116
Views
36
CrossRef citations to date
0
Altmetric
Review

Cancer testis antigen and immunotherapy

, &
Pages 11-19 | Published online: 18 Apr 2013

Abstract

The identification of cancer testis (CT) antigens has been an important advance in determining potential targets for cancer immunotherapy. Multiple previous studies have shown that CT antigen vaccines, using both peptides and dendritic cell vaccines, can elicit clinical and immunologic responses in several different tumors. This review details the expression of melanoma antigen family A, 1 (MAGE-A1), melanoma antigen family A, 3 (MAGE-A3), and New York esophageal squamous cell carcinoma-1 (NY-ESO-1) in various malignancies, and presents our current understanding of CT antigen based immunotherapy.

Introduction

The past two decades have witnessed major strides in the treatment of several pediatric and adult cancers, particularly with the use of multiagent chemotherapy, radiation therapy, and recently, monoclonal antibodies. Nevertheless, a subset of these patients will develop resistance to these modalities, leaving few treatment options with curative potential. In addition, patients with high risk metastatic disease continue to have dismal treatment outcomes, despite these advances. Therefore, for patients with relapsed, therapy refractory disease and tumors at high risk for recurrence, new treatment strategies are desperately needed.

Over the past two decades numerous groups have investigated immune-based therapies for patients with relapsed cancer. The success in using adoptive cellular immunotherapy to fight viral infections following allogeneic stem cell transplantation has encouraged some groups to focus their efforts on the infusion of cancer antigen specific, or otherwise activated, T lymphocytes.Citation1,Citation2 There is a long history of clinical investigation with cancer vaccines for a variety of malignant solid tumors. The recognition that dendritic cells (DC) play a key role in antigen presentation led to several groups using DC pulsed with cancer relevant antigens, while other groups have used whole tumor antigens or human leukocyte antigen (HLA) restricted epitopes.Citation3,Citation4 Several different antigens have been targeted in these strategies, most notably the cancer testis (CT) antigens. These tumor proteins are of interest since they are expressed on several malignant solid tumors, as well as some leukemias, and have a restricted pattern of expression, thereby limiting the possibility of an immune response directed against normal host tissues. These antigens can also be epigenetically upregulated on tumors following exposure to demethylating chemotherapy agents, potentially making tumors more susceptible to killing by antigen-specific T cells that have been stimulated following a CT antigen vaccine. In this review we will summarize past studies which target these antigens and future directions in CT antigen-based immunotherapy.

Cancer immunotherapy

An improved understanding of cellular immunology has helped to facilitate the rational design of cancer immunotherapy strategies. While conventional therapy such as chemotherapy and radiation are useful for the majority of patients, the use of these modalities alone may be insufficient for patients with relapsed cancer or for those who initially present with advanced disease. Chemotherapy often has limited efficacy in patients with relapsed disease, for whom intensification of conventional therapy to overcome drug resistance can lead to significant morbidity.

Immunotherapy can specifically target, or in general, modulate cellular immune responses against cancer proteins and has the potential to provide long-lasting responses. Adoptive transfer of autologous in vitro generated and expanded effector T cells is one such effective method. Initial studies in adoptive immunotherapy were performed in the allogeneic stem cell transplant setting to fight serious, potentially life-threatening viral infections, such as cytomegalovirus and Epstein–Barr virus. While adoptive immunotherapy has been largely successful against several viral infectionsCitation5Citation7 this approach has had limited success against cancer. The precursor frequency of cancer antigen-specific cells is very low, and the expansion of these cells requires multiple stimulations. In addition, the low avidity of expanded T cells against cancer antigens and the short life span of adoptively transferred effector T cells are practical limitations of adoptive immunotherapy. Several strategies have been developed to overcome these challenges, such as the use of chimeric antigen receptors,Citation8 T cells genetically engineered to express T cell receptors (TCRs) with high affinity and specificity,Citation9,Citation10 and bispecific antibodies to promote T cell recognition of tumors.Citation11

