Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 19.
Published in final edited form as: Cancer Biol Ther. 2009 Aug 1;8(15):1440–1449. doi: 10.4161/cbt.8.15.9133

Enhancing immune responses to tumor-associated antigens

Jack P Higgins 1, Michael B Bernstein 1, James W Hodge 1,*
PMCID: PMC7236598  NIHMSID: NIHMS1587292  PMID: 19556848

Abstract

The goal of vaccine-based cancer immunotherapy is to induce a tumor-specific immune response that ultimately reduces tumor burden. However, the immune system is often tolerant to antigens presented by the tumor, as the cancer originates from within a patient and is therefore recognized as self. This article reviews selected clinical strategies for overcoming this immune tolerance, and approaches to enhance generation of immunity to tumor-associated antigens by activating innate immunity, potentiating adaptive immunity, reducing immunosuppression, and enhancing tumor immunogenicity. Success in the field of cancer vaccines has yet to be fully realized, but intelligent choice of immunomodulators, tumor antigens and patient populations will likely lead to clinically relevant uses for cancer vaccines.

Keywords: tumor-associated antigen, immunotherapy, vaccine, costimulation

Introduction

Following the vision of Dr. William Coley, tumor immunologists have sought to develop cancer therapies by directing a patient’s own immune system to destroy tumor cells.1 Advances in our understanding of genomics, proteomics and immunology have led to the clinical development of numerous cancer immunotherapies. Identification of tumor-associated antigens (TAAs) has facilitated the development of various cancer vaccination strategies to elicit tumor-specific immunity potentially capable of reducing tumor burden. However, strategies to induce tumor-specific immunity in patients must overcome several obstacles: (1) insufficient and poorly functional populations of antigen-presenting cells (APCs) and lymphocytes; (2) inducing immunity potent enough to overcome potential tolerance mechanisms without inducing unacceptable autoimmune toxicities; (3) poor immunogenicity of antigens expressed by tumor cells; and (4) immunoregulatory pathways that dampen the tumor-specific immune response. In numerous clinical trials to date, various strategies have been used in combination with cancer vaccine modalities in an effort to enhance the immune response to TAAs, including cytokine support, toll-like receptor (TLR) agonists, costimulation, chemotherapy, radiotherapy, ablative therapy, immunoregulatory modulation, adjuvants and vector-driven immunity.

Tumor-Associated Antigens

Many potential targets for cancer immunotherapy have been identified to date by a variety of methods (Table 1). Methods of identifying immunologically relevant tumor antigens include patient antibody responses, genomics, biochemical strategies, SEREX and reverse immunology.26 Most TAAs that are over-expressed in malignant tissue are also expressed in normal tissue or originate as oncofetal antigens, and thus peripheral or thymic tolerance to TAAs often exists. It is important to note that when an antigen is targeted for cancer immunotherapy, the activated T-cells induced by vaccination recognize only the peptide-major histocompatibility complex (MHC) complexes of the tumor antigen on the cell surface, and not the surface protein. Thus, targets of vaccine therapy need not be cell-surface proteins. Additionally, a secondary, costimulatory signal is needed to fully activate adaptive T-cell responses. Identification of lipid antigens for natural killer T-cells and tumor stroma-associated antigens will further expand the repertoire of available tumor targets.79 A current challenge is to determine which of these TAAs warrant further development.

Table 1.

Partial list of current and potential targets for cancer vaccine therapy

Carcinoma-associated antigens Endogenous Retroviral gene products Leukemia/lymphoma antigens Melanoma antigens Viral antigens
Brachyury HERV-H Aberrant class II MAGE HPV
CA125 HERV-K Anti-idiotype MART
CEA B1 gp100
EGFR CD19 Tyrosinase
HER-2/neu CD20 CD2
KSA CD22 CD3
Mesothelin CD25 GM2
MUC-1 CD36
NY-ESO
p53
PAGE-4
PAP
PSA
PSCA
PSMA
Ras
sTn
TARP
VEGF

Approaches to Inducing Tumor-Specific Immunity

Several modalities that seek to reduce tumor burden by inducing tumor-specific immunity are currently under investigation in clinical trials. A review by Fournier and Schirrmacher recently described the state of the art in 2008 for cancer vaccines and autologous cell-based therapies (summarized in Table 2).10 Unfortunately, each of these classes of immunotherapies has had disappointing Phase III trial results. However, careful attention to clinical trial design, primary endpoint selection, patient population selection, combinational therapy approaches and vaccine delivery could potentially lead to successful treatment strategies for a variety of malignancies.

Table 2.

Partial list of current and potential cancer vaccine modalities

Tumor cells Protein/molecule-based Antigen-presenting cells Vector-based
Autologous Protein (HSP) Unmodified DNA/RNA
Allogeneic Peptide Apoptotic body loaded Yeast
Irradiated Glycopeptide Gene modified
Oncolysates Glycolipid mAb fusion proteins Peptide pulsed RNA transfected Bacterial: Salmonella
Gene modified: Anti-idiotype mAb Tumor lysate loaded Listeria
GM-CSF Tumor cell fusion Lactobacillus
Costimulatory molecules
Lymphotactin Viral: Vaccinia Adenovirus RNA replicons Avipox Fowlpox Canarypox MVA

Approaches to Enhancing Generation of TAA-Specific Immunity

TAAs are weakly immunogenic due to host immunotolerance, as well as the tumor’s immunosuppressive microenvironment and low expression of costimulatory molecules.11,12 Immunomodulators provide an opportunity to enhance the immunogenicity of TAAs, leading to the generation of TAA-specific adaptive immunity (Table 3). The following sections review the clinical applications of such immunomodulators and their potential for improving the efficacy of current cancer vaccine strategies.

Table 3.

Partial list of immunomodulators used for cancer vaccine modalities

Type Example Proposed mechanism of action
Cytokine GM-CSF mDCs activation → IL-1 & IL-12 → T-cell activation → IFNy1618
IL-7 Expansion of CD8+ & CD4+ T cells31
IL-12 T-cell activation → IFNγ137
IL-21 Enhanced cytolytic activity of NK & CD8+ T cells → IFNγ34,138
TLR agonist Imiquimod pDC activation → IFA-α → mDC maturation → IL-12 → T-cell activation63,64
CpG pDC activation → IFA-α → mDC maturation → IL-12 → T-cell activation68
MPL mDC activation → IL-12 → T-cell activation → IFNγ68
Costimulation B7–1 B7–1 → bind CD28 → T-cell activation43
ICAM-1 ICAM-1 → bind LFA-1 → T-cell activation43
LFA-3 LFA-3 → bind CD2 → T-cell activation43
Anti-CTLA-4 Block CTLA-4 binding to B7–1 → reduced costimulatory inhibition43
Chemotherapy Doxorubicin Tumor apoptosis → DC activation → T-cell immunity85
Radiotherapy EBRT Increased MHC I expression → enhanced CTL-mediated killing93
Ablative therapy RFA DC maturation → enhanced TAA presentation → T-cell immunity99
Cryoablation Antigen depot → enhanced APC antigen presentation → T-cell immunity102
NSAID Celecoxib Increased MHC expression → enhanced TAA presentation → T-cell immunity118
Treg modulation Denileukin diftitox Reduced Tregs prevents T-cell downregulation119
Classical adjuvant Montanide ISA 51 (IFA) Antigen depot → enhanced APC antigen presentation → T-cell immunity74

Cytokines

TNFα, IFNα and IL-2 are FDA-approved cytokines currently used for the treatment of soft tissue sarcoma, renal cell carcinoma and/or melanoma.1315 Other FDA-approved cytokines used to enhance lymphocyte and/or platelet recovery following chemotherapeutic regimens include granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte-colony-stimulating factor, erythropoietin and IL-11. Several of these cytokines, along with others, have demonstrated the ability to enhance APC or T-cell function. Increased APC function correlates with heightened levels of MHC, cell adhesion, and costimulatory molecules, which can lead to improved antigen presentation and induction of adaptive immune responses. Specifically, GM-CSF activates dendritic cells (DCs), which leads to production of IL-1 and IL-12 and activation of T-cells (Table 3).1618 In the production of the sipuleucel-T vaccine, an autologous DC-based therapy, GM-CSF is used to activate and mature DCs in vitro to enhance antigen uptake prior to patient reinfusion.19 A Phase III trial of sipuleucel-T demonstrated a significant increase in median survival of 4.5 mon in patients with hormone-refractory prostate cancer (25.9 vs. 21.4 mon; n = 127).20 However, as the primary endpoint of progression-free survival was not met, a similar Phase III trial measuring overall survival as the primary endpoint is ongoing, with reports of a survival advantage in patients receiving sipuleucel-T treatment.21,22 In another modality, GM-CSF (100 or 500 μg subcutaneously) has been used in combination with a follicular lymphoma-KLH anti-idiotype fusion antibody to induce tumor-specific CD4+ and CD8+ T-cell responses in 19 of 20 patients with follicular lymphoma,23 and is currently being evaluated in a Phase III trial in this patient population.24 Using a different vaccine platform, GM-CSF in combination with protein immunogens has also demonstrated an enhanced antigen-specific immune response.23,25 GM-CSF-secreting irradiated allogeneic whole tumor cells (GVAX) have also been used in the treatment of prostate cancer.26 A Phase I/II trial of GVAX in 80 patients with metastatic prostate cancer demonstrated a dose-dependent survival advantage that did not reach statistical significance.27 However, two Phase III trials using GVAX alone or in combination with docetaxel and prednisone were terminated due to a low probability of meeting the primary survival endpoint.28,29 A review by Parmiani et al. suggested that the adjuvant use of high-dose GM-CSF (≥100 μg) and repeated dosing may expand myeloid-derived suppressor cells and thus dampen the tumor-specific adaptive immune response.30 Overall, the use of GM-CSF in combination with various vaccination strategies has demonstrated some clinical benefit.

