41+ months). It is not possible to determine if the vaccine enhanced EFS from this small study (38).
Strategies aiming to generate CTLs must take into account mechanisms by which neuroblastoma cells may avoid immune elimination. These include decreased expression of peptide presenting HLA class I molecules by tumor cells, which can impair target peptide recognition by CTLs (25, 39–41). Also, neuroblastoma cells express low levels of antigen processing genes, including LMP-2, LMP-7, and TAP-1, which are necessary for preparation of peptides from proteins for presentation by HLA class I molecules to CTLs (42, 43). Neuroblastoma cells may suppress activation of CTLs as they have been reported to produce soluble MICA, which inhibits activation of NK cells and T cells via their NKG2D receptors (44) and 4Ig-B7-H3/CD276, which inhibits activation of NK cells (45) and T cells (46). Neuroblastoma cells also induce monocytes to release HLA-G, which suppresses both CTL and NK mediated cytotoxicity by interacting with inhibitory receptors or inducing apoptosis via CD8 ligation or the Fas-FasL pathway (47, 48). Finally, as discussed below, neuroblastoma cells recruit monocytes where they differentiate in the tumor microenvironment into M2 polarized macrophages to secrete potentially immunosuppressive cytokines including IL-6, IL-10, and TGFβ1 (49). Thus, effective CTL anti-tumor responses require that these escape mechanisms be evaluated and, if present, be overcome.
NKT cells
Vα24-invariant (type I) natural killer T cells (NKTs) react to self- and microbial-derived glycolipids presented by the monomorphic HLA class-I-like molecule CD1d. They express an invariant TCR α-chain, Vα24-Jα18 which is preferentially paired with Vβ11 (50). NKT cells are cytotoxic for tumor cells only if the latter expresses CD1d, and neuroblastoma cells do not express this surface molecule. However, NKT cells, if stimulated, secrete IL-2, which activates NK cells, and so indirectly NKT cells can mediate cytotoxicity against neuroblastoma cells (51). NKT cells were found to infiltrate primary neuroblastomas from patients with metastatic disease, which was due to tumor cell secretion of the chemokine MCP-1/CCL2. CCL2 expression by tumors was found to be inversely correlated with MYCN proto-oncogene amplification and expression, and MYCN-high/CCL2-low expression was associated with the absence of NKT cells in primary tumors (52). Further studies showed that MYCN down-regulates expression of CCL2 and so impacts localization of NKT cells into neuroblastoma tumors. This was the first demonstration that an oncogene can regulate infiltration of immune system cells into the tumor microenvironment (53). A possible role for NKT cells in mediating anti-tumor responses is suggested by the discovery that NKT cells can kill monocytes pulsed with tumor lysates. Cotransfer of human monocytes and NKT cells to neuroblastoma-bearing NOD/SCID mice decreased monocyte number at the tumor site and suppressed tumor growth compared with mice transferred with monocytes alone (54). Clinical therapy trials attempting to enhance NKT antitumor functions have not yet been performed in patients with neuroblastoma.
NK cells
A number of studies support the importance of NK cells in the immune response to cancer. Intra-tumoral NK cells have been demonstrated in colorectal carcinoma, gastric carcinoma, and squamous cell lung cancer, and, in general, patients whose tumors have the highest levels have the best survival (55–58). NK cell immunoglobulin receptor FcγRIIIa functional polymorphism correlates with response to the IgG1 anti-CD20 mAb rituximab in patients with non-Hodgkin’s lymphoma and Waldenstrom’s macroglobulinemia indicating the importance of NK mediated ADCC (59–61). Neuroblastoma cells often do not express surface HLA class I and II molecules and so may be much better targets for NK cells than for CTLs (40, 41, 62–64). Two recent studies suggest an anti-tumor role for NK cells in high-risk neuroblastoma. In one, killer immunoglobulin-like receptor (KIR) and HLA gene polymorphisms which interact to govern NK cell function correlated with disease progression and survival in patients with high-risk disease treated with autologous hematopoietic stem cell transplant (AHSCT). Those with a “missing ligand” KIR-HLA compound genotype had a 46% lower risk of death at three years after AHSCT compared to patients who possessed all ligands for inhibitory KIR (65). In another, patients with relapsed or refractory neuroblastoma had a significantly better response to the anti-GD2 immunocytokine hu14.18/IL-2 if they had a KIR-HLA ligand mismatch (66). To date, studies of the relationship of FcγRIIIa functional polymorphism to outcome in patients with neuroblastoma who were treated with the ch14.18 mAb have not been performed.
NK cells are activated to be cytotoxic and secrete IFNγ by IL-2. NK cytotoxicity is perforin-dependent or -independent. The perforin-dependent pathway includes perforin, granzymes, and granulysin, whereas the perforin-independent includes FASL and TRAIL. Perforin-dependent cytotoxicity may be triggered by natural cytotoxicity receptors (NCR) NKp30, NKp44, and NKp46, the C-type lectin heterodimeric receptor CD94/NKG2C, 2B4 (CD244), NKG2D, and DNAM1 (67–69). NCR and DNAM1 have been demonstrated to be involved in NK cell cytotoxicity against neuroblastoma cells (70, 71).
IL-2 alone has been tested in phase I and II trials for patients with neuroblastoma, and, although immune effects were documented, no objective tumor responses were observed (72–74). Lenalidomide is an immune modulating drug that activates T cells to secrete IL-2, which in turn activates NK cell cytotoxicity and ADCC (75, 76). Clinical trials in children and adults demonstrated increased numbers of NK cells and cytotoxicity, decreased T regulatory cells, and increased secretion of IL-2, IL-15, and GM-CSF after 21 days of lenalidomide treatment (77, 78). In the phase I study of lenalidomide in children and adolescents with relapsed or refractory solid tumors no objective responses occurred, but the drug was well tolerated without reaching a maximum tolerated dose (77). Thus, lenalidomide may be useful for activating NK cells to enhance mAb immunotherapy of neuroblastoma.
Anti-Tumor Cell Monoclonal Antibodies
The development of monoclonal antibody (mAb) technology (79) ultimately resulted in creation of therapeutically effective anti-tumor cell mAbs including rituximab (anti-CD20), cetuximab (anti-EGFR), trastuzumab (anti-HER2), and ch14.18 (anti-GD2) (7, 80–83). Soon after the technology became available, multiple different mAbs were produced that recognize neuroblastoma cell associated antigens (84–95). Some were used to develop immunocytology, which improved detection and quantification of neuroblastoma cells in bone marrow and blood compared to morphologic evaluation (96, 97). Immunocytology evaluation of bone marrow and blood during induction chemotherapy provided prognostic information (98). It also demonstrated that 20% of autologous marrows that were harvested for AHSCT had detectible tumor cells (98). To minimize the possibility of infusing tumorigenic cells with the autologous transplant (99), mAbs were used for ex vivo immunomagnetic removal (“purging”) of tumor cells from autologous bone marrow (100). Purged autologous bone marrow with no immunocytology detectible tumor cells was used for AHSCT in the phase III randomized study that established that consolidation with myeloablative chemoradiotherapy was more effective than with nonmyeloablative chemotherapy (5, 6). Immunocytology showed that only 1% of peripheral blood stem cell collections had detectible tumor cells, and a randomized phase III study demonstrated equivalent event-free survival when purged vs. non-purged PBSC were used for hematopoietic reconstitution (neither had detectible tumor cells by immunocytology) (Kreissman, et al., in preparation, 2011). Thus, both immunocytology and immunomagnetic ex vivo purging contributed to improving treatment for patients with high-risk disease.
The most significant contribution of mAbs to treatment of high-risk patients came from the discovery that neuroblastoma cells express a high level of disialoganglioside (GD2) and from the generation of mAbs to this surface molecule (88, 91, 93, 94, 101, 102). Initial clinical studies demonstrated that anti-GD2 mAbs could induce responses in patients with neuroblastoma (95, 103–105), probably due to complement dependent cytotoxicity and antibody dependent cellular cytotoxicity (ADCC) (106–108). IL-2 and GM-CSF were shown to enhance ADCC in vitro by activating cytotoxic natural killer cells and neutrophils (106, 109–112). A phase I/IB study tested the combination of IL-2 and murine anti-GD2 antibody 14G2A in patients with recurrent neuroblastoma (113, 114). Phase I studies also tested the chimeric anti-GD2 mAb ch14.18 in refractory or relapsed patients (115) and in patients in first response following myeloablative therapy and AHSCT (116). A subsequent phase I study evaluated the combination of GM-CSF, IL-2 and ch14.18 therapy combined with cisRA following myeloablative therapy and AHSCT and found the regimen to be tolerable (117). This progression of clinical trials culminated in the recently completed phase III randomized study of cisRA together with ch14.18, IL-2, and GMCSF vs. cisRA only for children with high-risk neuroblastoma who had a clinical response to induction therapy and myeloablative consolidation therapy/AHSCT. Immunotherapy after consolidation significantly improved EFS (66+/−5% vs. 46+/−5% at 2 years, P=0.01) and overall survival (86+/−4% vs. 75+/−5% at 2 years, P=0.02 (7). This was the first demonstration that antibody based therapy improves EFS and overall survival. Although EFS was improved by adding immunotherapy to cisRA, approximately 40% of patients still relapsed during or after this therapy (7). Additionally, the combination of ch14.18 with IL-2 and GM-CSF has significant toxicities, including neuropathic pain, fever without neutropenia, infection, hypokalemia, hypotension, and capillary leak syndrome. Thus, a search for new agents to combine with ch14.18 to improve efficacy and decrease toxicity is justified.