Several immune evasion mechanisms pose major obstacles for the practical application of immunotherapy against cancer. Tumor cells can evade the immune system by (a) downregulating the expression of major histocompatibility complex (MHC) class I and class II molecules that are required for antigen presentation to T cells; (b) downregulating costimulatory molecules, such as CD80 and CD86, which are required for optimal activation of T cells; (c) upregulating coinhibitory molecules, such as cytotoxic T-lymphocyte antigen 4 (CTLA-4) ligands and programmed cell death ligand 1 (PDL-1), on tumor cells;Citation10 and (d) recruiting regulatory T cells (Tregs) that produce immunosuppressive cytokines at the tumor site. For example, high expression of CTLA-4 has been correlated with increased T cell dysfunction in melanoma patients.Citation12 CTLA-4 and programmed cell death 1 (PD-1) are expressed on activated T cells and contribute to T cell exhaustion. The upregulation and ligation of CTLA-4/PD-1 (on T cells) with CTLA-4 ligands and PDL-1 (on tumor cells) dampens effector T cell activation and negatively attenuates adaptive immune responses.Citation13 Researchers have developed strategies to overcome the immunosuppressive tumor microenvironment by blocking the inhibitory pathways.Citation14 Therefore antibodies blocking CTLA-4 or PD-1 on T cells can prevent the inhibitory signals typically transmitted through these receptors and prevent effector cells from entering into the exhaustion phase, thereby extending the life and function of activated T cells. It seems logical to combine genetically targeted therapies/adoptive immunotherapy with negative regulatory blockade to minimize the chances of tumor resistance and escape. Accordingly, Treg depletion followed by PD-1/PDL-1 blockade has shown some efficacy in the treatment of acute myeloid leukemia (AML).Citation15 In a Phase I clinical trial of antibody-mediated PD-1 blockade, an objective response (complete response [CR] or partial response [PR]) was observed in those with non-small-cell lung cancer ([NSCLC] 18%), melanoma (28%), and renal cell cancer ([RCC] 27%).Citation16 A similar Phase I trial using antibody-mediated blockade of PDL-1 induced durable tumor regression and prolonged stabilization of disease in patients with advanced cancers.Citation17 A study has evaluated the contributions of CTLA-4 blockade on effector T cells and Treg populations in a mouse model of melanoma.Citation18 It revealed that CTLA-4 blockade on effector cells significantly improves tumor protection while blockade of Tregs completely fails to enhance antitumor responses, and a concomitant blockade of both effector and Tregs leads to maximal antitumor activity. CTLA-4 blockade with ipilimumab (an anti-CTLA-4 antibody) has resulted in some clinical responses in patients with melanoma, ovarian cancer, prostate cancer, and RCC.Citation19 A Phase III trial showed that ipilimumab, when given with or without a glycoprotein (gp)100 peptide vaccine, improved the overall survival to 10 months when compared to 6.4 months with gp100 alone in patients with metastatic melanoma.Citation20 Several Phase II studies suggest that ipilimumab is effective in patients with melanoma and brain metastases.Citation21,Citation22 In a Phase II trial of ipilimumab plus fotemustine in 86 patients with advanced melanoma, of whom 20 patients had asymptomatic brain metastases at baseline. 40 of 86 (46.5%) patients in the study population achieved disease control similar to 10 of 20 patients (50%) with brain metastases.Citation23 Furthermore, ipilimumab when combined with decarbazine improved the overall survival to 47% when compared to decarbazine alone (36%).Citation24 These results suggest that blocking the immune checkpoints can improve overall survival in cancer patients.

Cancer vaccines and immunotherapy

The success of a cancer vaccine is dependent on the ability of a patient to mount a primary or memory immune response against cancer antigens used in the vaccine. Thus far, the majority of cancer vaccine studies have focused on patients with relapsed or therapy refractory disease, but there is a growing interest on the potential to use this approach to prevent relapse in patients who are at high risk for recurrence. Three main types of cancer vaccines that have been used in previous studies, including cellular vaccines, largely consist of DCs pulsed with cancer relevant antigens or tumor cell lysates, protein- or peptide-based vaccines, and vector-based vaccines where plasmid DNA and viral/bacterial/yeast vectors are used to deliver tumor-specific antigens.Citation25 Potential problems with using whole cell lysates, peptides, or plasmid DNA approaches include the immunogenicity of the vaccine, the majority of cancer reactive T cells exist in low numbers and are difficult to expand, and that most tumors have developed multiple means to evade the immune system. Adjuvants can be used to enhance vaccine immunogenicity and thereby increase the likelihood of eliciting a T cell response. Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been used as an adjuvant in several types of tumors including melanoma, colorectal carcinoma, RCC, and lymphoma. For example, an idiotypic protein vaccine together with GM-CSF resulted in complete molecular remission (by polymerase chain reaction [PCR]) in 8 of 11 lymphoma patients and tumor-specific cytotoxic CD4+ and CD8+ cells were found in 95% of the patients.Citation26 DCs play a central role in initiating antitumor responses by activating innate and adaptive immune cells. Different DC subsets express distinct toll-like receptors (TLRs), such as TLRs 1 to 8, and upon stimulation, upregulate costimulatory molecules, pro-inflammatory cytokines, and chemokines which can assist in priming tumor-specific T cells. Therefore, different types of TLR agonists have been used as adjuvants along with DC-based vaccines in treating glioblastoma, breast cancer, melanoma, RCC, and leukemia.Citation27 A list of clinical trials using DC as therapeutic vaccines has been detailed in a comprehensive review of cancer immunotherapy with these antigen presenting cells.Citation28

Cancer testis antigens

An ideal tumor antigen for immunotherapy should be (a) expressed specifically on tumor cells and not on healthy cells, (b) stably and homogenously expressed on all/majority of tumor cells, (c) vital for the existence of cancer cells, and (d) targeted by tumor antigen-specific cytotoxic T lymphocytes.Citation29 Identification of such tumor antigens would enhance the success of cancer vaccines.