Several other cytokines, including IL-2, IL-7, IL-12, IL-15 and IL-21, have a critical role in enhancing T-cell function.3134 The most studied of these, IL-2, has demonstrated clinical responses in metastatic melanoma and metastatic renal cell carcinoma.15,35 In vitro, IL-2 induces proliferation of CD8+ T-cells capable of lysing tumor cells.36 However, the combination of IL-2 with current peptide-based or tumor cell-based cancer vaccination strategies has yet to provide significant clinical benefit. Specifically, in patients with metastatic melanoma or metastatic renal cell carcinoma, multivalent autologous tumor lysates bound to silica beads in combination with IL-2 infusion (1.75 × 106 IU/m2/day) provided no survival benefit compared to vaccination alone.37 Similarly, other clinical trials using autologous tumor cell or peptide vaccines in combination with IL-2 have not yet demonstrated a significant clinical benefit.38,39 Thus, the potential of IL-2 to enhance immunity to TAAs delivered by peptide or tumor cell lysate vaccination has not yet been realized, but determining the optimal dose, route and schedule for the combined use of cytokines and cancer vaccines may result in clinical benefit.

A different approach to the use of IL-2 in cancer vaccination utilizes a modified vaccinia Ankara (MVA) virus carrying mucin (MUC)-1 and IL-2 transgenes (MVA/MUC1/IL-2; TG4010).40 A clinical study in prostate cancer patients with evidence of prostate-specific antigen (PSA) progression evaluated the significance of the timing of MVA/MUC1/IL-2 vaccinations.41 Patients in Arm 1 received 6 weekly vaccinations, then were vaccinated once every 3 w; patients in Arm 2 were vaccinated once every 3 w. A significant improvement in PSA doubling time was observed in Arm 1 compared to Arm 2. Finally, the use of IL-2 to expand lymphocytes for adoptive transfer is being utilized in clinical trials with evidence of clinical efficacy.42

Overall, optimizing the use of cytokines to enhance generation of tumor-specific immunity is a challenging and ongoing process, but increased understanding of proper dosage and routes of administration, coupled with investigation of additional cytokines such as IL-7, IL-12, IL-15 and IL-21, could improve the clinical efficacy of cancer vaccines.

Costimulation

Efficient activation of T-cells requires two signals. The first signal is mediated through the presentation of a peptide-MHC complex on the surface of the APC to the T-cell receptor on the surface of the T-cell. The second signal involves binding of a T-cell costimulatory molecule on the surface of the APC to its ligand on the surface of the T-cell. One of the most studied costimulatory molecules is B7–1 (CD80), which interacts with either of two T-cell ligands, leading to upregulation (CD28) or downregulation (CTLA-4) of T-cell function (Table 3).43 In a clinical setting, an allogeneic breast cancer cell line vaccine expressing B7–1, used in combination with GM-CSF or BCG, was evaluated in 30 women with metastatic breast cancer.44 Following vaccination, increased CD8+IFNγ+ PBMCs were isolated, but no clinical responses were reported. Another trial used a canarypox viral vector expressing carcinoembryonic antigen (CEA) and B7–1 (ALVAC-CEA/B7–1) in combination with chemotherapy (FOLFIRI) in 118 metastatic colorectal cancer patients.45 Vaccination with ALVAC-CEA/B7–1 increased CEA-specific T-cell responses, but clinical responses observed were similar to other trials using the FOLFIRI chemotherapy regimen alone. Kaufman et al. intratumorally injected a recombinant vaccinia (rV) virus expressing B7–1 (rV-B7–1) in 12 patients with metastatic melanoma.46 Following vaccination, gene expression profiles from fine needle aspirations of melanoma lesions found an increase in CD8 and IFNγ expression in patients who exhibited a partial response, compared to patients with stable disease or progressive disease, suggesting an association between adaptive immune activation and clinical response. Another clinical trial of 28 patients with metastatic androgen-independent prostate cancer used docetaxel in combination with a vaccination strategy of rV-PSA admixed with rV-B7–1, followed by a similar recombinant fowlpox (rF) vector (rF-PSA) with GM-CSF.47 PSA velocities stabilized or decreased in nine of nine patients who received vaccine in combination with docetaxel and in eight of 11 vaccinated patients who received docetaxel upon disease progression, compared to 13 of 17 patients in a historical control who received docetaxel alone. Induction of PSA-specific T-cell responses was reported, as well as induction of tumor-specific immunity to separate antigens (PSMA, PAP and MUC-1). The generation of immunity to additional TAAs not expressed by the vaccine (antigen cascade) may provide insight as to why immunity to the vaccine-delivered antigens alone often does not correlate with clinical responses, as antigen cascade may be necessary in order to induce this effect. Overall, cancer vaccine strategies incorporating B7–1 have demonstrated the generation of TAA immunity in patients with various malignancies.

Additional costimulatory molecules such as intercellular adhesion molecule (ICAM)-1 and lymphocyte function-associated antigen (LFA)-3 have been used in combination with B7–1 (the triad designated TRICOM) in cancer vaccines for various malignancies.48 Delivering TRICOM via a poxviral vector leads to increased expression of these three costimulatory molecules on the surface of infected cells, which enhances T-cell costimulation and activation. In a preclinical CEA-transgenic mouse model, vaccination with an rV vector containing CEA and TRICOM (rV-CEA/TRICOM) increased the avidity of CEA-specific cytotoxic T lymphocytes (CTLs) 100-fold compared to rV-CEA only.49 This enhancement of T-cell function demonstrates how adequate costimulation can enhance the quality of immune responses to TAAs. In a Phase I study of 15 patients with metastatic prostate cancer, an rV-PSA/TRICOM prime was followed by boosts of rF-PSA/TRICOM with or without GM-CSF.48 Of six HLA-A*0201 patients, four demonstrated an increase in PSA-specific T-cell ELISPOT responses, and PSA velocities decreased in nine of 15 vaccinated patients. In another trial, 25 patients with metastatic carcinoma were vaccinated with rV-CEA/MUC1/TRICOM followed by boosts with a similar fowlpox vector, both in combination with GM-CSF.50 Immune responses to MUC-1 or CEA were observed in nine of 16 vaccinated patients, and one patient with clear cell ovarian cancer and one with metastatic breast cancer demonstrated clinical responses radiographically. Together, these data demonstrate that increasing the costimulatory signal with TRICOM can enhance the generation of TAA-specific immunity and potentially lead to clinical benefits in cancer patients.

As described above, the costimulatory molecule B7–1 expressed by APCs can either activate (bind CD28) or downregulate (bind CTLA-4) T-cell responses. Ipilimumab is a CTLA-4-specific antibody used to reduce binding of B7–1 to CTLA-4 and thus potentiate T-cell activation.51 Clinical trials using ipilimumab as a monotherapy in metastatic melanoma patients or in combination with GM-CSF in metastatic prostate cancer patients have demonstrated some clinical responses and expansion of activated CD8+ T-cells.52,53 A study combining ipilimumab with a gp100 peptide vaccine in 14 patients with metastatic melanoma reported that 11 patients demonstrated gp100-specific immune responses following in vitro sensitization and significant increases in CD3+CD4+ and CD3+CD4 (presumed CD8+) T-cells following treatment; two patients had complete clinical responses and one had a partial clinical response.54 Blocking CTLA-4 downregulation of T-cell responses in combination with vaccination is being further evaluated in several ongoing clinical trials.

Another approach to enhancing TAA-specific immunity is to activate APCs through costimulation. CD40 is one such target expressed on DCs, B-cells and monocytes.55 Ligation of CD40 expressed on APCs leads to activation, whereas ligation of CD40 expressed by some carcinomas and melanoma can lead to apoptotic cell death.55,56 A CD40 agonist monoclonal antibody (mAb) (CP-870,893) was used in a Phase I dose escalation trial of 29 patients with advanced solid tumors.57 This study reported a dose-related increase in CD86+CD19+ B-cells following treatment, with four of 15 melanoma patients showing objective partial responses at restaging (43 d). Targeting CD40 has the benefit of APC activation and a direct cytolytic effect on tumors, and could prove beneficial for the treatment of various malignancies. Combining this CD40 mAb with autologous cell transfer or cancer vaccines may further improve patient outcomes.