Antibody-cytokine fusion proteins (immunocytokines) combine the targeting ability of antibodies with the functional activity of cytokines, and such molecules may improve antibody-based therapy by delivering cytokines to the microenvironment to both activate effector cells and modulate the microenvironment. Immunocytokines, depending upon their molecular composition, could create a microenvironment that results in 1) complement dependent cytotoxicity against tumor cells; 2) ADCC against tumor cells mediated by activated neutrophils, natural killer cells, and/or monocytes/macrophages; and 3) attraction of dendritic cells that process tumor associated antigens and then induce antitumor immune responses by T and/or B cells. To date, immunocytokine research has focused on ADCC mediated by NK cells and on induction of CTL. An anti-GD2/IL-2 immunocytokine eradicated hepatic metastases of neuroblastomas in SCID mice that had been reconstituted with human lymphokine (IL-2) activated killer cells (118–120). In contrast, the combination of monoclonal anti-GD2 antibody and IL-2 at doses equivalent to the immunocytokine only reduced tumor load. In a syngeneic murine model of GD2 expressing melanoma, targeting with an anti-GD2 antibody/IL-2 immunocytokine resulted in generation of CD8+ T lymphocytes that could eradicate tumor as well as prevent tumor growth (120). Based upon these data, phase I and II studies have tested a humanized anti-GD2/IL-2 immunocytokine (hu14.18/IL-2) in patients with refractory or relapsed neuroblastoma. In the phase I study of 27 patients, treatment with hu14.18/IL2 caused elevated serum levels of soluble IL-2 receptor alpha (sIL2Rα) and lymphocytosis. There were no measurable complete or partial responses to hu14.18/IL2; however, three patients showed evidence of antitumor activity (121). In the phase II study, 39 patients with recurrent or refractory neuroblastoma were enrolled (36 evaluable). No responses were seen for patients with disease measurable by standard radiographic criteria (stratum 1) (n = 13). Of 23 patients with disease evaluable only by 123I-metaiodobenzylguanidine (MIBG) scintigraphy and/or bone marrow histology (stratum 2), five patients (21.7%) responded; all had a complete response of 9, 13, 20, 30, and 35+ months duration. Grade 3 and 4 non-hematologic toxicities included capillary leak, hypoxia, pain, rash, allergic reaction, elevated transaminases, and hyperbilirubinemia, which were reversible within a few days of completing a treatment course. These results support further testing of hu14.18/IL2 in children with non-bulky high-risk neuroblastoma (122).
Another strategy for improving antibody-based immunotherapy is to identify drugs that both activate NK cells for ADCC and prevent immunosuppression by the tumor microenvironment. Lenalidomide is an immune modulating drug that activates T cells to secrete IL-2, which in turn activates NK cell cytotoxicity and ADCC (75, 76). Preclinical in vitro data and mouse xenograft studies have shown that lenalidomide can enhance ADCC mediated by NK cells with rituximab (anti-CD20) against lymphoma and chronic lymphocytic leukemia cells (123–125), with SGN-40 (anti-CD40) against multiple myeloma and chronic lymphocytic leukemia cells (126, 127), and with trastuzumab (anti-HER2/neu) and cetuximab (anti-EGFR) against solid tumor cell lines (128). A phase I trial in children and adolescents with refractory solid tumors demonstrated increased NK cell numbers and cytotoxicity, decreased T regulatory cells, and increased serum IL-2, IL-15, and GM-CSF after 21 days of lenalidomide treatment (77). Our pre-clinical research demonstrated that lenalidomide enhances IL-2-mediated activation of NK cells, prevents their suppression by IL-6 and TGFβ1, which are in the neuroblastoma microenvironment, and increases ADCC in vitro and in NOD/SCID mice with mAb ch14.18 (Xu, et al., submitted for publication). Based upon these clinical and pre-clinical data, a phase I trial to test lenalidomide in combination with ch14.18 in patients with refractory or relapsed neuroblastoma is being developed by the New Approaches to Neuroblastoma Therapy (NANT) consortium.
Adoptive Cell Therapy with T cells and NK cells
Adoptive cell therapy (ACT) for high-risk neuroblastoma has focused upon growing autologous T cells ex vivo and transfecting them with cDNA encoding chimeric antigen receptors (CARs) that ligate tumor cell surface antigens and also provide activating signals to the T cells (129). CARs usually use a single chain fraction variable (scFv) antibody-derived motif for recognizing a cell surface antigen; importantly, such recognition is independent of antigen processing or MHC-restricted presentation. Primary human T cells with a CD28-like CAR specific for GD2 had enhanced survival and proliferation upon receptor stimulation (130). T cells engineered to express an anti-GD2 CAR (scFv from mAb 14.G2a linked to TCR ζ) recognized and lysed GD2-expressing neuroblastoma cells and secreted IFNγ in an antigen-specific manner. However, functionality declined over time in vitro, and antigenic stimulation did not induce proliferation (131). It was then shown that EBV-specific T cells, which were transduced with the anti-GD2 CAR gene, could be expanded and maintained long-term in the presence of EBV-infected B cells. These T cells efficiently lysed both EBV infected cells and GD2 expressing cells (132). Next, it was shown in a clinical trial that enrolled 11 patients that infusion of these genetically modified cells was safe and was associated with tumor regression or necrosis in four of the eight evaluable patients (two responses, two stable disease) (133). T cells expressing a CAR that targets the L1-CAM molecule on neuroblastoma cells were engineered by fusing the scFv of mAb CE7 to human IgG1 hinge-Fc, the transmembrane portion of human CD4, and the cytoplasmic tail of the human CD3-zeta chain. Primary human CD8+ CTL clones expressing this CAR specifically lysed human neuroblastoma cells and secreted GM-CSF, TNFα, and IFNγ (134). A clinical study of six patients in which autologous CD8+ T cells with anti-L1-CAM CAR were tested showed no overt toxicities to tissues known to express L1-CAM. Persistence of the CTL clones in the circulation was short (1––7 days) in patients with bulky disease but longer (42 days) in a patient with limited disease whose tumor partially responded (135). Migration to sites of disease is essential, and since MYCN non-amplified neuroblastomas express the chemokine CCL2 (52), it was determined if engineering T cells to express an anti-GD2 CAR and CCR2b, a receptor for CCL2, enhanced their migration. Injection of such T cells intravenously into SCID mice with subcutaneous neuroblastoma demonstrated specific migration, intra-tumor T cell expansion, and enhanced anti-tumor effect compared to anti-GD2 CAR T cells without CCR2b (136). Finally, expression of constitutively active Akt in anti-GD2 CAR T cells increased their proliferation and cytokine production, provided resistance to apoptosis, rendered them relatively resistant to suppression by and conversion into regulatory T cells, and increased cytotoxicity for GD2 expressing neuroblastoma cells in vitro (137).
Adoptive cell therapy with NK cells alone and especially combined with mAbs has therapeutic potential for a wide variety of human malignancies, including neuroblastoma. Recently, the ability to harvest large numbers of peripheral blood lymphocytes by leukapheresis, deplete alloreactive T cells, and activate the remaining NK cells with IL-2 has enabled haploidentical NK cell adoptive immunotherapy for AML in adults where remission in 5 of 19 poor-prognosis patients was associated with KIR mismatch (138). In a similar study, KIR-mismatched NK cell infusion was reported to be safe as post-remission consolidation therapy for children with AML with no relapses reported in the 10 patients treated (139). As noted above, patients with stage 4 neuroblastoma undergoing AHSCT who lacked a HLA ligand had a 46% lower risk of death and a 34% lower risk of progression at 3 years compared to those who possessed all ligands for their inhibitory KIRs. Among all KIR-HLA combinations, 16 patients lacking the HLA-C1 ligand for KIR2DL2/KIR2DL3 had the highest 3-year survival rate of 81%. In this study, survival was more strongly associated with "missing ligand" than with tumor MYCN gene amplification (65).
The inability to obtain large numbers of pure NK cells has limited testing the potential efficacy of NK adoptive cell therapy. A novel ex vivo expansion method for NK cells that uses an artificial antigen-presenting cell (aAPC) genetically engineered to express membrane bound IL-21, which signals primarily through STAT3, resolves this problem (140). The IL-21-expressing aAPCs promote sustained proliferation of mature NK cells without senescence by increasing the telomere length in the expanded cells. The IL-21 aAPC enable large-scale expansion of NK cells from a small volume of peripheral blood, sufficient to deliver multiple infusions of NK cells at high cell doses. These expanded NK cells display a highly-activated phenotype and participate in ADCC with mAb ch14.18 to kill neuroblastoma cells in vitro and in models of neuroblastoma in NOD/SCID mice. This pre-clinical data, as well as pre-clinical and clinical data for lenalidomide, provide the basis for developing a phase I clinical trial by MD Anderson Cancer Center and the NANT consortium that will test autologous, ex vivo expanded and activated NK cells combined with lenalidomide and mAb ch14.18 in patients with refractory or relapsed neuroblastoma.