CT antigens are tumor proteins with a restricted pattern of expression, generally limited to germ cell and trophoblast tissue, but are also expressed in various human cancers. Their stable and specific expression on tumor cells and lack of expression on normal tissues make them an attractive target for cancer immunotherapy. Based on the frequency of CT antigen expression, Chen et alCitation30 and Caballero and ChenCitation31 classified certain types of cancers including melanoma, ovarian cancer, lung cancer, and bladder cancer as “CT-rich” tumors; RCC, colorectal cancer, and lymphoma/leukemia as “CT-poor” tumors; and breast cancer, bladder cancer, and prostate cancer as “CT-intermediate” tumors. CT antigens are divided into two groups: CT-X (encoded on X chromosome) and non-X CT antigens. An excellent review by Simpson et al summarizes the characteristics and functions of these two types of CT antigens.Citation29 Until 2004, there were around 40 CT antigens identified,Citation33 but by 2012, the number of CT antigens identified had increased to 110.Citation32 Our review will focus mainly on melanoma antigen family (MAGE)-A1, MAGE-A3, and New York esophageal squamous cell carcinoma (NY-ESO-1), three of the initially identified and most widely studied CT antigens in melanoma.

MAGE-A1 and MAGE-A3 are members of the MAGE gene family that are expressed on male germ line cells and placenta, as well as in melanoma, bladder cancer, breast cancer, prostate cancer, and NSCLC.Citation33 NY-ESO-1 is another CT antigen found on several tumors, including in ovarian cancer, lung cancer, melanoma, as well as some sarcomas and neuroblastomas.Citation34 Expression rates of MAGE-A1 and MAGE-A3 were 53.7% and 36.6%, respectively, in ovarian cancer.Citation35 Several MAGE-A1 peptides restricted to individual HLA alleles have been reported in healthy donors.Citation36Citation38 The frequency of expression of MAGE-A1 and NY-ESO-1 in bladder cancer versus liver cancer was 22% and 80% versus 80% and 29%, respectively.Citation33 In pharyngeal tumors, MAGE-A and NY-ESO-1 were detectable in 70% and 33.3% of tumors, respectively.Citation39 In NSCLC patients, the expression of NY-ESO-1 was only 8.3%,Citation40 while its expression in synovial sarcoma was 80%,Citation41 and its expression was 100% in myxoid/round cell liposarcoma patients.Citation42 Screening neuroblastoma cell lines for these antigens by reverse transcriptase-PCR (RT-PCR) has revealed that 44% are positive for MAGE-A1, 21% for MAGE-A3, and 30%–82% for NY-ESO-1, and immunohistochemical analysis has shown a good correlation between gene and protein expression.Citation43 In addition, in neuroblastoma, a higher level of NY-ESO-1 expression has been reported in patients with later stage disease.Citation44 The frequency of MAGE-A1 expression increased from 20% (in primary tumors) to 51% with advanced disease (in distant metastases), while NY-ESO-1 expression remained at 45%, regardless of stage of disease in melanoma patients.Citation45 In malignant gammopathies, the expression pattern of MAGE-A1, MAGE-A3, and NY-ESO-1 was heterogeneous, and the expression of these antigens was greater in patients with stage III extramedullary plasmacytoma or high risk myeloma relative to low risk disease groups.Citation46 This indicates that levels of expression of CT antigens vary depending upon the type of cancer and the stage of a patient’s disease, with many tumors having increased expression of CT antigens upon progression/relapse.Citation45Citation47

The expression of CT antigens on tumors has been correlated with the presence of CT antigen-specific B and T cell responses. Studies in adult patients have demonstrated that MAGE-A1 and MAGE-A3 specific T cells are present and can be augmented with a vaccine, or by stimulation of these T cells in culture.Citation48Citation51 There is also a correlation between the detection of MAGE-A3 specific CD8+ T cells and regression of tumors in melanoma patients.Citation52 MAGE-specific CD8+ T cell responses have been reported in AML patients.Citation53 In adult T cell leukemia/lymphoma cells, NY-ESO-1 and MAGE-A3 were expressed in 61.4% and 31.6% of cells, respectively. This study detected NY-ESO-1 specific antibodies in 11.6%, and NY-ESO-1 specific CD8+ T cell responses in 55.6%, of adult T cell leukemia/lymphoma patients.Citation54 Another study demonstrated CD8+ T cell responses in 10 of 11 patients with NY-ESO-1 positive melanoma who had NY-ESO-1 antibodies, but not in patients with NY-ESO-1 negative tumors or those lacking antibodies.Citation55,Citation56 There has also been a report on the detection of interferon-γ (IFN-γ) producing NY-ESO-1 specific T cells in neuroblastoma patients.Citation45 These studies indicate that MAGE-A1, MAGE-A3, and NY-ESO-1 are immunogenic and capable of eliciting T and B cell responses.