Toll-Like Receptor Agonists

Among the primary signaling mechanisms of the innate system that discriminates between self and non-self are pattern-recognition receptors. Expressed on APCs, these receptors recognize pathogen-associated molecular patterns such as bacterial lipopolysaccharide (LPS), bacterial CpG motifs, viral DNA, dsRNA, mannose and yeast.58 TLRs comprise one such class of pattern-recognition receptors. Several synthetic TLR agonists are under clinical investigation, and one TLR-7 agonist (imiquimod) is FDA-approved for superficial basal cell carcinoma, genital warts and actinic keratosis.59 Direct application of topical imiquimod has demonstrated clinical responses in various cutaneous metastases of the skin.60,61 This antitumor activity is likely directed through TLR-dependent (TLR-7/TLR-8 activation of APCs) and independent (tumor apoptosis) pathways.62 One immune-mediated mechanism of action for imiquimod is TLR-7 activation of plasmacytoid DCs (pDCs), leading to IFNα production that activates and matures myeloid-derived DCs (mDCs), leading to production of proinflammatory cytokines and enhanced antigen presentation that activates adaptive immunity (Table 3).63,64 Imiquimod has been used clinically in combination with peptide/protein-based vaccines for malignant melanoma and prostate cancer.6567 In a trial of nine patients with malignant melanoma, vaccination with NY-ESO-1 protein in combination with topical imiquimod at a healthy skin extremity generated NY-ESO-1-specific CD4+ T-cell responses in six of eight patients.66 A Phase I/II trial of 19 hormone-sensitive prostate cancer patients following biochemical failure compared a peptide vaccine (11 HLA-A*0201 peptides and two HLA class II peptides) alone or in combination with imiquimod, GM-CSF, hyperthermia, or a MUC-1-mRNA/protamine complex adjuvant.67 Of four patients who received vaccine plus imiquimod, three demonstrated an increase of PSA doubling time and one showed an interim decline followed by a rise in PSA. Only four of 15 patients comprising all other groups reported similar PSA responses. Together, these reports demonstrate the ability of a TLR-7 agonist (imiquimod), used in combination with peptide/protein-based vaccines, to enhance the generation of TAA-specific immunity and lead to clinical benefits.

Bacterial CpG DNA motifs that signal through TLR-9 have been used in combination with various vaccination strategies to treat infectious diseases and cancer.68 Several synthetic CpG oligodeoxynucleotides signal through TLR-9 and lead to B-cell activation, pDC production of IFNα, and/or mDC production of IL-12 (Table 3).68 One TLR-9 agonist (PF-3512676) used in a Phase II trial of stage IIIB/IV non-small cell lung cancer (NSCLC) in combination with platinum-based chemotherapy demonstrated a trend in enhanced overall survival that did not reach statistical significance.69 Additionally, two Phase III trials in NSCLC have been discontinued due to lack of benefit of PF-3512676 over standard-of-care chemotherapy.70 Another vaccine using a TLR-9 agonist (VaxImmune™) in combination with a MAGE-A3 TAA (ASCI; GSK1572932A) is currently being evaluated in a Phase III trial of patients with early-stage (IB, II, IIIA) NSCLC.71 In a Phase II trial of 182 stage IB/II NSLC patients randomized 2:1 to receive MAGE-A3 vaccine versus placebo, endpoints for disease-free interval, disease-free survival, and overall survival were improved in the vaccine arm, but not to significant levels.72 Further, microarray analysis of tumor biopsies revealed a genetic signature associated with a high risk of postoperative relapse, which seemed to correlate with improved clinical efficacy of the vaccine.73 More information about patient and tumor biology will help to identify the most appropriate patient populations for immunotherapy. Oil emulsions such as montanide ISA-51 (IFA) form an antigen depot and facilitate the slow release of peptide antigens to reduce the induction of immunological tolerance caused by presentation of antigen through nonprofessional APCs.74 This approach, in combination with TLR agonists such as CpG, has demonstrated induction of TAA-specific T-cell responses in melanoma patients.75,76

Another TLR agonist of interest is monophosphoryl lipid (MPL) A, which targets TLR-4.77 MPL is a derivate of LPS and induces a Th1-like cytokine response similar to LPS.78 L-BLP25 (Stimuvax®) is a liposomal vaccine that incorporates MPL and carries TAAs targeting the extracellular core protein of MUC-1. In a phase IIB trial, 171 patients with stage IIIB/IV NSCLC were randomized to either low-dose cyclophosphamide administered 3 d before weekly L-BLP25 vaccinations plus best supportive care, or best supportive care alone.79 At a 2-y follow-up, a 4.4-mon survival advantage was noted in the vaccine arm, but did not reach statistical significance. A subgroup of patients with stage IIIB locoregional NSCLC appeared to respond better to L-BLP25 vaccination than patients with more progressive disease. L-BLP25 is currently being investigated in combination with cyclophosphamide versus placebo in a Phase III trial of patients with stage III NSCLC.80 Targeting patients with less advanced disease may prove beneficial for this and other vaccination strategies.

Chemotherapy

Given the potential for lymphopenia and immunosuppression, using chemotherapeutic agents to enhance tumor-specific immune responses would appear to be counterintuitive. However, a growing body of evidence suggests that drugs commonly used in cancer chemotherapy can augment the antitumor effects of immunotherapeutic modalities. One mechanism proposed for this observed enhancement of the immune response is chemotherapy-induced apoptosis of tumor cells. The induction of tumor apoptosis by certain cytotoxic agents can not only activate DCs, but also provide them with an increased supply of tumor-specific antigens for presentation and cross-presentation to T-cells.8183 Furthermore, certain cytotoxic drugs can modulate tumor phenotype or induce immunogenic cell death, thus enhancing immune-mediated cell killing. In murine cell lines, 5-fluorouracil treatment and doxorubicin-induced caspase-dependent apoptosis enhanced CTL lysis of tumor cells.84,85 This ability of specific cytotoxic drugs to enhance the immunogenicity of tumor cells makes the combination of chemotherapy and immunotherapy attractive.

Another benefit to combining cytotoxic agents with immunotherapeutic approaches is the onset of homeostatic T-cell proliferation following drug-induced lymphopenia. The phenomenon of homeostatic T-cell proliferation was demonstrated in a pilot trial of patients with metastatic melanoma who were treated with expanded tumor-infiltrating lymphocytes (TILs) and IL-2 after a lymphodepleting regimen of cyclophosphamide and fludarabine.86 Results of this trial suggested that conditioning with nonmyeloablative chemotherapy before adoptive transfer of activated tumor-specific T-cells can enhance tumor regression and increase the rate of objective clinical responses. These findings correlated with those of Muranski et al. who reported that lymphodepletion enables the host to accommodate transferred T-cells and increases T-cell survival compared to competing cell populations.87

While these studies demonstrate the benefit of combining lymphodepletion by cytotoxic agents with passive immunotherapy (adoptive T-cell transfer), others show that combining active immunotherapy (vaccination) with chemotherapy has clinical promise as well. As described above, docetaxel plus a recombinant poxvirus-based vaccine (rV-PSA + rV-B7–1) increased antigen-specific T-cell responses to PSA, as well as to cascade antigens derived from the tumor.46 Similar results were found when patients with extensive small cell lung cancer were vaccinated with a DC-based p53 vaccine in combination with standard-of-care chemotherapy.88 Specifically, 16 of 28 patients (57%) demonstrated p53-specific T-cell responses to vaccination. In addition, the authors observed a high rate of objective clinical responses to chemotherapy (61.9%) immediately following vaccination, which was closely associated with the induction of immunologic responses. These clinical trials show that chemotherapy can be administered with immunotherapy without inhibiting the generation of vaccine-specific immune responses. These studies also suggest that, despite the potency of cytotoxic anticancer drugs, certain agents have immunomodulatory activity and can thus be successfully combined with different immunotherapeutic approaches to potentiate tumor-specific immunity.

Radiotherapy

Radiotherapy, like chemotherapy, has traditionally been considered immunosuppressive and thus an unlikely candidate for combination with immunotherapeutic approaches. However, the combination of radiation and immunotherapy holds particular promise as a strategy for cancer treatment. Evidence suggests that this combination may prevent cancer cells from escaping immune recognition through two main mechanisms: (a) radiation-induced tumor-cell death increases the source of tumor antigens for immune responses, and (b) radiation modulates tumor-cell phenotype, allowing for efficient immune-cell access and increased sensitivity to T-cell killing.89

Because of their low expression of costimulatory molecules, tumor cells often do not generate potent immune responses.11,12 However, previous studies have demonstrated that irradiation of tumor cells can induce recognition and phagocytosis signals for DCs and cause the release of “danger” signals required for DC activation.90,91 Furthermore, nonlethal doses of radiation have the ability to modulate tumor-cell phenotype to enhance T-cell-mediated killing.92,93 In addition, antigens released by dying tumor cells following radiation can be presented and cross-presented by DCs, thereby activating tumor-specific immune responses. Radiation-treated tumor cells could therefore serve as an in situ autologous tumor vaccine, augmenting antitumor immunity that could eradicate residual primary tumor cells as well as distant metastases.89,94 These preclinical studies have demonstrated the synergism of radiotherapy and immunotherapy in enhancing tumor-specific immune responses and immune-mediated tumor killing, and have provided the rationale for their combined use in the clinic.