Microenvironment Promotion of Tumor Growth and Immunosuppression
The tumor microenvironment, whether primary or metastatic, is a complex milieu created by tumor cells interacting with normal host cells. Like the yin and yang concept, immune system cells can eliminate tumor cells, promote their growth and dissemination, and suppress antitumor immune responses (141, 142). The importance of the microenvironment in which neuroblastoma cells grow has not been widely appreciated for developing new therapies. The first suggestion that immune system cells promote neuroblastoma progression came from gene expression analysis of high-risk, metastatic neuroblastomas without MYCN gene amplification using Affymetrix® microarrays and TaqMan® low-density arrays (143). This research revealed that high expression of genes related to B lymphocytes, macrophages, and inflammation, including IL-6, IL-6R, IL-10, and TGFβ1, was associated with poor 5-year EFS (54, 143). Furthermore, high-risk primary neuroblastomas had CD68+ tumor-associated monocytes/macrophages (TAMs) expressing IL-6, and bone marrows with neuroblastoma metastases had CD33+CD14+ myelomonocytic cells, also expressing IL-6 (54). Experimentally, neuroblastoma cells stimulate blood monocytes to secrete IL-6, and TAMs stimulate the growth of neuroblastoma cells in NOD/SCID mice, at least partially via IL-6 secretion (54). TGFβ1 activates the SMAD2/3 pathway in human NK cells, which results in suppression of ADCC and IFNγ, TNFα, and GM-CSF secretion in vitro (144, 145). The possible role of B lymphocytes in neuroblastoma progression is not yet clear. However, in murine cancer models, B cells have been shown to secrete IL-10 as well as antibodies that form immune complexes in the microenvironment, and both contribute to M2 polarization of TAMs. (141, 146, 147) (142)
T regulatory cells (Tregs), which have a CD4+ CD25+ Foxp3+ phenotype, have not been evaluated in human neuroblastoma primary tumors or metastatic sites. These cells secrete TGFβ1 and IL-10, which are present in primary tumors from patients with stage 4 disease and which are known to suppress immune responses. Tregs have been studied using murine neuroblastoma models. Inhibition of CD25+ Tregs by treating mice with an anti-CD25 mAb partially depleted CD4+ Foxp3+ T cells and enhanced vaccine-induced immunity to challenge with Neuro-2A neuroblastoma cells (148). In another study, treatment of mice with an anti-CD4 mAb depleted 90% of Tregs and potentiated a Neuro-2A/IL-21 cell vaccine. 80% of mice did not develop disseminated disease after intravenous injection of tumor cells, and this was dependent upon CD8+ T cells (149). In a third study, mice bearing subcutaneous Neuro-2A (AGN2a subclone) neuroblastoma tumors were treated with total body irradiation followed by transplantation with syngeneic bone marrow combined with T cells. Mice receiving CD25+ depleted T cells had increased post-transplantation vaccine-induced immunity and survival (150).
Future Directions
New immunotherapeutic strategies for improving survival of patients with high-risk neuroblastoma will require continued improvement in understanding tumor and host cell biology. Biologic and immunobiologic knowledge will drive developmental therapeutics research with in vitro and in vivo models of neuroblastoma. Pre-clinical strategies that are promising and preferably supported by clinical data from studies of adults with cancer will be tested in early phase clinical trials. Some strategies may show convincing benefit in well-designed early phase studies, but most are likely to require randomized phase III trials to definitively evaluate their efficacy. There are several important considerations in this discovery to development to delivery, bench to bedside to bench paradigm.
In discovery research, interactions of neuroblastoma and immune system cells that eliminate tumor cells (yin) and that promote immune suppression and tumor growth (yang) in primary and metastatic sites will be defined. Understanding how tumor cells create microenvironments that enhance their survival and dissemination while suppressing anti-tumor immune responses is particularly important. Well-characterized human neuroblastoma cell lines, which represent high-risk disease, provide models for in vitro and in vivo studies using human cells. Primary, short-term cultures of neuroblastoma cells, such as bone marrow metastases, allow studies of autologous interactions. Murine transgenic neuroblastomas that are MYCN amplified or non-amplified provide models of human neuroblastoma in immune competent mice, and these mice can be genetically manipulated to test hypotheses about immune system cells, pathways, and effector molecules. All models should be correlated with data from human primary tumors and bone marrow metastases to obtain clinical validation.
In the development phase, pre-clinical therapeutic research, which is based upon discovered immunobiology, tests new treatments against neuroblastoma cells in primary and metastatic sites. A key goal must be to successfully target the tumor microenvironment to tip the balance in favor of anti-tumor immunotherapy. This research uses the same models used in discovery to integrate immunobiology with immunotherapy. It is advisable to prioritize immunotherapeutic strategies that are most applicable for clinical delivery to patients and so, for example, to focus upon drugs and mAbs that already are approved or are likely to be approved by the FDA rather than agents that have not yet entered the clinical testing pipeline. Such prioritization will maximize the likelihood that agents shown to be effective in pre-clinical studies will be available for clinical testing. In this context, vaccine and adoptive cell therapy strategies, to be successful, will need to focus upon both efficacy in pre-clinical models and upon methods for efficient broad clinical application. An important aspect of this phase of immunotherapy research is the parallel development of assays for pharmacokinetics, pharmacodynamics, and tumor response, the latter seeking to provide an early surrogate of efficacy.
In the delivery phase, phase I clinical trials test the new immunotherapeutic strategy to determine appropriate dose and schedule, pharmacology and pharmacodynamics, and, within the context of such trials, anti-tumor activity. These trials generally require a consortium of several institutions with expertise in early phase trials for efficient implementation of protocols and accrual of patients with refractory or recurrent disease. Initial information about biomarker strategies should be obtained in early phase clinical studies. Definitive testing of the treatment and of the biomarkers is obtained from phase II and III clinical trials.
In developing new immunotherapies, it is important to consider their role in the overall treatment of high-risk neuroblastoma. Current dose intensive chemotherapy is likely to be at the limit of both anti-tumor efficacy and patient tolerance, and post consolidation therapy, even though including antibody immunotherapy, does not eradicate minimal residual disease (MRD) in many patients. Appropriate immunotherapy strategies will be developed that will contribute to improving induction, consolidation, and post-consolidation treatments. These new strategies will come from learning how to harness the power of the immune system to eliminate tumor cells and from learning how to integrate immunotherapy with non-specific and targeted drug therapies.
Acknowledgements
This work was supported by grants to R. Seeger from the National Cancer Institute (2R01 CA60104-16 and 5 P01 CA81403-12), the ThinkCure Foundation, the Bogart Pediatric Cancere Research Foundation, and the T.J. Martell Foundation for Leukemia, Cancer, and AIDS Research.