Clinical trials have been reported using DC-based vaccines, whole protein vaccines, or HLA restricted epitopes for MAGE-A1 and MAGE-A3 positive malignancies. Chianese-Bullock et al gave vaccines consisting of MAGE-A1, MAGE-A10, and gp100 peptides with GM-CSF and incomplete Freund’s adjuvant to patients with stage IIB to IV melanoma.Citation49 There were increases in MAGE-A1 specific IFN-γ production postvaccination, and cytotoxic T lymphocyte (CTL) from these patients lysed tumor cells expressing MAGE-A1. MacKensen et al reported on the results of a MAGE-A1 and MAGE-A3 peptide loaded DC vaccine in 14 melanoma patients.Citation57 Clinical and immunologic responses were seen in two patients, and increased melanoma peptide specific immune responses were seen in four patients.Citation57 Thurner et al reported the use of MAGE-A3 peptide pulsed mature DC at doses of 3 × 106 DC per vaccine, given at 14 day intervals.Citation51 Significant expansion of MAGE-A3 specific CD8+ cytotoxic T cells was induced in 8 of 11 patients, with regression of individual metastases in 6 of 11 patients. The ongoing clinical trials with CT antigens, MAGE-A1, MAGE-A3, and NY-ESO-1 are presented in .

Table 1 Ongoing clinical trials with the cancer testis antigens MAGE-A1, MAGE-A3, and NY-ESO-1

The majority of clinical trials with NY-ESO-1 tumor vaccines have used either individual HLA restricted epitopes or whole protein, with or without adjuvants. Most of these studies have demonstrated enhancement of T and B cell responses to this antigen postvaccination. Some of the initial clinical trials with NY-ESO-1 peptide vaccines used HLA-A2 restricted peptides, and demonstrated that CD8+ T cell responses can be expanded postvaccination.Citation56,Citation58,Citation59 Bender et al used an HLA-A2 restricted NY-ESO-1 peptide for vaccination, and reported that three of nine seronegative patients developed CD8+ T cell responses.Citation60 One study used full length NY-ESO-1 protein with the ISCOMATRIX™ adjuvant in 46 patients with fully resected, NY-ESO-1 positive tumors.Citation61 These investigators found high titer antibody responses, as well as CD4+ and CD8+ T cell responses, against a wide range of NY-ESO-1 epitopes postvaccination. There was improved survival, with only two of 19 relapses in the group receiving adjuvant and protein, in comparison with nine of 16 relapses in the group receiving protein alone. Upon further evaluation, persisting anti-NY-ESO-1 immunity was detected in ten of 14 recipients who had previously received vaccine with ISCOMATRIX™ adjuvant, while immunity only persisted in three of 14 recipients who received vaccine alone.Citation62

Combination therapy

A major focus of research during the past two decades has been to identify methods to overcome the mechanisms used by tumors to evade the immune system. Different approaches including conventional therapy, molecular-targeted therapy, and immunotherapy have been combined in an attempt to improve clinical outcomes. This includes using chemotherapy and blockade of immune checkpoints,Citation20,Citation63,Citation64 cancer vaccines and radiation therapy,Citation65 cancer vaccines and chemotherapy,Citation66,Citation67 cancer vaccines and molecular-targeted agents,Citation68 and molecular-targeted agents and blockade of immune checkpoints.Citation69 Current available combinations of immunotherapy and molecular-targeted therapy for cancer treatment are summarized in a review by Vanneman and Dranoff.Citation70 Depletion of Tregs in combination with a cancer vaccine is another approach. Tregs can be depleted by using anti-CD25 monoclonal antibodiesCitation71,Citation72 and studies show that chemotherapy agents such as cyclophosphamide can deplete/suppress Tregs.Citation73,Citation74 Among the different approaches available, we will focus our discussion on combining immunotherapy (using CT antigens) and chemotherapy, especially on the use of decitabine ([DAC] 5-aza-2′-deoxycytidine), a demethylating chemotherapeutic agent that epigenetically upregulates the expression of CT antigens, and review how CT antigens have been targeted in clinical trials.

The success of immunotherapy is largely dependent on the recognition of cancer cells expressing CT antigens by antigen-specific T cells, and this is dependent on antigen expression in the context of MHC class I and class II molecules. In cancer cells, hypermethylation of promoters leads to the downregulation of expression of CT antigensCitation75 and MHC molecules,Citation76 which are required for antigen presentation and recognition by antigen-specific cytotoxic T cells. Since not all tumors express CT antigens, one way to upregulate the expression of CT antigens and MHC molecules, and enhance tumor cell killing by antigen-specific cytotoxic T lymphocytes, would be to reverse hypermethylation by using demethylating agents. DAC is a potent inhibitor of DNA methylation, and the doses associated with the demethylating action of DAC are much lower than those required for cytotoxicity.Citation77Citation80 Several groups have demonstrated that demethylating agents, such as DAC, upregulate the expression of MAGE-A1, MAGE-A3, and NY-ESO-1 in a number of tumor cell lines,Citation81Citation84 potentially making these tumors more susceptible to MAGE-A1, MAGE-A3, and NY-ESO-1 mediated killing.