A recent clinical trial combined a recombinant cancer vaccine with standard definitive radiotherapy in patients with localized prostate cancer.95 Patients were assigned to receive definitive radiotherapy with or without vaccine to determine if the vaccine could induce an immune response in the presence of tumor irradiation. The vaccine regimen consisted of a priming vaccination of rV-PSA admixed with rV-B7–1, followed by monthly booster vaccinations with rF-PSA. Patients received local radiotherapy either alone or in combination with vaccine (between the fourth and sixth vaccinations). Of 17 patients in the combination arm who completed all eight vaccinations, 13 (76%) had at least three-fold increases in circulating PSA-specific T-cells, while no detectable PSA-specific T-cell responses were noted in the radiotherapy-only arm.95 Patients in the combination arm also showed evidence of antigen cascade through generation of T-cell responses to prostate-associated antigens other than PSA, providing indirect evidence of immune-mediated tumor killing. Chi et al. also attempted to show enhanced tumor-specific immunity by combining vaccine with radiation therapy. In a Phase I trial, patients with refractory hepatoma received 8 Gy of external beam radiation followed by intratumoral injection of immature DCs on days 2 and 24.96 Results of this study reported seven of ten evaluable patients had increases in α-fetoprotein-specific T-cell ELISPOT responses. Several of these patients also showed partial clinical responses, reinforcing the relationship between induction of antitumor immunity and reduction of disease burden. Taken together, these studies demonstrate that radiation can enhance tumor-specific immune responses and, in combination with immunotherapeutic approaches, may increase tumor cell killing compared to either modality alone.

Ablative Therapy

Another approach to inducing immunogenic tumor cell death is radiofrequency ablation (RFA), which converts radiofrequency waves into heat to achieve local temperatures sufficient for tissue destruction.97 A clinical study of 20 patients with hepatocellular carcinoma (HCC) demonstrated a significant increase in circulating T-cell ELISPOT responses to tumor tissue following RFA treatment and an increase in circulating natural killer (NK) cells (CD3CD56+).98 Another trial using PBMCs from 19 patients with HCC demonstrated that RFA significantly increased maturation markers (CD83, CD86) of activated monocytes and monocyte-derived DCs.99 Further, exposure of patient-derived, HCC-specific T-cell lines (CD4+ or CD8+) to RFA-treated HCC tumor increased T-cell IFNγ production. This could be due to increased antigen presentation, increased expression of costimulatory molecules, and/or reduced immunosuppression by the tumor cells. A clinical trial of 14 lung cancer patients undergoing RFA demonstrated a significant increase in total white blood cells three days post-RFA, a significant increase in proinflammatory plasma cytokines and chemokines three days post-RFA, and a significant decrease in circulating regulatory T-cells (Tregs) (CD4+CD25 hiCD69) 30 d post-RFA.100 Overall, these clinical reports demonstrate that RFA can enhance DC maturation, increase frequency of tumor-specific T-cells, and reduce Treg numbers, making RFA and immunotherapy an appealing combinational approach for cancer therapy.101

Cryoablation is a type of ablative therapy that dates back to the 1840s.102 Early clinical studies using cryoablation demonstrated regression of untreated metastatic lesions and indications of antitumor immunity.103106 More recent clinical studies have demonstrated increases in serum cytokines and generation of tumor-specific T-cells following cryoabaltion.107,108 The simplicity of cryoablation, coupled with minimal negative effects on the immune system, make this procedure an attractive candidate for combination with immunotherapy.

High-Intensity Focused Ultrasound

High-intensity focused ultrasound (HIFU) is a recently developed and promising technique that uses a transducer to generate high-frequency ultrasound waves focused on a single point in the tumor. HIFU can deliver a large amount of energy that is subsequently converted to thermal energy, resulting in tumor cell destruction with minimal damage to surrounding tissue.109 HIFU is receiving increasing interest for the clinical management of patients with various types of cancer, including prostate, breast, liver, pancreas, kidney, bone and soft tissue.110,111

Wu et al. investigated whether tumor antigens expressed on breast cancer cells are preserved after HIFU treatment and could serve as a potential source of antigens to stimulate the immune system.112 Primary lesions in 23 patients with biopsy-proven breast cancer were treated with HIFU and then analyzed for changes in expression of tumor antigen. Results demonstrated that several tumor antigens remained in the tumor debris after HIFU ablation, potentially indicating the generation of a TAA source for DCs. A more recent study investigated the status and function of TILs after HIFU ablation of breast cancer.113 In this trial, 48 patients were randomized to receive HIFU prior to modified radical mastectomy or surgery alone. Compared to surgery alone, treatment with HIFU and surgery significantly increased tumor-infiltrating T-cells, B-cells and NK cells. Additionally, HIFU treatment significantly increased Fas-ligand+, granzyme+ and perforin+ TILs compared to surgery alone, indicating the enhanced ability of lymphocytes to induce apoptosis or cytolytic death of tumor cells. These studies suggest that HIFU-treated tumor can serve as a source of tumor antigens, and that HIFU can increase the number and function of TILs.

Nonsteroidal Anti-Inflammatory Drugs

Nonsteroidal anti-inflammatory drugs (NSAIDs) have been gaining significant interest among oncologists as more is learned about their potential antitumor effects. A growing body of data suggests that NSAIDs can interfere with complex processes related to tumorigenesis, angiogenesis, metastasis and inhibition of apoptosis.114116 Attention has focused on cyclooxygenase-2 inhibitors, including celecoxib (Celebrex®) and indomethacin, to confirm their antiangiogenic and proapoptotic functions, as well as to determine their ability to initiate tumor-specific immune responses.

One study treated colorectal carcinoma patients preoperatively with oral NSAIDs and evaluated MHC and TIL gene expression.117 In this study, genes belonging to the MHC locus, including those responsible for peptide loading, antigen presentation and cytolytic activity, were upregulated in response to short-term preoperative NSAID treatment. Furthermore, NSAID pretreatment increased infiltration of CD4+ and CD8+ T-cells in the tumor stroma and decreased expression of molecules associated with immunosuppressive Tregs (FOXP3, IL-10). Similar findings were noted in a study that investigated the activity of celecoxib in patients with newly diagnosed, untreated nasopharyngeal carcinoma.118 Tumor biopsies were obtained before and after a 14-d course of celecoxib. Results of this study showed MHC class II DMB increased expression two-fold, the greatest change in expression level of any of the 35 genes examined. Clearly, utilizing NSAIDs in cancer treatment regimens is still in the beginning stages. However, the ability of these drugs to inhibit angiogenesis, induce apoptosis and upregulate genes crucial for initiating an immune response may enhance the function and antitumor effects of other immunotherapeutic modalities.

Regulatory T-cells

The ability of tumors to evade immune system surveillance enables tumor cells to grow and survive. CD4+CD25+FOXP3+ Tregs are important suppressors of active tumor-specific immune responses in cancer.119 Depleting Tregs and reducing their functionality has been shown to improve endogenous antitumor immunity and the efficacy of active immunotherapy in animal models for cancer, suggesting that inhibiting Treg function could also improve on the limited successes of immunotherapy in human cancers.119122 Denileukin diftitox (Ontak®) is a recombinant fusion protein consisting of IL-2 and diphtheria toxin targeting IL-2 receptor (CD25). It has been approved by the FDA to treat cutaneous T-cell leukemia/lymphoma, which is characterized by large numbers of malignant CD4+CD25+ T-cells.123,124 Based on the antitumor effects of Treg reduction in leukemia/lymphoma patients, Curiel et al. designed a Phase 0/I clinical trial in patients with any advanced-stage epithelial carcinoma to test if denileukin diftitox would reduce Tregs, and thereby reduce immune suppression and boost antitumor immunity in these patient populations.125 Results of this study demonstrated that denileukin diftitox decreased the number of circulating Tregs and was associated with improved cellular immunity in patients with ovarian, lung, breast or pancreatic cancer. Additional reports confirmed that denileukin diftitox reduced Treg numbers and improved tumor-specific T-cell responses in individuals with renal cell carcinoma and melanoma.126,127 Importantly, the enhanced anti-tumor immune responses following Treg reduction also resulted in regression of melanoma metastases, confirming clinical benefit.126 Numerous clinical trials are currently recruiting patients to further evaluate the clinical applicability of reducing Tregs, including Phase II trials in patients with metastatic pancreatic cancer, ovarian cancer, and advanced refractory breast cancer.128130 These and other studies will demonstrate how reducing Tregs can enhance current cancer vaccination strategies.

Daclizumab is an mAb targeting CD25 that was FDA-approved in 1997 for renal allograft rejection.131 This antibody blocks interaction of IL-2 with CD25, preventing T-cell proliferation, and thus could have a potential role in the treatment of T-cell leukemia or lymphoma.132 Recently, the combination of daclizumab and cytotoxic depsipeptide significantly increased the survival of mice bearing T-cell leukemia tumors.133 This demonstrates another approach to targeting CD25 for combinational cancer immunotherapy.