Abbreviations
- mAb
monoclonal antibody
- CTL
cytotoxic T lymphocyte
- GD2
disialoganglioside
- EFS
event-free survival
- NKT
Vα24-invariant (type I) natural killer T cell
- NK cell
natural killer cell
- ADCC
antibody dependent cellular cytotoxicity
- AHSCT
autologous hematopoietic stem cell transplantation
- TAM
tumor associated macrophage
- Tregs
T regulatory cells
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosures of potential conflicts of interest: none
Reference List
- 1.Seeger RC, Brodeur GM, Sather H, Dalton A, Siegel SE, Wong KY, Hammond D. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N.Engl.J.Med. 1985;313:1111–1116. doi: 10.1056/NEJM198510313131802. [DOI] [PubMed] [Google Scholar]
- 2.Shimada H, Stram DO, Chatten J, Joshi VV, Hachitanda Y, Brodeur GM, Lukens JN, Matthay KK, Seeger RC. Identification of subsets of neuroblastomas by combined histopathologic and N-myc analysis. J.Natl.Cancer Inst. 1995;87:1470–1476. doi: 10.1093/jnci/87.19.1470. [DOI] [PubMed] [Google Scholar]
- 3.Schmidt ML, Lal A, Seeger RC, Maris JM, Shimada H, O'Leary M, Gerbing RB, Matthay KK. Favorable prognosis for patients 12 to 18 months of age with stage 4 nonamplified MYCN neuroblastoma: a Children's Cancer Group Study. J Clin Oncol. 2005;23:6474–6480. doi: 10.1200/JCO.2005.05.183. [DOI] [PubMed] [Google Scholar]
- 4.London WB, Castleberry RP, Matthay KK, Look AT, Seeger RC, Shimada H, Thorner P, Brodeur G, Maris JM, Reynolds CP, et al. Evidence for an age cutoff greater than 365 days for neuroblastoma risk group stratification in the Children's Oncology Group. J Clin Oncol. 2005;23:6459–6465. doi: 10.1200/JCO.2005.05.571. [DOI] [PubMed] [Google Scholar]
- 5.Matthay KK, Villablanca JG, Seeger RC, Stram DO, Harris RE, Ramsay NK, Swift P, Shimada H, Black CT, Brodeur GM, et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children's Cancer Group. N.Engl.J.Med. 1999;341:1165–1173. doi: 10.1056/NEJM199910143411601. [DOI] [PubMed] [Google Scholar]
- 6.Matthay KK, Reynolds CP, Seeger RC, Shimada H, Adkins ES, Haas-Kogan D, Gerbing RB, London WB, Villablanca JG. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children's oncology group study. J.Clin.Oncol. 2009;27:1007–1013. doi: 10.1200/JCO.2007.13.8925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, Smith M, Anderson B, Villablanca JG, Matthay KK, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363:1324–1334. doi: 10.1056/NEJMoa0911123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hellstrom IE, Hellstrom KE, Pierce GE, Bill AH. Demonstration of cellbound and humoral immunity against neuroblastoma cells. Proc Natl Acad Sci U S A. 1968;60:1231–1238. doi: 10.1073/pnas.60.4.1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bill AH. The implications of immune reactions to neuroblastoma. Surgery. 1969;66:415–418. [PubMed] [Google Scholar]
- 10.Bill AH, Morgan A. Evidence for immune reactions to neuroblastoma and future possibilities for investigation. J Pediatr Surg. 1970;5:111–116. doi: 10.1016/0022-3468(70)90266-6. [DOI] [PubMed] [Google Scholar]
- 11.D'Angio GJ, Evans AE, Koop CE. Special pattern of widespread neuroblastoma with a favourable prognosis. Lancet. 1971;1:1046–1049. doi: 10.1016/s0140-6736(71)91606-0. [DOI] [PubMed] [Google Scholar]
- 12.Evans AE, Gerson J, Schnaufer L. Spontaneous regression of neuroblastoma. Natl Cancer Inst Monogr. 1976;44:49–54. [PubMed] [Google Scholar]
- 13.Cole WH. Spontaneous regression of cancer and the importance of finding its cause. Natl Cancer Inst Monogr. 1976;44:5–9. [PubMed] [Google Scholar]
- 14.van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, Knuth A, Boon T. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254:1643–1647. doi: 10.1126/science.1840703. [DOI] [PubMed] [Google Scholar]
- 15.Traversari C, van der Bruggen P, Luescher IF, Lurquin C, Chomez P, Van Pel A, De Plaen E, Amar-Costesec A, Boon T. A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E. J Exp Med. 1992;176:1453–1457. doi: 10.1084/jem.176.5.1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Boon T, Coulie PG, Van den Eynde BJ, van der Bruggen P. Human T cell responses against melanoma. Annu Rev Immunol. 2006;24:175–208. doi: 10.1146/annurev.immunol.24.021605.090733. [DOI] [PubMed] [Google Scholar]
- 17.Chomez P, De Backer O, Bertrand M, De Plaen E, Boon T, Lucas S. An overview of the MAGE gene family with the identification of all human members of the family. Cancer Res. 2001;61:5544–5551. [PubMed] [Google Scholar]
- 18.Scanlan MJ, Gure AO, Jungbluth AA, Old LJ, Chen YT. Cancer/testis antigens: an expanding family of targets for cancer immunotherapy. Immunol Rev. 2002;188:22–32. doi: 10.1034/j.1600-065x.2002.18803.x. [DOI] [PubMed] [Google Scholar]
- 19.Corrias MV, Scaruffi P, Occhino M, De Bernardi B, Tonini GP, Pistoia V. Expression of MAGE-1, MAGE-3 and MART-1 genes in neuroblastoma. Int J Cancer. 1996;69:403–407. doi: 10.1002/(SICI)1097-0215(19961021)69:5<403::AID-IJC9>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
- 20.Ishida H, Matsumura T, Salgaller ML, Ohmizono Y, Kadono Y, Sawada T. MAGE-1 and MAGE-3 or-6 expression in neuroblastoma-related pediatric solid tumors. Int.J.Cancer. 1996;69:375–380. doi: 10.1002/(SICI)1097-0215(19961021)69:5<375::AID-IJC4>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
- 21.Soling A, Schurr P, Berthold F. Expression and clinical relevance of NY-ESO-1, MAGE-1 and MAGE-3 in neuroblastoma. Anticancer Res. 1999;19:2205–2209. [PubMed] [Google Scholar]
- 22.Wolfl M, Jungbluth AA, Garrido F, Cabrera T, Meyen-Southard S, Spitz R, Ernestus K, Berthold F. Expression of MHC class I, MHC class II, and cancer germline antigens in neuroblastoma. Cancer Immunol Immunother. 2005;54:400–406. doi: 10.1007/s00262-004-0603-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bao L, Dunham K, Lucas K. MAGE-A1, MAGE-A3, and NY-ESO-1 can be upregulated on neuroblastoma cells to facilitate cytotoxic T lymphocyte-mediated tumor cell killing. Cancer Immunol Immunother. 2011 doi: 10.1007/s00262-011-1037-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Andersen MH, Svane IM, Becker JC, Straten PT. The universal character of the tumor-associated antigen survivin. Clin Cancer Res. 2007;13:5991–5994. doi: 10.1158/1078-0432.CCR-07-0686. [DOI] [PubMed] [Google Scholar]
- 25.Coughlin CM, Fleming MD, Carroll RG, Pawel BR, Hogarty MD, Shan X, Vance BA, Cohen JN, Jairaj S, Lord EM, et al. Immunosurveillance and survivin-specific T-cell immunity in children with high-risk neuroblastoma. J Clin Oncol. 2006;24:5725–5734. doi: 10.1200/JCO.2005.05.3314. [DOI] [PubMed] [Google Scholar]
- 26.Morandi F, Chiesa S, Bocca P, Millo E, Salis A, Solari M, Pistoia V, Prigione I. Tumor mRNA-transfected dendritic cells stimulate the generation of CTL that recognize neuroblastoma-associated antigens and kill tumor cells: immunotherapeutic implications. Neoplasia. 2006;8:833–842. doi: 10.1593/neo.06415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Croce M, Meazza R, Orengo AM, Fabbi M, Borghi M, Ribatti D, Nico B, Carlini B, Pistoia V, Corrias MV, et al. Immunotherapy of neuroblastoma by an Interleukin-21-secreting cell vaccine involves survivin as antigen. Cancer Immunol Immunother. 2008;57:1625–1634. doi: 10.1007/s00262-008-0496-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Fest S, Huebener N, Bleeke M, Durmus T, Stermann A, Woehler A, Baykan B, Zenclussen AC, Michalsky E, Jaeger IS, et al. Survivin minigene DNA vaccination is effective against neuroblastoma. Int J Cancer. 2009;125:104–114. doi: 10.1002/ijc.24291. [DOI] [PubMed] [Google Scholar]
- 29.Huebener N, Fest S, Strandsby A, Michalsky E, Preissner R, Zeng Y, Gaedicke G, Lode HN. A rationally designed tyrosine hydroxylase DNA vaccine induces specific antineuroblastoma immunity. Mol Cancer Ther. 2008;7:2241–2251. doi: 10.1158/1535-7163.MCT-08-0109. [DOI] [PubMed] [Google Scholar]
- 30.Huebener N, Fest S, Hilt K, Schramm A, Eggert A, Durmus T, Woehler A, Stermann A, Bleeke M, Baykan B, et al. Xenogeneic immunization with human tyrosine hydroxylase DNA vaccines suppresses growth of established neuroblastoma. Mol Cancer Ther. 2009;8:2392–2401. doi: 10.1158/1535-7163.MCT-09-0107. [DOI] [PubMed] [Google Scholar]
- 31.Sarkar AK, Nuchtern JG. Lysis of MYCN-amplified neuroblastoma cells by MYCN peptide-specific cytotoxic T lymphocytes. Cancer Res. 2000;60:1908–1913. [PubMed] [Google Scholar]