There have been several in vitro studies showing the effects of demethylating chemotherapy on the expression of CT antigens. One study demonstrated that the use of DAC could result in the restoration of MHC class I and MAGE antigens on melanoma cells.Citation85 Another group demonstrated that the treatment of ovarian cancer cell lines with DAC resulted in the upregulation of MAGE-A1 and MAGE-A3 expression, as well as MHC class I molecules.Citation81 Sigalotti et al treated 33 patients with AML or myelodysplastic syndrome (MDS) with DAC, and measured the expression of several CT antigens by RT-PCR.Citation86 In 31 of 33 patients who had no CT antigen expression prior to treatment, de novo expression of MAGE-A1 and NY-ESO-1 was observed in all but one patient 15 days after treatment. Weber et al demonstrated that MAGE-A1 expression was upregulated on several malignant melanoma cell lines following exposure to DAC,Citation83 and other studies have demonstrated that DAC can increase the expression of NY-ESO-1 on malignant glioma cell lines.Citation87,Citation88 Our group recently demonstrated that the majority of neuroblastoma cell lines had increased expression of MAGE-A1, MAGE-A3, and NY-ESO-1, on both a molecular and protein level, after 5 days exposure to DAC, and that this effect was associated with enhanced tumor cell killing by CT antigen specific CTL.Citation89 Upregulation of CT antigens and enhanced killing of tumor cells following treatment with DAC by CT antigen specific T cells suggests that immunotherapy using CT antigens in combination with DAC can be a potential strategy to treat relapsed patients.

Our ongoing Phase I clinical trial combining DAC and a DC vaccine targeting MAGE-A1, MAGE-A3, and NY-ESO-1 for patients with relapsed neuroblastoma demonstrated a complete response in our first patient. The clinical outcome was correlated with a robust increase in the number of MAGE-A3 specific CD8+ and CD4+ T cells, and the patient remains disease free 1 year following his vaccination.Citation90 This study indicates that a combination of demethylation-based chemotherapy followed by vaccine formulations containing CT antigens can elicit antigen-specific immune responses, potentially leading to an intensified antitumor effect.

Clinical trials are currently underway using genetically engineered NY-ESO-1 specific T cells for patients with synovial sarcoma, TCRs specific for MAGEA3/A6/B18 or NY-ESO-1/L antigen family member (LAGE) for patients with ovarian cancer, and TCRs specific for MAGE-A3 and NY-ESO-1 for patients with melanoma. Adoptive transfer of autologous T cells transduced with TCR directed against NY-ESO-1 has shown an objective clinical response in 4 of 6 patients with synovial cell sarcoma and in 5 of 11 patients with melanoma.Citation91 This study demonstrated a partial response lasting 18 months in 1 of 6 patients with synovial cell sarcoma and a complete regression, that lasted over 12 months, in 2 of 11 patients with melanoma.

Conclusion

CT antigens are ideal targets for immunotherapy and success of CT antigen based immunotherapy is largely dependent on the recognition of cancer cells expressing CT antigens by antigen-specific T cells. Combination therapy that includes a combination of different immunotherapeutic modalities, or combination of immunotherapy with DAC and/or other chemotherapy/irradiation, or both could overcome the obstacles related to effective antitumor immunity. Such a combination therapy should primarily target upregulation of CT antigen expression and pro-apoptotic molecules on tumor cells, enhance the expression of MHC class I and class II molecules and costimulatory molecules on antigen presenting cells, and downregulate the expression of coinhibitory molecules on the surface of T cells. A combination therapy using agents to target all three types of cells could result in an antitumor immune response, and further studies addressing issues of cell dosage, timing, and necessary sequence of agents used could improve clinical outcomes.

Disclosure

The authors report no conflicts of interest in this work.