Cyclophosphamide is a cytotoxic drug with demonstrated ability to reduce lymphocyte populations without being myeloablative.134 Clinical studies have also demonstrated that low metronomic doses of cyclophosphamide can reduce Treg populations without reducing CD8+ T-cell populations.135 However, single high doses of cyclophosphamide appear to be ineffective for Treg reduction.136 Thus, proper dosing and scheduling of cyclophosphamide treatment in combination with cancer vaccination could likely enhance the generation of tumor-specific cytolytic immunity.

Conclusions

Initial steps in the development of cancer vaccines have led to the identification of TAAs and the development of novel vaccine strategies. Elucidation of the mechanisms involved in antigen recognition, processing and presentation by APCs, along with identification of signals that activate or suppress T-cells, has led to the discovery and exploitation of immunomodulators to enhance the generation of tumor-specific immunity. Combining cancer vaccines with traditional cytotoxic agents or radiation therapy provides an opportunity to improve patient survival and quality of life. Additional knowledge acquired from basic, translational and clinical studies will most likely translate the science of tumor immunology into clinically relevant modalities for a range of cancers.

Abbreviations:

APC

antigen-presenting cell

BCG

bacillus calmette-guerin

CEA

carcinoembryonic antigen

CTL

cytotoxic T lymphocyte

DC

dendritic cell

FDA

Food and Drug Administration

FOLFIRI

folinic acid/fluorouracil/irinotecan

GM-CSF

granulocyte-macrophage colony-stimulating factor

HCC

hepatocellular carcinoma

HIFU

high-intensity focused ultrasound

ICAM

intercellular adhesion molecule

LFA

lymphocyte function-associated antigen

LPS

lipopolysaccharide

mAb

monoclonal antibody

mDC

myeloid-derived dendritic cell

MHC

major histocompatibility complex

MPL

monophosphoryl lipid

MUC

mucin

MVA

modified vaccinia ankara

NK

natural killer

NSAID

nonsteroidal anti-inflammatory drug

NSCLC

non-small cell lung cancer

PAP

prostatic acid phosphatase

PBMC

peripheral blood mononuclear cell

pDC

plasmacytoid dendritic cell

PSA

prostate-specific antigen

PSMA

prostate-specific membrane antigen

rF

recombinant fowlpox

RFA

radiofrequency ablation

rV

recombinant vaccinia

TAA

tumor-associated antigen

TIL

tumor-infiltrating lymphocyte

TLR

toll-like receptor

Treg

regulatory T-cell

References

  • 1.Wiemann B, Starnes CO. Coley’s toxins, tumor necrosis factor and cancer research: a historical perspective. Pharmacol Ther 1994; 64:529–64. [DOI] [PubMed] [Google Scholar]
  • 2.Schlichtholz B, Legros Y, Gillet D, Gaillard C, Marty M, Lane D, et al. The immune response to p53 in breast cancer patients is directed against immunodominant epitopes unrelated to the mutational hot spot. Cancer Res 1992; 52:6380–4. [PubMed] [Google Scholar]
  • 3.De Plaen E, Lurquin C, Van Pel A, Mariame B, Szikora JP, Wolfel T, et al. Immunogenic (tum-) variants of mouse tumor P815: cloning of the gene of tum-antigen P91A and identification of the tum-mutation. Proc Natl Acad Sci USA 1988; 85:2274–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Mandelboim O, Berke G, Fridkin M, Feldman M, Eisenstein M, Eisenbach L. CTL induction by a tumour-associated antigen octapeptide derived from a murine lung carcinoma. Nature 1994; 369:67–71. [DOI] [PubMed] [Google Scholar]
  • 5.Sahin U, Tureci O, Schmitt H, Cochlovius B, Johannes T, Schmits R, et al. Human neoplasms elicit multiple specific immune responses in the autologous host. Proc Natl Acad Sci USA 1995; 92:11810–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Viatte S, Alves PM, Romero P. Reverse immunology approach for the identification of CD8 T-cell-defined antigens: advantages and hurdles. Immunol Cell Biol 2006; 84:318–30. [DOI] [PubMed] [Google Scholar]
  • 7.Terabe M, Berzofsky JA. The role of NKT-cells in tumor immunity. Adv Cancer Res 2008; 101:277–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hofmeister V, Schrama D, Becker JC. Anti-cancer therapies targeting the tumor stroma. Cancer Immunol Immunother 2008; 57:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Maccalli C, Scaramuzza S, Parmiani G. TNK cells (NKG2D+ CD8+ or CD4+ T lymphocytes) in the control of human tumors. Cancer Immunol Immunother 2009; 58:801–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Fournier P, Schirrmacher V. Randomized clinical studies of anti-tumor vaccination: state of the art in 2008. Expert Rev Vaccines 2009; 8:51–66. [DOI] [PubMed] [Google Scholar]
  • 11.Li G, Ali SA, McArdle SE, Mian S, Ahmad M, Miles A, et al. Immunity to tumour antigens. Curr Pharm Des 2005; 11:3501–9. [DOI] [PubMed] [Google Scholar]
  • 12.Dougan M, Dranoff G. Immune therapy for cancer. Annu Rev Immunol 2009; 27:83–117. [DOI] [PubMed] [Google Scholar]
  • 13.Lans TE, Grunhagen DJ, de Wilt JH, van Geel AN, Eggermont AM. Isolated limb perfusions with tumor necrosis factor and melphalan for locally recurrent soft tissue sarcoma in previously irradiated limbs. Ann Surg Oncol 2005; 12:406–11. [DOI] [PubMed] [Google Scholar]
  • 14.Motzer RJ, Bacik J, Murphy BA, Russo P, Mazumdar M. Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. J Clin Oncol 2002; 20:289–96. [DOI] [PubMed] [Google Scholar]
  • 15.Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999; 17:2105–16. [DOI] [PubMed] [Google Scholar]
  • 16.Fagerberg J Granulocyte-macrophage colony-stimulating factor as an adjuvant in tumor immunotherapy. Med Oncol 1996; 13:155–60. [PubMed] [Google Scholar]
  • 17.Allison AC. The mode of action of immunological adjuvants. Dev Biol Stand 1998; 92:3–11. [PubMed] [Google Scholar]
  • 18.Coronel A, Boyer A, Franssen JD, Romet-Lemonne JL, Fridman WH, Teillaud JL. Cytokine production and T-cell activation by macrophage-dendritic cells generated for therapeutic use. Br J Haematol 2001; 114:671–80. [DOI] [PubMed] [Google Scholar]
  • 19.Harzstark AL, Small EJ. Sipuleucel-T for the treatment of prostate cancer. Drugs Today (Barc) 2008; 44:271–8. [DOI] [PubMed] [Google Scholar]
  • 20.Small EJ, Schellhammer PF, Higano CS, Redfern CH, Nemunaitis JJ, Valone FH, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol 2006; 24:3089–94. [DOI] [PubMed] [Google Scholar]
  • 21.Herper M Data Shows Provenge Extends Cancer Survival. Available from: http://www.forbes.com/2009/04/28/dendreon-provenge-cancer-business-healthcare-dendreon.html.
  • 22.Provenge® (Sipuleucel-T) Active Cellular Immunotherapy Treatment of Metastatic Prostate Cancer After Failing Hormone Therapy. Available from: http://www.clinicaltrials.gov/ct2/results?term=NCT00065442.
  • 23.Bendandi M, Gocke CD, Kobrin CB, Benko FA, Sternas LA, Pennington R, et al. Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma. Nat Med 1999; 5:1171–7. [DOI] [PubMed] [Google Scholar]
  • 24.Study of the BiovaxId Tumor Derived Idiotype Vaccine in Patients With Follicular Lymphoma (BiovaxID). Available from: http://www.clinicaltrials.gov/ct2/results?term=NCT00091676+.
  • 25.Ullenhag GJ, Frodin JE, Jeddi-Tehrani M, Strigard K, Eriksson E, Samanci A, et al. Durable carcinoembryonic antigen (CEA)-specific humoral and cellular immune responses in colorectal carcinoma patients vaccinated with recombinant CEA and granulocyte/macrophage colony-stimulating factor. Clin Cancer Res 2004; 10:3273–81. [DOI] [PubMed] [Google Scholar]
  • 26.Ward JE, McNeel DG. GVAX: an allogeneic, whole-cell, GM-CSF-secreting cellular immunotherapy for the treatment of prostate cancer. Expert Opin Biol Ther 2007; 7:1893–902. [DOI] [PubMed] [Google Scholar]
  • 27.Higano CS, Corman JM, Smith DC, Centeno AS, Steidle CP, Gittleman M, et al. Phase 1/2 dose-escalation study of a GM-CSF-secreting, allogeneic, cellular immunotherapy for metastatic hormone-refractory prostate cancer. Cancer 2008; 113:975–84. [DOI] [PubMed] [Google Scholar]
  • 28.Docetaxel in Combination With GVAX® Immunotherapy Versus Docetaxel and Prednisone in Prostate Cancer Patients. Available from: www.clinicaltrials.gov/ct2/results?term=NCT00133224+.