- 32.Katsanis E, Orchard PJ, Bausero MA, Gorden KB, McIvor RS, Blazar BR. Interleukin-2 gene transfer into murine neuroblastoma decreases tumorigenicity and enhances systemic immunity causing regression of preestablished retroperitoneal tumors. J Immunother Emphasis Tumor Immunol. 1994;15:81–90. doi: 10.1097/00002371-199402000-00001. [DOI] [PubMed] [Google Scholar]
- 33.Bausero MA, Panoskaltsis-Mortari A, Blazar BR, Katsanis E. Effective immunization against neuroblastoma using double-transduced tumor cells secreting GMCSF and interferon-gamma. J Immunother Emphasis Tumor Immunol. 1996;19:113–124. doi: 10.1097/00002371-199603000-00004. [DOI] [PubMed] [Google Scholar]
- 34.Bowman L, Grossmann M, Rill D, Brown M, Zhong WY, Alexander B, Leimig T, Coustan-Smith E, Campana D, Jenkins J, et al. IL-2 adenovector-transduced autologous tumor cells induce antitumor immune responses in patients with neuroblastoma. Blood. 1998;92:1941–1949. [PubMed] [Google Scholar]
- 35.Bowman LC, Grossmann M, Rill D, Brown M, Zhong WY, Alexander B, Leimig T, Coustan-Smith E, Campana D, Jenkins J, et al. Interleukin-2 gene-modified allogeneic tumor cells for treatment of relapsed neuroblastoma. Hum Gene Ther. 1998;9:1303–1311. doi: 10.1089/hum.1998.9.9-1303. [DOI] [PubMed] [Google Scholar]
- 36.Rousseau RF, Haight AE, Hirschmann-Jax C, Yvon ES, Rill DR, Mei Z, Smith SC, Inman S, Cooper K, Alcoser P, et al. Local and systemic effects of an allogeneic tumor cell vaccine combining transgenic human lymphotactin with interleukin-2 in patients with advanced or refractory neuroblastoma. Blood. 2003;101:1718–1726. doi: 10.1182/blood-2002-08-2493. [DOI] [PubMed] [Google Scholar]
- 37.Russell HV, Strother D, Mei Z, Rill D, Popek E, Biagi E, Yvon E, Brenner M, Rousseau R. Phase I trial of vaccination with autologous neuroblastoma tumor cells genetically modified to secrete IL-2 and lymphotactin. J Immunother. 2007;30:227–233. doi: 10.1097/01.cji.0000211335.14385.57. [DOI] [PubMed] [Google Scholar]
- 38.Russell HV, Strother D, Mei Z, Rill D, Popek E, Biagi E, Yvon E, Brenner M, Rousseau R. A phase 1/2 study of autologous neuroblastoma tumor cells genetically modified to secrete IL-2 in patients with high-risk neuroblastoma. J Immunother. 2008;31:812–819. doi: 10.1097/CJI.0b013e3181869893. [DOI] [PubMed] [Google Scholar]
- 39.Lampson LA, Fisher CA. Weak HLA and beta 2-microglobulin expression of neuronal cell lines can be modulated by interferon. Proc Natl Acad Sci U S A. 1984;81:6476–6480. doi: 10.1073/pnas.81.20.6476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Main EK, Lampson LA, Hart MK, Kornbluth J, Wilson DB. Human neuroblastoma cell lines are susceptible to lysis by natural killer cells but not by cytotoxic T lymphocytes. J Immunol. 1985;135:242–246. [PubMed] [Google Scholar]
- 41.Reid GS, Shan X, Coughlin CM, Lassoued W, Pawel BR, Wexler LH, Thiele CJ, Tsokos M, Pinkus JL, Pinkus GS, et al. Interferon-gamma-dependent infiltration of human T cells into neuroblastoma tumors in vivo. Clin Cancer Res. 2009;15:6602–6608. doi: 10.1158/1078-0432.CCR-09-0829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Raffaghello L, Prigione I, Airoldi I, Camoriano M, Morandi F, Bocca P, Gambini C, Ferrone S, Pistoia V. Mechanisms of immune evasion of human neuroblastoma. Cancer Lett. 2005;228:155–161. doi: 10.1016/j.canlet.2004.11.064. [DOI] [PubMed] [Google Scholar]
- 43.Raffaghello L, Prigione I, Bocca P, Morandi F, Camoriano M, Gambini C, Wang X, Ferrone S, Pistoia V. Multiple defects of the antigen-processing machinery components in human neuroblastoma: immunotherapeutic implications. Oncogene. 2005;24:4634–4644. doi: 10.1038/sj.onc.1208594. [DOI] [PubMed] [Google Scholar]
- 44.Raffaghello L, Prigione I, Airoldi I, Camoriano M, Levreri I, Gambini C, Pende D, Steinle A, Ferrone S, Pistoia V. Downregulation and/or release of NKG2D ligands as immune evasion strategy of human neuroblastoma. Neoplasia. 2004;6:558–568. doi: 10.1593/neo.04316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Castriconi R, Dondero A, Augugliaro R, Cantoni C, Carnemolla B, Sementa AR, Negri F, Conte R, Corrias MV, Moretta L, et al. Identification of 4Ig-B7-H3 as a neuroblastoma-associated molecule that exerts a protective role from an NK cellmediated lysis. Proc Natl Acad Sci U S A. 2004;101:12640–12645. doi: 10.1073/pnas.0405025101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Leitner J, Klauser C, Pickl WF, Stockl J, Majdic O, Bardet AF, Kreil DP, Dong C, Yamazaki T, Zlabinger G, et al. B7-H3 is a potent inhibitor of human T-cell activation: No evidence for B7-H3 and TREML2 interaction. Eur J Immunol. 2009;39:1754–1764. doi: 10.1002/eji.200839028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Morandi F, Levreri I, Bocca P, Galleni B, Raffaghello L, Ferrone S, Prigione I, Pistoia V. Human neuroblastoma cells trigger an immunosuppressive program in monocytes by stimulating soluble HLA-G release. Cancer Res. 2007;67:6433–6441. doi: 10.1158/0008-5472.CAN-06-4588. [DOI] [PubMed] [Google Scholar]
- 48.Pistoia V, Morandi F, Wang X, Ferrone S. Soluble HLA-G: Are they clinically relevant? Semin Cancer Biol. 2007;17:469–479. doi: 10.1016/j.semcancer.2007.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sica A, Larghi P, Mancino A, Rubino L, Porta C, Totaro MG, Rimoldi M, Biswas SK, Allavena P, Mantovani A. Macrophage polarization in tumour progression. Semin Cancer Biol. 2008;18:349–355. doi: 10.1016/j.semcancer.2008.03.004. [DOI] [PubMed] [Google Scholar]
- 50.Metelitsa LS. Anti-tumor potential of type-I NKT cells against CD1d-positive and CD1d-negative tumors in humans. Clin Immunol. 2011;140:119–129. doi: 10.1016/j.clim.2010.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Metelitsa LS, Naidenko OV, Kant A, Wu HW, Loza MJ, Perussia B, Kronenberg M, Seeger RC. Human NKT cells mediate antitumor cytotoxicity directly by recognizing target cell CD1d with bound ligand or indirectly by producing IL-2 to activate NK cells. J Immunol. 2001;167:3114–3122. doi: 10.4049/jimmunol.167.6.3114. [DOI] [PubMed] [Google Scholar]
- 52.Metelitsa LS, Wu HW, Wang H, Yang Y, Warsi Z, Asgharzadeh S, Groshen S, Wilson SB, Seeger RC. Natural killer T cells infiltrate neuroblastomas expressing the chemokine CCL2. Journal of Experimental Medicine. 2004;199:1213–1221. doi: 10.1084/jem.20031462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Song L, Ara T, Wu HW, Woo CW, Reynolds CP, Seeger RC, DeClerck YA, Thiele CJ, Sposto R, Metelitsa LS. Oncogene MYCN regulates localization of NKT cells to the site of disease in neuroblastoma. J.Clin.Invest. 2007;117:2702–2712. doi: 10.1172/JCI30751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Song L, Asgharzadeh S, Salo J, Engell K, Wu HW, Sposto R, Ara T, Silverman AM, DeClerck YA, Seeger RC, et al. Valpha24-invariant NKT cells mediate antitumor activity via killing of tumor-associated macrophages. J Clin Invest. 2009;119:1524–1536. doi: 10.1172/JCI37869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Coca S, Perez-Piqueras J, Martinez D, Colmenarejo A, Saez MA, Vallejo C, Martos JA, Moreno M. The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer. 1997;79:2320–2328. doi: 10.1002/(sici)1097-0142(19970615)79:12<2320::aid-cncr5>3.0.co;2-p. [DOI] [PubMed] [Google Scholar]
- 56.Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Che X, Iwashige H, Aridome K, Hokita S, Aikou T. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer. 2000;88:577–583. [PubMed] [Google Scholar]
- 57.Villegas FR, Coca S, Villarrubia VG, Jimenez R, Chillon MJ, Jareno J, Zuil M, Callol L. Prognostic significance of tumor infiltrating natural killer cells subset CD57 in patients with squamous cell lung cancer. Lung Cancer. 2002;35:23–28. doi: 10.1016/s0169-5002(01)00292-6. [DOI] [PubMed] [Google Scholar]
- 58.Menon AG, Janssen-van Rhijn CM, Morreau H, Putter H, Tollenaar RA, van de Velde CJ, Fleuren GJ, Kuppen PJ. Immune system and prognosis in colorectal cancer: a detailed immunohistochemical analysis. Lab Invest. 2004;84:493–501. doi: 10.1038/labinvest.3700055. [DOI] [PubMed] [Google Scholar]
- 59.Dall'Ozzo S, Tartas S, Paintaud G, Cartron G, Colombat P, Bardos P, Watier H, Thibault G. Rituximab-dependent cytotoxicity by natural killer cells: influence of FCGR3A polymorphism on the concentration-effect relationship. Cancer Res. 2004;64:4664–4669. doi: 10.1158/0008-5472.CAN-03-2862. [DOI] [PubMed] [Google Scholar]