References

  • YeeCThompsonJAByrdDAdoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and anti-tumor effect of transferred T cellsProc Natl Acad Sci U S A20029925161681617312427970
  • DudleyMEWunderlichJRRobbinsPFCancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytesScience2002298559485085412242449
  • YuJSLiuGYingHYongWHBlackKLWheelerCJVaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant gliomaCancer Res200464144973497915256471
  • KonoKTakahashiASugaiHDendritic cells pulsed with HER-2/neu-derived peptides can induce specific T-cell responses in patients with gastric cancerClin Cancer Res20028113394340012429626
  • ComitoMASunQLucasKGImmunotherapy for Epstein-Barr virus-associated tumorsLeuk Lymphoma200445101981198715370241
  • RiddellSRGreenbergPDPrinciples for adoptive T cell therapy of human viral diseasesAnnu Rev Immunol1995135455867612234
  • WalterEAGreenbergPDGilbertMJReconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donorN Engl J Med199533316103810447675046
  • CurranKJPegramHJBrentjensRJChimeric antigen receptors for T cell immunotherapy: current understanding and future directionsJ Gene Med201214640541522262649
  • MorganRADudleyMEWunderlichJRCancer regression in patients after transfer of genetically engineered lymphocytesScience2006314579612612916946036
  • KerkarSPSanchez-PerezLYangSGenetic engineering of murine CD8+ and CD4+ T cells for preclinical adoptive immunotherapy studiesJ Immunother201134434335221499127
  • ChoiBDCaiMBignerDDMehtaAIKuanCTSampsonJHBispecific antibodies engage T cells for antitumor immunotherapyExpert Opin Biol Ther201111784385321449821
  • BaitschLBaumgaertnerPDevêvreEExhaustion of tumor-specific CD8(+) T cells in metastases from melanoma patientsJ Clin Invest201112162350236021555851
  • NordeWJHoboWvan der VoortRDolstraHCoinhibitory molecules in hematologic malignancies: targets for therapeutic interventionBlood2012120472873622563087
  • GaoJBernatchezCSharmaPRadvanyiLGHwuPAdvances in the development of cancer immunotherapiesTrends Immunol2013342909823031830
  • ZhouQMungerMEHighfillSLProgram death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemiaBlood2010116142484249320570856
  • TopalianSLHodiFSBrahmerJRSafety, activity, and immune correlates of anti-PD-1 antibody in cancerN Engl J Med2012366262443245422658127
  • BrahmerJRTykodiSSChowLQSafety and activity of anti-PD-L1 antibody in patients with advanced cancerN Engl J Med2012366262455246522658128
  • PeggsKSQuezadaSAChambersCAKormanAJAllisonJPBlockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodiesJ Exp Med200920681717172519581407
  • WeberJReview: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse eventsOncologist200712786487217673617
  • HodiFSO’DaySJMcDermottDFImproved survival with ipilimumab in patients with metastatic melanomaN Engl J Med2010363871172320525992
  • MargolinKErnstoffMSHamidOIpilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trialLancet Oncol201213545946522456429
  • WeberJSAminAMinorDSafety and clinical activity of ipilimumab in melanoma patients with brain metastases: retrospective analysis of data from a phase 2 trialMelanoma Res201121653053422051508
  • Di GiacomoAMAsciertoPAPillaLIpilimumab and fotemustine in patients with advanced melanoma (NIBIT-M1): an open-label, single-arm phase 2 trialLancet Oncol201213987988622894884
  • RobertCThomasLBondarenkoIIpilimumab plus dacarbazine for previously untreated metastatic melanomaN Engl J Med2011364262517252621639810
  • BolhassaniASafaiyanSRafatiSImprovement of different vaccine delivery systems for cancer therapyMol Cancer201110321211062
  • BendandiMGockeCDKobrinCBComplete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphomaNat Med19995101171117710502821
  • GnjaticSSawhneyNBBhardwajNToll-like receptor agonists: are they good adjuvants?Cancer J201016438239120693851
  • PaluckaKBanchereauJCancer immunotherapy via dendritic cellsNat Rev Cancer201212426527722437871
  • SimpsonAJCaballeroOLJungbluthAChenYTOldLJCancer/testis antigens, gametogenesis and cancerNat Rev Cancer20055861562516034368
  • ChenYTRossDSChiuRMultiple cancer/testis antigens are preferentially expressed in hormone-receptor negative and high-grade breast cancersPLoS One201163e1787621437249
  • CaballeroOLChenYTCancer/testis (CT) antigens: potential targets for immunotherapyCancer Sci2009100112014202119719775
  • RestifoNPDudleyMERosenbergSAAdoptive immunotherapy for cancer: harnessing the T cell responseNat Rev Immunol201212426928122437939
  • ScanlanMJSimpsonAJOldLJThe cancer/testis genes: review, standardization, and commentaryCancer Immun20044114738373
  • NicholaouTEbertLDavisIDDirections in the immune targeting of cancer: lessons learned from the cancer-testis Ag NY-ESO-1Immunol Cell Biol200684330331716681828
  • ZhangSZhouXYuHYuYExpression of tumor-specific antigen MAGE, GAGE and BAGE in ovarian cancer tissues and cell linesBMC Cancer20101016320423514