  • 29.GVAX® Vaccine for Prostate Cancer vs Docetaxel & Prednisone in Patients With Metastatic Hormone-Refractory Prostate Cancer. Available from: www.clinicaltrials.gov/ct2/results?term=NCT00089856.
  • 30.Parmiani G, Castelli C, Pilla L, Santinami M, Colombo MP, Rivoltini L. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann Oncol 2007; 18:226–32. [DOI] [PubMed] [Google Scholar]
  • 31.Rosenberg SA, Sportes C, Ahmadzadeh M, Fry TJ, Ngo LT, Schwarz SL, et al. IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. J Immunother 2006; 29:313–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Daud AI, DeConti RC, Andrews S, Urbas P, Riker AI, Sondak VK, et al. Phase I trial of interleukin-12 plasmid electroporation in patients with metastatic melanoma. J Clin Oncol 2008; 26:5896–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T, et al. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res 2009; 69:4010–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Davis ID, Brady B, Kefford RF, Millward M, Cebon J, Skrumsager BK, et al. Clinical and biological efficacy of recombinant human interleukin-21 in patients with stage IV malignant melanoma without prior treatment: a phase IIa trial. Clin Cancer Res 2009; 15:2123–9. [DOI] [PubMed] [Google Scholar]
  • 35.Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 1995; 13:688–96. [DOI] [PubMed] [Google Scholar]
  • 36.Letourneau S, Krieg C, Pantaleo G, Boyman O. IL-2- and CD25-dependent immunoregulatory mechanisms in the homeostasis of T-cell subsets. J Allergy Clin Immunol 2009; 123:758–62. [DOI] [PubMed] [Google Scholar]
  • 37.Dudek AZ, Mescher MF, Okazaki I, Math VT, Luo X, Curtsinger JM, et al. Autologous large multivalent immunogen vaccine in patients with metastatic melanoma and renal cell carcinoma. Am J Clin Oncol 2008; 31:173–81. [DOI] [PubMed] [Google Scholar]
  • 38.Elias EG, Zapas JL, McCarron EC, Beam SL, Hasskamp JH, Culpepper WJ. Sequential administration of GM-CSF (Sargramostim) and IL-2 +/− autologous vaccine as adjuvant therapy in cutaneous melanoma: an interim report of a phase II clinical trial. Cancer Biother Radiopharm 2008; 23:285–91. [DOI] [PubMed] [Google Scholar]
  • 39.Sosman JA, Carrillo C, Urba WJ, Flaherty L, Atkins MB, Clark JI, et al. Three phase II cytokine working group trials of gp100 (210M) peptide plus high-dose interleukin-2 in patients with HLA-A2-positive advanced melanoma. J Clin Oncol 2008; 26:2292–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Acres B, Bonnefoy JY. Clinical development of MVA-based therapeutic cancer vaccines. Expert Rev Vaccines 2008; 7:889–93. [DOI] [PubMed] [Google Scholar]
  • 41.Dreicer R, Stadler WM, Ahmann FR, Whiteside T, Bizouarne N, Acres B, et al. MVA-MUC1-IL2 vaccine immunotherapy (TG4010) improves PSA doubling time in patients with prostate cancer with biochemical failure. Invest New Drugs 2009. 4:379–86. [DOI] [PubMed] [Google Scholar]
  • 42.Tran KQ, Zhou J, Durflinger KH, Langhan MM, Shelton TE, Wunderlich JR, et al. Minimally cultured tumor-infiltrating lymphocytes display optimal characteristics for adoptive cell therapy. J Immunother 2008; 31:742–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ward RC, Kaufman HL. Targeting costimulatory pathways for tumor immunotherapy. Int Rev Immunol 2007; 26:161–96. [DOI] [PubMed] [Google Scholar]
  • 44.Dols A, Smith JW 2nd, Meijer SL, Fox BA, Hu HM, Walker E, et al. Vaccination of women with metastatic breast cancer, using a costimulatory gene (CD80)-modified, HLA-A2-matched, allogeneic, breast cancer cell line: clinical and immunological results. Hum Gene Ther 2003; 14:1117–23. [DOI] [PubMed] [Google Scholar]
  • 45.Kaufman HL, Lenz HJ, Marshall J, Singh D, Garett C, Cripps C, et al. Combination chemotherapy and ALVAC-CEA/B7.1 vaccine in patients with metastatic colorectal cancer. Clin Cancer Res 2008; 14:4843–9. [DOI] [PubMed] [Google Scholar]
  • 46.Kaufman HL, Deraffele G, Mitcham J, Moroziewicz D, Cohen SM, Hurst-Wicker KS, et al. Targeting the local tumor microenvironment with vaccinia virus expressing B7.1 for the treatment of melanoma. J Clin Invest 2005; 115:1903–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Arlen PM, Gulley JL, Parker C, Skarupa L, Pazdur M, Panicali D, et al. A randomized phase II study of concurrent docetaxel plus vaccine versus vaccine alone in metastatic androgen-independent prostate cancer. Clin Cancer Res 2006; 12:1260–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Arlen PM, Skarupa L, Pazdur M, Seetharam M, Tsang KY, Grosenbach DW, et al. Clinical safety of a viral vector based prostate cancer vaccine strategy. J Urol 2007; 178:1515–20. [DOI] [PubMed] [Google Scholar]
  • 49.Hodge JW, Chakraborty M, Kudo-Saito C, Garnett CT, Schlom J. Multiple costimulatory modalities enhance CTL avidity. J Immunol 2005; 174:5994–6004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gulley JL, Arlen PM, Tsang KY, Yokokawa J, Palena C, Poole DJ, et al. Pilot study of vaccination with recombinant CEA-MUC-1-TRICOM poxviral-based vaccines in patients with metastatic carcinoma. Clin Cancer Res 2008; 14:3060–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Weber J Ipilimumab: controversies in its development, utility and autoimmune adverse events. Cancer Immunol Immunother 2009; 58:823–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Weber JS, O’Day S, Urba W, Powderly J, Nichol G, Yellin M, et al. Phase I/II study of ipilimumab for patients with metastatic melanoma. J Clin Oncol 2008; 26:5950–6. [DOI] [PubMed] [Google Scholar]
  • 53.Fong L, Kwek SS, O’Brien S, Kavanagh B, McNeel DG, Weinberg V, et al. Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res 2009; 69:609–15. [DOI] [PubMed] [Google Scholar]
  • 54.Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci USA 2003; 100:8372–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tong AW, Stone MJ. Prospects for CD40-directed experimental therapy of human cancer. Cancer Gene Ther 2003; 10:1–13. [DOI] [PubMed] [Google Scholar]
  • 56.Eliopoulos AG, Davies C, Knox PG, Gallagher NJ, Afford SC, Adams DH, et al. CD40 induces apoptosis in carcinoma cells through activation of cytotoxic ligands of the tumor necrosis factor superfamily. Mol Cell Biol 2000; 20:5503–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Vonderheide RH, Flaherty KT, Khalil M, Stumacher MS, Bajor DL, Hutnick NA, et al. Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. J Clin Oncol 2007; 25:876–83. [DOI] [PubMed] [Google Scholar]
  • 58.Medzhitov R, Janeway C Jr. Innate immunity. N Engl J Med 2000; 343:338–44. [DOI] [PubMed] [Google Scholar]
  • 59.Meyer T, Stockfleth E. Clinical investigations of toll-like receptor agonists. Expert Opin Investig Drugs 2008; 17:1051–65. [DOI] [PubMed] [Google Scholar]
  • 60.Vidal D Topical imiquimod: mechanism of action and clinical applications. Mini Rev Med Chem 2006; 6:499–503. [DOI] [PubMed] [Google Scholar]
  • 61.Didona B, Benucci R, Amerio P, Canzona F, Rienzo O, Cavalieri R. Primary cutaneous CD30+ T-cell lymphoma responsive to topical imiquimod (Aldara). Br J Dermatol 2004; 150:1198–201. [DOI] [PubMed] [Google Scholar]
  • 62.Schon MP, Schon M. The small-molecule immune response modifier imiquimod—its mode of action and clinical use in the treatment of skin cancer. Expert Opin Ther Targets 2006; 10:69–76. [DOI] [PubMed] [Google Scholar]
  • 63.Stary G, Bangert C, Tauber M, Strohal R, Kopp T, Stingl G. Tumoricidal activity of TLR7/8-activated inflammatory dendritic cells. J Exp Med 2007; 204:1441–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Guiducci C, Coffman RL, Barrat FJ. Signalling pathways leading to IFNalpha production in human plasmacytoid dendritic cell and the possible use of agonists or antagonists of TLR7 and TLR9 in clinical indications. J Intern Med 2009; 265:43–57. [DOI] [PubMed] [Google Scholar]
  • 65.Shackleton M, Davis ID, Hopkins W, Jackson H, Dimopoulos N, Tai T, et al. The impact of imiquimod, a Toll-like receptor-7 ligand (TLR7L), on the immunogenicity of melanoma peptide vaccination with adjuvant Flt3 ligand. Cancer Immun 2004; 4:9. [PubMed] [Google Scholar]