- 60.Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, Watier H. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood. 2002;99:754–758. doi: 10.1182/blood.v99.3.754. [DOI] [PubMed] [Google Scholar]
- 61.Treon SP, Hansen M, Branagan AR, Verselis S, Emmanouilides C, Kimby E, Frankel SR, Touroutoglou N, Turnbull B, Anderson KC, et al. Polymorphisms in FcgammaRIIIA (CD16) receptor expression are associated with clinical response to rituximab in Waldenstrom's macroglobulinemia. J Clin Oncol. 2005;23:474–481. doi: 10.1200/JCO.2005.06.059. [DOI] [PubMed] [Google Scholar]
- 62.Handgretinger R, Kimmig A, Lang P, Daurer B, Kuci S, Bruchelt G, Treuner J, Niethammer D. Interferon-gamma upregulates the susceptibility of human neuroblastoma cells to interleukin-2-activated natural killer cells. Nat Immun Cell Growth Regul. 1989;8:189–196. [PubMed] [Google Scholar]
- 63.Foreman NK, Rill DR, Coustan-Smith E, Douglass EC, Brenner MK. Mechanisms of selective killing of neuroblastoma cells by natural killer cells and lymphokine activated killer cells. Potential for residual disease eradication. Br J Cancer. 1993;67:933–938. doi: 10.1038/bjc.1993.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Rossi AR, Pericle F, Rashleigh S, Janiec J, Djeu JY. Lysis of neuroblastoma cell lines by human natural killer cells activated by interleukin-2 and interleukin-12. Blood. 1994;83:1323–1328. [PubMed] [Google Scholar]
- 65.Venstrom JM, Zheng J, Noor N, Danis KE, Yeh AW, Cheung IY, Dupont B, O'Reilly RJ, Cheung NK, Hsu KC. KIR and HLA genotypes are associated with disease progression and survival following autologous hematopoietic stem cell transplantation for high-risk neuroblastoma. Clin Cancer Res. 2009;15:7330–7334. doi: 10.1158/1078-0432.CCR-09-1720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Delgado DC, Hank JA, Kolesar J, Lorentzen D, Gan J, Seo S, Kim K, Shusterman S, Gillies SD, Reisfeld RA, et al. Genotypes of NK cell KIR receptors, their ligands, and Fcgamma receptors in the response of neuroblastoma patients to Hu14.18-IL2 immunotherapy. Cancer Res. 2010;70:9554–9561. doi: 10.1158/0008-5472.CAN-10-2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Andre P, Castriconi R, Espeli M, Anfossi N, Juarez T, Hue S, Conway H, Romagne F, Dondero A, Nanni M, et al. Comparative analysis of human NK cell activation induced by NKG2D and natural cytotoxicity receptors. Eur J Immunol. 2004;34:961–971. doi: 10.1002/eji.200324705. [DOI] [PubMed] [Google Scholar]
- 68.Moretta L, Moretta A. Unravelling natural killer cell function: triggering and inhibitory human NK receptors. EMBO J. 2004;23:255–259. doi: 10.1038/sj.emboj.7600019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lanier LL. Natural killer cell receptor signaling. Curr Opin Immunol. 2003;15:308–314. doi: 10.1016/s0952-7915(03)00039-6. [DOI] [PubMed] [Google Scholar]
- 70.Sivori S, Parolini S, Marcenaro E, Castriconi R, Pende D, Millo R, Moretta A. Involvement of natural cytotoxicity receptors in human natural killer cellmediated lysis of neuroblastoma and glioblastoma cell lines. J Neuroimmunol. 2000;107:220–225. doi: 10.1016/s0165-5728(00)00221-6. [DOI] [PubMed] [Google Scholar]
- 71.Castriconi R, Dondero A, Corrias MV, Lanino E, Pende D, Moretta L, Bottino C, Moretta A. Natural killer cell-mediated killing of freshly isolated neuroblastoma cells: critical role of DNAX accessory molecule-1-poliovirus receptor interaction. Cancer Res. 2004;64:9180–9184. doi: 10.1158/0008-5472.CAN-04-2682. [DOI] [PubMed] [Google Scholar]
- 72.Truitt RL, Piaskowski V, Kirchner P, McOlash L, Camitta BM, Casper JT. Immunological evaluation of pediatric cancer patients receiving recombinant interleukin-2 in a phase I trial. J Immunother (1991) 1992;11:274–285. doi: 10.1097/00002371-199205000-00006. [DOI] [PubMed] [Google Scholar]
- 73.Bauer M, Reaman GH, Hank JA, Cairo MS, Anderson P, Blazar BR, Frierdich S, Sondel PM. A phase II trial of human recombinant interleukin-2 administered as a 4-day continuous infusion for children with refractory neuroblastoma, non-Hodgkin's lymphoma, sarcoma, renal cell carcinoma, and malignant melanoma. A Childrens Cancer Group study. Cancer. 1995;75:2959–2965. doi: 10.1002/1097-0142(19950615)75:12<2959::aid-cncr2820751225>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
- 74.Marti F, Pardo N, Peiro M, Bertran E, Amill B, Garcia J, Cubells J, Rueda F. Progression of natural immunity during one-year treatment of residual disease in neuroblastoma patients with high doses of interleukin-2 after autologous bone marrow transplantation. Exp Hematol. 1995;23:1445–1452. [PubMed] [Google Scholar]
- 75.Bartlett JB, Dredge K, Dalgleish AG. The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat Rev Cancer. 2004;4:314–322. doi: 10.1038/nrc1323. [DOI] [PubMed] [Google Scholar]
- 76.Hayashi T, Hideshima T, Akiyama M, Podar K, Yasui H, Raje N, Kumar S, Chauhan D, Treon SP, Richardson P, et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. Br J Haematol. 2005;128:192–203. doi: 10.1111/j.1365-2141.2004.05286.x. [DOI] [PubMed] [Google Scholar]
- 77.Berg SL, Cairo MS, Russell H, Ayello J, Ingle AM, Lau H, Chen N, Adamson PC, Blaney SM. Safety, pharmacokinetics, and immunomodulatory effects of lenalidomide in children and adolescents with relapsed/refractory solid tumors or myelodysplastic syndrome: a Children's Oncology Group Phase I Consortium report. J Clin Oncol. 2011;29:316–323. doi: 10.1200/JCO.2010.30.8387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bartlett JB, Michael A, Clarke IA, Dredge K, Nicholson S, Kristeleit H, Polychronis A, Pandha H, Muller GW, Stirling DI, et al. Phase I study to determine the safety, tolerability and immunostimulatory activity of thalidomide analogue CC-5013 in patients with metastatic malignant melanoma and other advanced cancers. Br J Cancer. 2004;90:955–961. doi: 10.1038/sj.bjc.6601579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–497. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
- 80.Maloney DG, Grillo-Lopez AJ, White CA, Bodkin D, Schilder RJ, Neidhart JA, Janakiraman N, Foon KA, Liles TM, Dallaire BK, et al. IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin's lymphoma. Blood. 1997;90:2188–2195. [PubMed] [Google Scholar]
- 81.Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, Bets D, Mueser M, Harstrick A, Verslype C, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–345. doi: 10.1056/NEJMoa033025. [DOI] [PubMed] [Google Scholar]
- 82.Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE, Jr, Davidson NE, Tan-Chiu E, Martino S, Paik S, Kaufman PA, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–1684. doi: 10.1056/NEJMoa052122. [DOI] [PubMed] [Google Scholar]
- 83.Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659–1672. doi: 10.1056/NEJMoa052306. [DOI] [PubMed] [Google Scholar]
- 84.Kennett RH, Gilbert F. Hybrid myelomas producing antibodies against a human neuroblastoma antigen present on fetal brain. Science. 1979;203:1120–1121. doi: 10.1126/science.424740. [DOI] [PubMed] [Google Scholar]
- 85.Seeger RC, Rosenblatt HM, Imai K, Ferrone S. Common antigenic determinants on human melanoma, glioma, neuroblastoma, and sarcoma cells defined with monoclonal antibodies. Cancer Res. 1981;41:2714–2717. [PubMed] [Google Scholar]
- 86.Howe AJ, Seeger RC, Molinaro GA, Ferrone S. Analysis of human tumor cells for Ia-like antigens with monoclonal antibodies. J.Natl.Cancer Inst. 1981;66:827–829. [PubMed] [Google Scholar]
- 87.Seeger RC, Danon YL, Rayner SA, Hoover F. Definition of a Thy-1 determinant on human neuroblastoma, glioma, sarcoma, and teratoma cells with a monoclonal antibody. J Immunol. 1982;128:983–989. [PubMed] [Google Scholar]
- 88.Schulz G, Cheresh DA, Varki NM, Yu A, Staffileno LK, Reisfeld RA. Detection of ganglioside GD2 in tumor tissues and sera of neuroblastoma patients. Cancer Res. 1984;44:5914–5920. [PubMed] [Google Scholar]
- 89.Honsik CJ, Jung G, Reisfeld RA. Lymphokine-activated killer cells targeted by monoclonal antibodies to the disialogangliosides GD2 and GD3 specifically lyse human tumor cells of neuroectodermal origin. Proc Natl Acad Sci U S A. 1986;83:7893–7897. doi: 10.1073/pnas.83.20.7893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Mujoo K, Spiro RC, Reisfeld RA. Characterization of a unique glycoprotein antigen expressed on the surface of human neuroblastoma cells. J Biol Chem. 1986;261:10299–10305. [PubMed] [Google Scholar]