  • WangXFCohenWMCastelliFASelective identification of HLA-DP4 binding T cell epitopes encoded by the MAGE-A gene familyCancer Immunol Immunother200756680781816988823
  • LuitenRDemotteNTineJvan der BruggenPA MAGE-A1 peptide presented to cytolytic T lymphocytes by both HLA-B35 and HLA-A1 moleculesTissue Antigens2000561778110958359
  • LuitenRvan der BruggenPA MAGE-A1 peptide is recognized on HLA-B7 human tumors by cytolytic T lymphocytesTissue Antigens200055214915210746786
  • Pastorcic-GrgicMSarcevicBDosenDJureticASpagnoliGCGrgicMPrognostic value of MAGE-A and NY-ESO-1 expression in pharyngeal cancerHead Neck20103291178118420029985
  • YoshidaNAbeHOhkuriTExpression of the MAGE-A4 and NY-ESO-1 cancer-testis antigens and T cell infiltration in non-small cell lung carcinoma and their prognostic significanceInt J Oncol20062851089109816596224
  • JungbluthAAAntonescuCRBusamKJMonophasic and biphasic synovial sarcomas abundantly express cancer/testis antigen NY-ESO-1 but not MAGE-A1 or CT7Int J Cancer200194225225611668506
  • PollackSMJungbluthAAHochBLNY-ESO-1 is a ubiquitous immunotherapeutic target antigen for patients with myxoid/round cell liposarcomaCancer2012118184564457022359263
  • WölflMJungbluthAAGarridoFExpression of MHC class I, MHC class II, and cancer germline antigens in neuroblastomaCancer Immunol Immunother200554440040615449039
  • RodolfoMLukschRStockertEAntigen-specific immunity in neuroblastoma patients: antibody and T-cell recognition of NY-ESO-1 tumor antigenCancer Res200363206948695514583496
  • BarrowCBrowningJMacGregorDTumor antigen expression in melanoma varies according to antigen and stageClin Cancer Res2006123 Pt 176477116467087
  • DhodapkarMVOsmanKTeruya-FeldsteinJExpression of cancer/testis (CT) antigens MAGE-A1, MAGE-A3, MAGE-A4, CT-7, and NY-ESO-1 in malignant gammopathies is heterogeneous and correlates with site, stage and risk status of diseaseCancer Immun20033912875607
  • SuyamaTShiraishiTZengYExpression of cancer/testis antigens in prostate cancer is associated with disease progressionProstate201070161778178720583133
  • ChauxPLuitenRDemotteNIdentification of five MAGE-A1 epitopes recognized by cytolytic T lymphocytes obtained by in vitro stimulation with dendritic cells transduced with MAGE-A1J Immunol199916352928293610453041
  • Chianese-BullockKAPressleyJGarbeeCMAGE-A1-, MAGE-A10-, and gp100-derived peptides are immunogenic when combined with granulocyte-macrophage colony-stimulating factor and montanide ISA-51 adjuvant and administered as part of a multipeptide vaccine for melanomaJ Immunol200517453080308615728523
  • CarrascoJVan PelANeynsBVaccination of a melanoma patient with mature dendritic cells pulsed with MAGE-3 peptides triggers the activity of nonvaccine anti-tumor cellsJ Immunol200818053585359318292586
  • ThurnerBHaendleIRöderCVaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanomaJ Exp Med1999190111669167810587357
  • ConnerotteTVan PelAGodelaineDFunctions of Anti-MAGE T-cells induced in melanoma patients under different vaccination modalitiesCancer Res200868103931394018483279
  • GoodyearOAgathanggelouANovitzky-BassoIInduction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasiaBlood2010116111908191820530795
  • NishikawaHMaedaYIshidaTCancer/testis antigens are novel targets of immunotherapy for adult T-cell leukemia/lymphomaBlood2012119133097310422323448
  • JägerENagataYGnjaticSMonitoring CD8 T cell responses to NY-ESO-1: correlation of humoral and cellular immune responsesProc Natl Acad Sci U S A20009794760476510781081
  • JägerEGnjaticSNagataYInduction of primary NY-ESO-1 immunity: CD8+ T lymphocyte and antibody responses in peptide-vaccinated patients with NY-ESO-1+ cancersProc Natl Acad Sci U S A20009722121981220311027314
  • MacKensenAHerbstBChenJLPhase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in vitro from CD34(+) hematopoietic progenitor cellsInt J Cancer200086338539210760827
  • ShackletonMDavisIDHopkinsWThe impact of imiquimod, a Toll-like receptor-7 ligand (TLR7L), on the immunogenicity of melanoma peptide vaccination with adjuvant Flt3 ligandCancer Immun20044915384929
  • ChenQJacksonHShackletonMCharacterization of antigen-specific CD8+ T lymphocyte responses in skin and peripheral blood following intradermal peptide vaccinationCancer Immun20055515755075
  • BenderAKarbachJNeumannALUD 00-009: phase 1study of intensive course immunization with NY-ESO-1 peptides in HLA-A2 positive patients with NY-ESO-1-expressing cancerCancer Immun200771617944437
  • DavisIDChenWJacksonHRecombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4(+) and CD8(+) T cell responses in humansProc Natl Acad Sci U S A200410129106971070215252201
  • NicholaouTChenWDavisIDImmunoediting and persistence of antigen-specific immunity in patients who have previously been vaccinated with NY-ESO-1 protein formulated in ISCOMATRIX™Cancer Immunol Immunother201160111625163721698545
  • RobertCThomasLBondarenkoIIpilimumab plus dacarbazine for previously untreated metastatic melanomaN Engl J Med2011364262517252621639810