  • 66.Adams S, O’Neill DW, Nonaka D, Hardin E, Chiriboga L, Siu K, et al. Immunization of malignant melanoma patients with full-length NY-ESO-1 protein using TLR7 agonist imiquimod as vaccine adjuvant. J Immunol 2008; 181:776–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, et al. Novel multi-peptide vaccination in HLA-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate 2009; 69:917–27. [DOI] [PubMed] [Google Scholar]
  • 68.Jurk M, Vollmer J. Therapeutic applications of synthetic CpG oligodeoxynucleotides as TLR9 agonists for immune modulation. Bio Drugs 2007; 21:387–401. [DOI] [PubMed] [Google Scholar]
  • 69.Manegold C, Gravenor D, Woytowitz D, Mezger J, Hirsh V, Albert G, et al. Randomized phase II trial of a toll-like receptor 9 agonist oligodeoxynucleotide, PF-3512676, in combination with first-line taxane plus platinum chemotherapy for advanced-stage non-small-cell lung cancer. J Clin Oncol 2008; 26:3979–86. [DOI] [PubMed] [Google Scholar]
  • 70.Coley Pharma announces Pfizer’s discontinuation of clinical trials for PF-3512676. Available from: www.reuters.com/article/inPlayBriefing/idUSIN20070620135825COLY20070620.
  • 71.GSK1572932A antigen-specific cancer immunotherapeutic as adjuvant therapy in patients with non-small cell lung cancer. Available from: www.clinicaltrials.gov/ct2/results?term=NCT00480025.
  • 72.Vansteenkiste J, Zielinski M, Linder A, Dahabre J, Esteban E, Malinowski W, et al. Final results of a multi-center, double-blind, randomized, placebo-controlled phase II study to assess the efficacy of MAGE-A3 immunotherapeutic as adjuvant therapy in stage IB/II non-small cell lung cancer (NSCLC). J Clin Oncol 2007; 25:7554. [Google Scholar]
  • 73.Vansteenkiste J, Zielinski M, Dahabre J, Linder A, Lehmann F, Gruselle O, et al. Association of gene expression signature and clinical efficacy of MAGE-A3 antigen-specific cancer immunotherapeutic (ASCI) as adjuvant therapy in resected stage IB/II non-small cell lung cancer (NSCLC). J Clin Oncol 2008; 26:7501. [Google Scholar]
  • 74.Kanodia S, Kast WM. Peptide-based vaccines for cancer: realizing their potential. Expert Rev Vaccines 2008; 7:1533–45. [DOI] [PubMed] [Google Scholar]
  • 75.Valmori D, Souleimanian NE, Tosello V, Bhardwaj N, Adams S, O’Neill D, et al. Vaccination with NY-ESO-1 protein and CpG in montanide induces integrated antibody/Th1 responses and CD8 T-cells through cross-priming. Proc Natl Acad Sci USA 2007; 104:8947–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Fourcade J, Kudela P, Andrade Filho PA, Janjic B, Land SR, Sander C, et al. Immunization with analog peptide in combination with CpG and montanide expands tumor antigen-specific CD8+ T-cells in melanoma patients. J Immunother 2008; 31:781–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ribi E, Cantrell JL, Takayama K, Qureshi N, Peterson J, Ribi HO. Lipid A and immunotherapy. Rev Infect Dis 1984; 6:567–72. [DOI] [PubMed] [Google Scholar]
  • 78.Baldridge JR, McGowan P, Evans JT, Cluff C, Mossman S, Johnson D, et al. Taking a Toll on human disease: Toll-like receptor 4 agonists as vaccine adjuvants and monotherapeutic agents. Expert Opin Biol Ther 2004; 4:1129–38. [DOI] [PubMed] [Google Scholar]
  • 79.Butts C, Murray N, Maksymiuk A, Goss G, Marshall E, Soulieres D, et al. Randomized phase IIB trial of BLP25 liposome vaccine in stage IIIB and IV non-small-cell lung cancer. J Clin Oncol 2005; 23:6674–81. [DOI] [PubMed] [Google Scholar]
  • 80.Cancer vaccine study for unresectable stage III non-small cell lung cancer. Available from: www.clinicaltrials.gov/ct2/results?term=NCT00409188.
  • 81.Baxevanis CN, Perez SA, Papamichail M. Combinatorial treatments including vaccines, chemotherapy and monoclonal antibodies for cancer therapy. Cancer Immunol Immunother 2009; 58:317–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Nowak AK, Lake RA, Marzo AL, Scott B, Heath WR, Collins EJ, et al. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T-cells. J Immunol 2003; 170:4905–13. [DOI] [PubMed] [Google Scholar]
  • 83.Nowak AK, Robinson BW, Lake RA. Synergy between chemotherapy and immunotherapy in the treatment of established murine solid tumors. Cancer Res 2003; 63:4490–6. [PubMed] [Google Scholar]
  • 84.Tanaka F, Yamaguchi H, Ohta M, Mashino K, Sonoda H, Sadanaga N, et al. Intratumoral injection of dendritic cells after treatment of anticancer drugs induces tumor-specific antitumor effect in vivo. Int J Cancer 2002; 101:265–9. [DOI] [PubMed] [Google Scholar]
  • 85.Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med 2005; 202:1691–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Robbins PF, Dudley ME, Wunderlich J, El-Gamil M, Li YF, Zhou J, et al. Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J Immunol 2004; 173:7125–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Muranski P, Boni A, Wrzesinski C, Citrin DE, Rosenberg SA, Childs R, et al. Increased intensity lymphodepletion and adoptive immunotherapy—how far can we go? Nat Clin Pract Oncol 2006; 3:668–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Antonia SJ, Mirza N, Fricke I, Chiappori A, Thompson P, Williams N, et al. Combination of p53 cancer vaccine with chemotherapy in patients with extensive stage small cell lung cancer. Clin Cancer Res 2006; 12:878–87. [DOI] [PubMed] [Google Scholar]
  • 89.Hodge JW, Guha C, Neefjes J, Gulley JL. Synergizing radiation therapy and immunotherapy for curing incurable cancers. Opportunities and challenges. Oncology 2008; 22:1064–70. [PMC free article] [PubMed] [Google Scholar]
  • 90.Demaria S, Formenti SC. Sensors of ionizing radiation effects on the immunological microenvironment of cancer. Int J Radiat Biol 2007; 83:819–25. [DOI] [PubMed] [Google Scholar]
  • 91.Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 2007; 13:54–61. [DOI] [PubMed] [Google Scholar]
  • 92.Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med 2006; 203:1259–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Garnett CT, Palena C, Chakraborty M, Tsang KY, Schlom J, Hodge JW. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res 2004; 64:7985–94. [DOI] [PubMed] [Google Scholar]
  • 94.Demaria S, Santori FR, Ng B, Liebes L, Formenti SC, Vukmanovic S. Select forms of tumor cell apoptosis induce dendritic cell maturation. J Leukoc Biol 2005; 77:361–8. [DOI] [PubMed] [Google Scholar]
  • 95.Gulley JL, Arlen PM, Bastian A, Morin S, Marte J, Beetham P, et al. Combining a recombinant cancer vaccine with standard definitive radiotherapy in patients with localized prostate cancer. Clin Cancer Res 2005; 11:3353–62. [DOI] [PubMed] [Google Scholar]
  • 96.Chi KH, Liu SJ, Li CP, Kuo HP, Wang YS, Chao Y, et al. Combination of conformal radiotherapy and intratumoral injection of adoptive dendritic cell immunotherapy in refractory hepatoma. J Immunother 2005; 28:129–35. [DOI] [PubMed] [Google Scholar]
  • 97.Liu LX, Zhang WH, Jiang HC. Current treatment for liver metastases from colorectal cancer. World J Gastroenterol 2003; 9:193–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zerbini A, Pilli M, Penna A, Pelosi G, Schianchi C, Molinari A, et al. Radiofrequency thermal ablation of hepatocellular carcinoma liver nodules can activate and enhance tumor-specific T-cell responses. Cancer Res 2006; 66:1139–46. [DOI] [PubMed] [Google Scholar]
  • 99.Zerbini A, Pilli M, Fagnoni F, Pelosi G, Pizzi MG, Schivazappa S, et al. Increased immunostimulatory activity conferred to antigen-presenting cells by exposure to antigen extract from hepatocellular carcinoma after radiofrequency thermal ablation. J Immunother 2008; 31:271–82. [DOI] [PubMed] [Google Scholar]
  • 100.Fietta AM, Morosini M, Passadore I, Cascina A, Draghi P, Dore R, et al. Systemic inflammatory response and downmodulation of peripheral CD25(+)Foxp3(+) T-regulatory cells in patients undergoing radiofrequency thermal ablation for lung cancer. Hum Immunol 2009; Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 101.Fagnoni FF, Zerbini A, Pelosi G, Missale G. Combination of radiofrequency ablation and immunotherapy. Front Biosci 2008; 13:369–81. [DOI] [PubMed] [Google Scholar]