- 91.Mujoo K, Cheresh DA, Yang HM, Reisfeld RA. Disialoganglioside GD2 on human neuroblastoma cells: target antigen for monoclonal antibody-mediated cytolysis and suppression of tumor growth. Cancer Res. 1987;47:1098–1104. [PubMed] [Google Scholar]
- 92.Wolff JM, Frank R, Mujoo K, Spiro RC, Reisfeld RA, Rathjen FG. A human brain glycoprotein related to the mouse cell adhesion molecule L1. J Biol Chem. 1988;263:11943–11947. [PubMed] [Google Scholar]
- 93.Cheung NK, Saarinen UM, Neely JE, Landmeier B, Donovan D, Coccia PF. Monoclonal antibodies to a glycolipid antigen on human neuroblastoma cells. Cancer Res. 1985;45:2642–2649. [PubMed] [Google Scholar]
- 94.Saito M, Yu RK, Cheung NK. Ganglioside GD2 specificity of monoclonal antibodies to human neuroblastoma cell. Biochem Biophys Res Commun. 1985;127:1–7. doi: 10.1016/s0006-291x(85)80117-0. [DOI] [PubMed] [Google Scholar]
- 95.Cheung NK, Lazarus H, Miraldi FD, Abramowsky CR, Kallick S, Saarinen UM, Spitzer T, Strandjord SE, Coccia PF, Berger NA. Ganglioside GD2 specific monoclonal antibody 3F8: a phase I study in patients with neuroblastoma and malignant melanoma. J Clin Oncol. 1987;5:1430–1440. doi: 10.1200/JCO.1987.5.9.1430. [DOI] [PubMed] [Google Scholar]
- 96.Moss TJ, Reynolds CP, Sather HN, Romansky SG, Hammond GD, Seeger RC. Prognostic value of immunocytologic detection of bone marrow metastases in neuroblastoma. N.Engl.J.Med. 1991;324:219–226. doi: 10.1056/NEJM199101243240403. [DOI] [PubMed] [Google Scholar]
- 97.Beiske K, Burchill SA, Cheung IY, Hiyama E, Seeger RC, Cohn SL, Pearson AD, Matthay KK. Consensus criteria for sensitive detection of minimal neuroblastoma cells in bone marrow, blood and stem cell preparations by immunocytology and QRT-PCR: recommendations by the International Neuroblastoma Risk Group Task Force. Br J Cancer. 2009;100:1627–1637. doi: 10.1038/sj.bjc.6605029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Seeger RC, Reynolds CP, Gallego R, Stram DO, Gerbing RB, Matthay KK. Quantitative tumor cell content of bone marrow and blood as a predictor of outcome in stage IV neuroblastoma: a Children's Cancer Group Study. J.Clin.Oncol. 2000;18:4067–4076. doi: 10.1200/JCO.2000.18.24.4067. [DOI] [PubMed] [Google Scholar]
- 99.Rill DR, Santana VM, Roberts WM, Nilson T, Bowman LC, Krance RA, Heslop HE, Moen RC, Ihle JN, Brenner MK. Direct demonstration that autologous bone marrow transplantation for solid tumors can return a multiplicity of tumorigenic cells. Blood. 1994;84:380–383. [PubMed] [Google Scholar]
- 100.Reynolds CP, Seeger RC, Vo DD, Black AT, Wells J, Ugelstad J. Model system for removing neuroblastoma cells from bone marrow using monoclonal antibodies and magnetic immunobeads. Cancer Res. 1986;46:5882–5886. [PubMed] [Google Scholar]
- 101.Ladisch S, Wu ZL. Detection of a tumour-associated ganglioside in plasma of patients with neuroblastoma. Lancet. 1985;1:136–138. doi: 10.1016/s0140-6736(85)91906-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wu ZL, Schwartz E, Seeger R, Ladisch S. Expression of GD2 ganglioside by untreated primary human neuroblastomas. Cancer Res. 1986;46:440–443. [PubMed] [Google Scholar]
- 103.Handgretinger R, Baader P, Dopfer R, Klingebiel T, Reuland P, Treuner J, Reisfeld RA, Niethammer D. A phase I study of neuroblastoma with the anti-ganglioside GD2 antibody 14.G2a. Cancer Immunol Immunother. 1992;35:199–204. doi: 10.1007/BF01756188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Murray JL, Cunningham JE, Brewer H, Mujoo K, Zukiwski AA, Podoloff DA, Kasi LP, Bhadkamkar V, Fritsche HA, Benjamin RS, et al. Phase I trial of murine monoclonal antibody 14G2a administered by prolonged intravenous infusion in patients with neuroectodermal tumors. J Clin Oncol. 1994;12:184–193. doi: 10.1200/JCO.1994.12.1.184. [DOI] [PubMed] [Google Scholar]
- 105.Handgretinger R, Anderson K, Lang P, Dopfer R, Klingebiel T, Schrappe M, Reuland P, Gillies SD, Reisfeld RA, Neithammer D. A phase I study of human/mouse chimeric antiganglioside GD2 antibody ch14.18 in patients with neuroblastoma. Eur J Cancer. 1995;31A:261–267. doi: 10.1016/0959-8049(94)00413-y. [DOI] [PubMed] [Google Scholar]
- 106.Kushner BH, Cheung NK. Clinically effective monoclonal antibody 3F8 mediates nonoxidative lysis of human neuroectodermal tumor cells by polymorphonuclear leukocytes. Cancer Res. 1991;51:4865–4870. [PubMed] [Google Scholar]
- 107.Barker E, Mueller BM, Handgretinger R, Herter M, Yu AL, Reisfeld RA. Effect of a chimeric anti-ganglioside GD2 antibody on cell-mediated lysis of human neuroblastoma cells. Cancer Res. 1991;51:144–149. [PubMed] [Google Scholar]
- 108.Bruchelt G, Handgretinger R, Fierlbeck G, Kimmig A, Dopfer R, Reisfeld RA, Treuner J, Niethammer D. Lysis of neuroblastoma cells by the ADCCreaction: granulocytes of patients with chronic granulomatous disease are more effective than those of healthy donors. Immunol Lett. 1989;22:217–220. doi: 10.1016/0165-2478(89)90194-6. [DOI] [PubMed] [Google Scholar]
- 109.Hank JA, Robinson RR, Surfus J, Mueller BM, Reisfeld RA, Cheung NK, Sondel PM. Augmentation of antibody dependent cell mediated cytotoxicity following in vivo therapy with recombinant interleukin 2. Cancer Res. 1990;50:5234–5239. [PubMed] [Google Scholar]
- 110.Kushner BH, Cheung NK. GM-CSF enhances 3F8 monoclonal antibodydependent cellular cytotoxicity against human melanoma and neuroblastoma. Blood. 1989;73:1936–1941. [PubMed] [Google Scholar]
- 111.Barker E, Reisfeld RA. A mechanism for neutrophil-mediated lysis of human neuroblastoma cells. Cancer Res. 1993;53:362–367. [PubMed] [Google Scholar]
- 112.Metelitsa LS, Gillies SD, Super M, Shimada H, Reynolds CP, Seeger RC. Antidisialoganglioside/granulocyte macrophage-colony-stimulating factor fusion protein facilitates neutrophil antibody-dependent cellular cytotoxicity and depends on FcgammaRII (CD32) and Mac-1 (CD11b/CD18) for enhanced effector cell adhesion and azurophil granule exocytosis. Blood. 2002;99:4166–4173. doi: 10.1182/blood.v99.11.4166. [DOI] [PubMed] [Google Scholar]
- 113.Hank JA, Surfus J, Gan J, Chew TL, Hong R, Tans K, Reisfeld R, Seeger RC, Reynolds CP, Bauer M, et al. Treatment of neuroblastoma patients with antiganglioside GD2 antibody plus interleukin-2 induces antibody-dependent cellular cytotoxicity against neuroblastoma detected in vitro. J Immunother Emphasis Tumor Immunol. 1994;15:29–37. doi: 10.1097/00002371-199401000-00004. [DOI] [PubMed] [Google Scholar]
- 114.Frost JD, Hank JA, Reaman GH, Frierdich S, Seeger RC, Gan J, Anderson PM, Ettinger LJ, Cairo MS, Blazar BR, et al. A phase I/IB trial of murine monoclonal anti-GD2 antibody 14.G2a plus interleukin-2 in children with refractory neuroblastoma: a report of the Children's Cancer Group. Cancer. 1997;80:317–333. doi: 10.1002/(sici)1097-0142(19970715)80:2<317::aid-cncr21>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
- 115.Yu AL, Uttenreuther-Fischer MM, Huang CS, Tsui CC, Gillies SD, Reisfeld RA, Kung FH. Phase I trial of a human-mouse chimeric antidisialoganglioside monoclonal antibody ch14.18 in patients with refractory neuroblastoma and osteosarcoma. J Clin Oncol. 1998;16:2169–2180. doi: 10.1200/JCO.1998.16.6.2169. [DOI] [PubMed] [Google Scholar]
- 116.Ozkaynak MF, Sondel PM, Krailo MD, Gan J, Javorsky B, Reisfeld RA, Matthay KK, Reaman GH, Seeger RC. Phase I study of chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocytemacrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-cell transplantation: a Children's Cancer Group Study. J Clin Oncol. 2000;18:4077–4085. doi: 10.1200/JCO.2000.18.24.4077. [DOI] [PubMed] [Google Scholar]