  • RosenblattJGlotzbeckerBMillsHPD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccineJ Immunother201134540941821577144
  • GulleyJLArlenPMBastianACombining a recombinant cancer vaccine with standard definitive radiotherapy in patients with localized prostate cancerClin Cancer Res20051193353336215867235
  • ArlenPMGulleyJLParkerCA randomized phase II study of concurrent docetaxel plus vaccine versus vaccine alone in metastatic androgen-independent prostate cancerClin Cancer Res20061241260126916489082
  • LaheruDLutzEBurkeJAllogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activationClin Cancer Res20081451455146318316569
  • FarsaciBHigginsJPHodgeJWConsequence of dose scheduling of sunitinib on host immune response elements and vaccine combination therapyInt J Cancer201213081948195921633954
  • StaggJLoiSDivisekeraUAnti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapyProc Natl Acad Sci U S A2011108177142714721482773
  • VannemanMDranoffGCombining immunotherapy and targeted therapies in cancer treatmentNat Rev Cancer201212423725122437869
  • RechAJVonderheideRHClinical use of anti-CD25 antibody daclizumab to enhance immune responses to tumor antigen vaccination by targeting regulatory T cellsAnn N Y Acad Sci200911749910619769742
  • JacobsJFPuntCJLesterhuisWJDendritic cell vaccination in combination with anti-CD25 monoclonal antibody treatment: a phase I/II study in metastatic melanoma patientsClin Cancer Res201016205067507820736326
  • ZhaoJCaoYLeiZYangZZhangBHuangBSelective depletion of CD4+CD25+Foxp3+ regulatory T cells by low-dose cyclophosphamide is explained by reduced intracellular ATP levelsCancer Res201070124850485820501849
  • van der MostRGCurrieAJMahendranSTumor eradication after cyclophosphamide depends on concurrent depletion of regulatory T cells: a role for cycling TNFR2-expressing effector-suppressor T cells in limiting effective chemotherapyCancer Immunol Immunother20095881219122819052741
  • De SmetCLurquinCLethéBDNA methylation is the primary silencing mechanism for a set of germ line- and tumor-specific genes with a CpG-rich promoterMol Cell Biol199919117327733510523621
  • YeQShenYWangXHypermethylation of HLA class I gene is associated with HLA class I down-regulation in human gastric cancerTissue Antigens2010751303919883394
  • MarksPRifkindRARichonVMBreslowRMillerTKellyWKHistone deacetylases and cancer: causes and therapiesNat Rev Cancer20011319420211902574
  • YangHHoshinoKSanchez-GonzalezBKantarjianHGarcia-ManeroGAntileukemia activity of the combination of 5-aza-2′-deoxycytidine with valproic acidLeuk Res200529773974815927669
  • Garcia-ManeroGGoreSDFuture directions for the use of hypomethylating agentsSemin Hematol2005423 Suppl 2S50S5916015506
  • IssaJPGharibyanVCortesJPhase II study of low-dose decitabine in patients with chronic myelogenous leukemia resistant to imatinib mesylateJ Clin Oncol200523173948395615883410
  • AdairSJHoganKTTreatment of ovarian cancer cell lines with 5-aza-2′-deoxycytidine upregulates the expression of cancer-testis antigens and class I major histocompatibility complex-encoded moleculesCancer Immunol Immunother200958458960118791715
  • SchrumpDSFischetteMRNguyenDMPhase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleuraClin Cancer Res200612195777578517020984
  • WeberJSalgallerMSamidDExpression of the MAGE-1 tumor antigen is up-regulated by the demethylating agent 5-aza-2′-deoxycytidineCancer Res1994547176617717511051
  • AlmstedtMBlagitko-DorfsNDuque-AfonsoJThe DNA demethylating agent 5-aza-2′-deoxycytidine induces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leukemia cellsLeuk Res201034789990520381863
  • SerranoATanzarellaSLionelloIRexpression of HLA class I antigens and restoration of antigen-specific CTL response in melanoma cells following 5-aza-2′-deoxycytidine treatmentInt J Cancer200194224325111668505
  • SigalottiLAltomonteMColizziF5-Aza-2′-deoxycytidine (decitabine) treatment of hematopoietic malignancies: a multimechanism therapeutic approach?Blood2003101114644464612756166
  • OiSNatsumeAItoMSynergistic induction of NY-ESO-1 antigen expression by a novel histone deacetylase inhibitor, valproic acid, with 5-aza-2′-deoxycytidine in glioma cellsJ Neurooncol2009921152219030781
  • NatsumeAWakabayashiTTsujimuraKThe DNA demethylating agent 5-aza-2′-deoxycytidine activates NY-ESO-1 antigenicity in orthotopic human gliomaInt J Cancer2008122112542255318240144
  • BaoLDunhamKLucasKMAGE-A1, MAGE-A3, and NY-ESO-1 can be upregulated on neuroblastoma cells to facilitate cytotoxic T lymphocyte-mediated tumor cell killingCancer Immunol Immunother20116091299130721626030
  • KrishnadasDKShapiroTLucasKComplete remission following decitabine/dendritic cell vaccine for relapsed neuroblastomaPediatrics20131311e336e34123266925
  • RobbinsPFMorganRAFeldmanSATumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1J Clin Oncol2011297917929021282551