  • 102.Sabel MS. Cryo-immunology: a review of the literature and proposed mechanisms for stimulatory versus suppressive immune responses. Cryobiology 2009; 58:1–11. [DOI] [PubMed] [Google Scholar]
  • 103.Alblin RJ, Soanes WA, Gonder MJ. Prospects for cryo-immunotherapy in cases of metastasizing carcinoma of the prostate. Cryobiology 1971; 8:271–9. [DOI] [PubMed] [Google Scholar]
  • 104.Ablin RJ, Guinan PD, Bruns GR, Sheik HI, Sadoughi N, Bush IM. Evaluation of cellular immunologic responsiveness in the clinical management of patients with prostatic cancer II. Effect of oestrogen, cryosurgery and transurethral resection on thymic-dependent lymphocytic blastogenesis. Urol Int 1976; 31:383–400. [DOI] [PubMed] [Google Scholar]
  • 105.Ablin RJ, Bruns GR, Guinan PD, Sadoughi N, Sheik HA, Bush IM. Alterations in host responsiveness in patients with prostatic cancer following cryosurgery or transurethral resection. Am Surg 1977; 43:144–50. [PubMed] [Google Scholar]
  • 106.Weyer U, Petersen I, Ehrke C, Carstensen A, Nussgen A, Russ C, et al. Immunomodulation by cryosurgery in malignant melanoma. Onkologie 1989; 12:291–6. [DOI] [PubMed] [Google Scholar]
  • 107.Osada S, Imai H, Tomita H, Tokuyama Y, Okumura N, Matsuhashi N, et al. Serum cytokine levels in response to hepatic cryoablation. J Surg Oncol 2007; 95:491–8. [DOI] [PubMed] [Google Scholar]
  • 108.Si T, Guo Z, Hao X. Immunologic response to primary cryoablation of high-risk prostate cancer. Cryobiology 2008; 57:66–71. [DOI] [PubMed] [Google Scholar]
  • 109.Zhang HG, Mehta K, Cohen P, Guha C. Hyperthermia on immune regulation: a temperature’s story. Cancer Lett 2008; 271:191–204. [DOI] [PubMed] [Google Scholar]
  • 110.Kennedy JE. High-intensity focused ultrasound in the treatment of solid tumours. Nat Rev Cancer 2005; 5:321–7. [DOI] [PubMed] [Google Scholar]
  • 111.Wu F Extracorporeal high intensity focused ultrasound in the treatment of patients with solid malignancy. Minim Invasive Ther Allied Technol 2006; 15:26–35. [DOI] [PubMed] [Google Scholar]
  • 112.Wu F, Wang ZB, Cao YD, Zhou Q, Zhang Y, Xu ZL, et al. Expression of tumor antigens and heat-shock protein 70 in breast cancer cells after high-intensity focused ultrasound ablation. Ann Surg Oncol 2007; 14:1237–42. [DOI] [PubMed] [Google Scholar]
  • 113.Lu P, Zhu XQ, Xu ZL, Zhou Q, Zhang J, Wu F. Increased infiltration of activated tumor-infiltrating lymphocytes after high intensity focused ultrasound ablation of human breast cancer. Surgery 2009; 145:286–93. [DOI] [PubMed] [Google Scholar]
  • 114.Wang D, Dubois RN. Prostaglandins and cancer. Gut 2006; 55:115–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Trifan OC, Hla T. Cyclooxygenase-2 modulates cellular growth and promotes tumori-genesis. J Cell Mol Med 2003; 7:207–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Soslow RA, Dannenberg AJ, Rush D, Woerner BM, Khan KN, Masferrer J, et al. COX-2 is expressed in human pulmonary, colonic and mammary tumors. Cancer 2000; 89:2637–45. [DOI] [PubMed] [Google Scholar]
  • 117.Lonnroth C, Andersson M, Arvidsson A, Nordgren S, Brevinge H, Lagerstedt K, et al. Preoperative treatment with a non-steroidal anti-inflammatory drug (NSAID) increases tumor tissue infiltration of seemingly activated immune cells in colorectal cancer. Cancer Immun 2008; 8:5. [PMC free article] [PubMed] [Google Scholar]
  • 118.Soo RA, Wu J, Aggarwal A, Tao Q, Hsieh W, Putti T, et al. Celecoxib reduces microvessel density in patients treated with nasopharyngeal carcinoma and induces changes in gene expression. Ann Oncol 2006; 17:1625–30. [DOI] [PubMed] [Google Scholar]
  • 119.Ruter J, Barnett BG, Kryczek I, Brumlik MJ, Daniel BJ, Coukos G, et al. Altering regulatory T-cell function in cancer immunotherapy: a novel means to boost the efficacy of cancer vaccines. Front Biosci 2009; 14:1761–70. [DOI] [PubMed] [Google Scholar]
  • 120.Tanaka H, Tanaka J, Kjaergaard J, Shu S. Depletion of CD4+ CD25+ regulatory cells augments the generation of specific immune T-cells in tumor-draining lymph nodes. J Immunother 2002; 25:207–17. [DOI] [PubMed] [Google Scholar]
  • 121.Steitz J, Bruck J, Lenz J, Knop J, Tuting T. Depletion of CD25(+) CD4(+) T-cells and treatment with tyrosinase-related protein 2-transduced dendritic cells enhance the interferon alpha-induced, CD8(+) T-cell-dependent immune defense of B16 melanoma. Cancer Res 2001; 61:8643–6. [PubMed] [Google Scholar]
  • 122.Sutmuller RP, van Duivenvoorde LM, van Elsas A, Schumacher TN, Wildenberg ME, Allison JP, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T-cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med 2001; 194:823–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Tiemessen MM, Mitchell TJ, Hendry L, Whittaker SJ, Taams LS, John S. Lack of suppressive CD4+CD25+FOXP3+ T-cells in advanced stages of primary cutaneous T-cell lymphoma. J Invest Dermatol 2006; 126:2217–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Foss FM. DAB(389)IL-2 (ONTAK): a novel fusion toxin therapy for lymphoma. Clin Lymphoma 2000; 1:110–6. [DOI] [PubMed] [Google Scholar]
  • 125.Barnett B, Kryczek I, Cheng P, Zou W, Curiel TJ. Regulatory T-cells in ovarian cancer: biology and therapeutic potential. Am J Reprod Immunol 2005; 54:369–77. [DOI] [PubMed] [Google Scholar]
  • 126.Rasku MA, Clem AL, Telang S, Taft B, Gettings K, Gragg H, et al. Transient T-cell depletion causes regression of melanoma metastases. J Transl Med 2008; 6:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Dannull J, Su Z, Rizzieri D, Yang BK, Coleman D, Yancey D, et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T-cells. J Clin Invest 2005; 115:3623–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Denileukin Diftitox in Treating Patients With Advanced Breast Cancer That Did Not Respond to Previous Treatment. Available from: http://www.clinicaltrials.gov/ct2/results?term=NCT00425672.
  • 129.A Pilot Study Evaluating the Efficacy of Regulatory T-Cell Suppression by Ontak in Metastatic Pancreatic Cancer. Available from: http://www.clinicaltrials.gov/ct2/results?term=NCT00726037.
  • 130.Ovarian Dendritic Cell Vaccine Trial. Available from: http://www.clinicaltrials.gov/ct2/results?term=NCT00703105.
  • 131.Tsurushita N, Hinton PR, Kumar S. Design of humanized antibodies: from anti-Tac to Zenapax. Methods 2005; 36:69–83. [DOI] [PubMed] [Google Scholar]
  • 132.Waldmann TA. Anti-Tac (daclizumab, Zenapax) in the treatment of leukemia, autoimmune diseases, and in the prevention of allograft rejection: a 25-year personal odyssey. J Clin Immunol 2007; 27:1–18. [DOI] [PubMed] [Google Scholar]
  • 133.Chen J, Zhang M, Ju W, Waldmann TA. Effective treatment of a murine model of adult T-cell leukemia using depsipeptide and its combination with unmodified daclizumab directed toward CD25. Blood 2009; 113:1287–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Brode S, Cooke A. Immune-potentiating effects of the chemotherapeutic drug cyclophosphamide. Crit Rev Immunol 2008; 28:109–26. [DOI] [PubMed] [Google Scholar]
  • 135.Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T-cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother 2007; 56:641–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Audia S, Nicolas A, Cathelin D, Larmonier N, Ferrand C, Foucher P, et al. Increase of CD4+ CD25+ regulatory T-cells in the peripheral blood of patients with metastatic carcinoma: a Phase I clinical trial using cyclophosphamide and immunotherapy to eliminate CD4+ CD25+ T lymphocytes. Clin Exp Immunol 2007; 150:523–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Gee K, Guzzo C, Che Mat NF, Ma W, Kumar A. The IL-12 family of cytokines in infection, inflammation and autoimmune disorders. Inflamm Allergy Drug Targets 2009; 8:40–52. [DOI] [PubMed] [Google Scholar]
  • 138.Skak K, Kragh M, Hausman D, Smyth MJ, Sivakumar PV. Interleukin 21: combination strategies for cancer therapy. Nat Rev Drug Discov 2008; 7:231–40. [DOI] [PubMed] [Google Scholar]

RESOURCES