- 117.Gilman AL, Ozkaynak MF, Matthay KK, Krailo M, Yu AL, Gan J, Sternberg A, Hank JA, Seeger R, Reaman GH, et al. Phase I study of ch14.18 with granulocyte-macrophage colony-stimulating factor and interleukin-2 in children with neuroblastoma after autologous bone marrow transplantation or stem-cell rescue: a report from the Children's Oncology Group. J Clin Oncol. 2009;27:85–91. doi: 10.1200/JCO.2006.10.3564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sabzevari H, Gillies SD, Mueller BM, Pancook JD, Reisfeld RA. A recombinant antibody-interleukin 2 fusion protein suppresses growth of hepatic human neuroblastoma metastases in severe combined immunodeficiency mice. Proc Natl Acad Sci U S A. 1994;91:9626–9630. doi: 10.1073/pnas.91.20.9626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Pancook JD, Becker JC, Gillies SD, Reisfeld RA. Eradication of established hepatic human neuroblastoma metastases in mice with severe combined immunodeficiency by antibody-targeted interleukin-2. Cancer Immunol Immunother. 1996;42:88–92. doi: 10.1007/s002620050256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Becker JC, Varki N, Gillies SD, Furukawa K, Reisfeld RA. Long-lived and transferable tumor immunity in mice after targeted interleukin-2 therapy. J Clin Invest. 1996;98:2801–2804. doi: 10.1172/JCI119107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Osenga KL, Hank JA, Albertini MR, Gan J, Sternberg AG, Eickhoff J, Seeger RC, Matthay KK, Reynolds CP, Twist C, et al. A phase I clinical trial of the hu14.18-IL2 (EMD 273063) as a treatment for children with refractory or recurrent neuroblastoma and melanoma: a study of the Children's Oncology Group. Clin Cancer Res. 2006;12:1750–1759. doi: 10.1158/1078-0432.CCR-05-2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Shusterman S, London WB, Gillies SD, Hank JA, Voss SD, Seeger RC, Reynolds CP, Kimball J, Albertini MR, Wagner B, et al. Antitumor activity of hu14.18-IL2 in patients with relapsed/refractory neuroblastoma: a Children's Oncology Group (COG) phase II study. J Clin Oncol. 2010;28:4969–4975. doi: 10.1200/JCO.2009.27.8861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hernandez-Ilizaliturri FJ, Reddy N, Holkova B, Ottman E, Czuczman MS. Immunomodulatory drug CC-5013 or CC-4047 and rituximab enhance antitumor activity in a severe combined immunodeficient mouse lymphoma model. Clin.Cancer Res. 2005;11:5984–5992. doi: 10.1158/1078-0432.CCR-05-0577. [DOI] [PubMed] [Google Scholar]
- 124.Wu L, Adams M, Carter T, Chen R, Muller G, Stirling D, Schafer P, Bartlett JB. lenalidomide enhances natural killer cell and monocyte-mediated antibodydependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells. Clin.Cancer Res. 2008;14:4650–4657. doi: 10.1158/1078-0432.CCR-07-4405. [DOI] [PubMed] [Google Scholar]
- 125.Zhang L, Qian Z, Cai Z, Sun L, Wang H, Bartlett JB, Yi Q, Wang M. Synergistic antitumor effects of lenalidomide and rituximab on mantle cell lymphoma in vitro and in vivo. Am.J.Hematol. 2009 doi: 10.1002/ajh.21468. [DOI] [PubMed] [Google Scholar]
- 126.Tai YT, Li XF, Catley L, Coffey R, Breitkreutz I, Bae J, Song W, Podar K, Hideshima T, Chauhan D, et al. Immunomodulatory drug lenalidomide (CC- 5013, IMiD3) augments anti-CD40 SGN-40-induced cytotoxicity in human multiple myeloma: clinical implications. Cancer Res. 2005;65:11712–11720. doi: 10.1158/0008-5472.CAN-05-1657. [DOI] [PubMed] [Google Scholar]
- 127.Lapalombella R, Gowda A, Joshi T, Mehter N, Cheney C, Lehman A, Chen CS, Johnson AJ, Caligiuri MA, Tridandapani S, et al. The humanized CD40 antibody SGN-40 demonstrates pre-clinical activity that is enhanced by lenalidomide in chronic lymphocytic leukaemia. Br J Haematol. 2009;144:848–855. doi: 10.1111/j.1365-2141.2008.07548.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Wu L, Parton A, Lu L, Adams M, Schafer P, Bartlett JB. Lenalidomide enhances antibody-dependent cellular cytotoxicity of solid tumor cells in vitro: influence of host immune and tumor markers. Cancer Immunol Immunother. 2011;60:61–73. doi: 10.1007/s00262-010-0919-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Sadelain M. T-cell engineering for cancer immunotherapy. Cancer J. 2009;15:451–455. doi: 10.1097/PPO.0b013e3181c51f37. [DOI] [PubMed] [Google Scholar]
- 130.Krause A, Guo HF, Latouche JB, Tan C, Cheung NK, Sadelain M. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med. 1998;188:619–626. doi: 10.1084/jem.188.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Rossig C, Bollard CM, Nuchtern JG, Merchant DA, Brenner MK. Targeting of G(D2)-positive tumor cells by human T lymphocytes engineered to express chimeric T-cell receptor genes. Int J Cancer. 2001;94:228–236. doi: 10.1002/ijc.1457. [DOI] [PubMed] [Google Scholar]
- 132.Rossig C, Bollard CM, Nuchtern JG, Rooney CM, Brenner MK. Epstein-Barr virus-specific human T lymphocytes expressing antitumor chimeric T-cell receptors: potential for improved immunotherapy. Blood. 2002;99:2009–2016. doi: 10.1182/blood.v99.6.2009. [DOI] [PubMed] [Google Scholar]
- 133.Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, Huls MH, Liu E, Gee AP, Mei Z, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–1270. doi: 10.1038/nm.1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Gonzalez S, Naranjo A, Serrano LM, Chang WC, Wright CL, Jensen MC. Genetic engineering of cytolytic T lymphocytes for adoptive T-cell therapy of neuroblastoma. J.Gene Med. 2004;6:704–711. doi: 10.1002/jgm.489. [DOI] [PubMed] [Google Scholar]
- 135.Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet HB, Bautista C, Chang WC, Ostberg JR, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–833. doi: 10.1038/sj.mt.6300104. [DOI] [PubMed] [Google Scholar]
- 136.Craddock JA, Lu A, Bear A, Pule M, Brenner MK, Rooney CM, Foster AE. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J Immunother. 2010;33:780–788. doi: 10.1097/CJI.0b013e3181ee6675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Sun J, Dotti G, Huye LE, Foster AE, Savoldo B, Gramatges MM, Spencer DM, Rooney CM. T cells expressing constitutively active Akt resist multiple tumor-associated inhibitory mechanisms. Mol Ther. 2010;18:2006–2017. doi: 10.1038/mt.2010.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, McKenna D, Le C, Defor TE, Burns LJ, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051–3057. doi: 10.1182/blood-2004-07-2974. [DOI] [PubMed] [Google Scholar]
- 139.Rubnitz JE, Inaba H, Ribeiro RC, Pounds S, Rooney B, Bell T, Pui CH, Leung W. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol. 2010;28:955–959. doi: 10.1200/JCO.2009.24.4590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Somanchi SS, Senyukov VV, Denman CJ, Lee DA. Expansion, purification, and functional assessment of human peripheral blood NK cells. J Vis Exp. 2011 doi: 10.3791/2540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–896. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]
- 142.Mantovani A. B cells and macrophages in cancer: yin and yang. Nat Med. 2011;17:285–286. doi: 10.1038/nm0311-285. [DOI] [PubMed] [Google Scholar]
- 143.Asgharzadeh S, Pique-Regi R, Sposto R, Wang H, Yang Y, Shimada H, Matthay K, Buckley J, Ortega A, Seeger RC. Prognostic significance of gene expression profiles of metastatic neuroblastomas lacking MYCN gene amplification. J.Natl.Cancer Inst. 2006;98:1193–1203. doi: 10.1093/jnci/djj330. [DOI] [PubMed] [Google Scholar]
- 144.Trotta R, Col JD, Yu J, Ciarlariello D, Thomas B, Zhang X, Allard J, 2nd, Wei M, Mao H, Byrd JC, et al. TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol. 2008;181:3784–3792. doi: 10.4049/jimmunol.181.6.3784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Bellone G, Aste-Amezaga M, Trinchieri G, Rodeck U. Regulation of NK cell functions by TGF-beta 1. J Immunol. 1995;155:1066–1073. [PubMed] [Google Scholar]
- 146.Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar S, Korets L, Lam J, Tawfik D, DeNardo DG, et al. FcRgamma activation regulates inflammationassociated squamous carcinogenesis. Cancer Cell. 2010;17:121–134. doi: 10.1016/j.ccr.2009.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.DeNardo DG, Andreu P, Coussens LM. Interactions between lymphocytes and myeloid cells regulate pro- versus anti-tumor immunity. Cancer Metastasis Rev. 2010;29:309–316. doi: 10.1007/s10555-010-9223-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Johnson BD, Jing W, Orentas RJ. CD25+ regulatory T cell inhibition enhances vaccine-induced immunity to neuroblastoma. J Immunother. 2007;30:203–214. doi: 10.1097/01.cji.0000211336.91513.dd. [DOI] [PubMed] [Google Scholar]
- 149.Croce M, Corrias MV, Orengo AM, Brizzolara A, Carlini B, Borghi M, Rigo V, Pistoia V, Ferrini S. Transient depletion of CD4(+) T cells augments IL-21- based immunotherapy of disseminated neuroblastoma in syngeneic mice. Int J Cancer. 2010;127:1141–1150. doi: 10.1002/ijc.25140. [DOI] [PubMed] [Google Scholar]
- 150.Jing W, Yan X, Hallett WH, Gershan JA, Johnson BD. Depletion of CD25+ T cells from hematopoietic stem cell grafts increases posttransplantation vaccine-induced immunity to neuroblastoma. Blood. 2011;117:6952–6962. doi: 10.1182/blood-2010-12-326108. [DOI] [PMC free article] [PubMed] [Google Scholar]