3,689
Views
20
CrossRef citations to date
0
Altmetric
Review

Trial Watch: Therapeutic vaccines in metastatic renal cell carcinoma

, , , , &
Article: e1001236 | Received 27 Nov 2014, Accepted 18 Dec 2014, Published online: 21 May 2015

Abstract

Despite the renaissance of cancer immunotherapy, no novel immunotherapy has been approved for the treatment of renal cell cancer (RCC) since the availability of recombinant cytokines (interleukin-2, interferon-α). All vaccine trials have failed to meet their endpoints although they have highlighted potential predictive biomarkers (e.g., pre-existing immune response, hematological parameters, tumor burden). Recent advances in immunomodulatory therapies have prompted the study of combination treatments targeting the tumor immunosuppressive microenvironment consisting of regulatory T-cells (Treg), myeloid suppressor cells, and cytokines. Approaches under investigation are use of inhibitors to curb the overexpression of immune checkpoint ligands by tumor cells (e.g., anti-CTLA-4, anti-PD-1/PD-L1) and exploiting the immunomodulatory effects of anti-angiogenic agents that are the current standard of metastatic RCC care. Phase III trials are focusing on the possible synergy between therapeutic vaccines (e.g., IMA-901 and AGS-003) and anti-angiogenic agents.

Introduction

Kidney cancer accounts for 2.5% of all cancers, with an annual global incidence of 273,000 cases leading to 111,000 deaths. The major histologic subtype (80% of cases) is clear cell renal cell carcinoma (ccRCC) followed by papillary subtype 1 and 2, Bellini tumor and chromophobe carcinoma. ccRCC is a chemoresistant tumor probably because the cells derive from the luminal cells of proximal tubules that express a high intrinsic level of multidrug resistance (MDR-1) protein. About 30% of ccRCCs are diagnosed at the metastatic stage (mRCC).Citation1

Standard of care has improved significantly over the past decade. Localized ccRCC is treated by nephrectomy but the relapse rate is 30%. The benefit of nephrectomy in a metastatic setting was previously proven Citation2 but is again under investigation at the era of TKIs (CARMENA trial, NCT00930033). No adjuvant immunotherapy has yet been approved. Several anti-angiogenic agents such as inhibitors of the vascular endothelial growth factor (VEGF) pathway (sunitinib, sorafenib, axitinib, pazopanib, and bevacizumab in association with interferon) or inhibitors of the mammalian target of rapamycin (temsirolimus, everolimus) have been approved for the treatment of relapse or synchronous metastases.Citation3–10 These agents improve overall clinical outcome but a plateau has been reached in terms of efficacy and median overall survival (OS) in patients with advanced disease.

Before the advent of anti-angiogenic agents, cytokines (interferon-α (IFNα) and interleukin-2 (IL-2)) were used to stimulate both innate and adaptive immunity. Cytokines induce natural killer (NK) cell proliferation and differentiation, and promote survival, proliferation and differentiation of T-cells into effector T-helper (Th) cells,Citation11 with an added contentious effect of IL-2 on Treg.Citation12 In a meta-analysis, overall response rate (ORR) to cytokines was 12.9% (vs. 4.5% in the control arm) and complete response rate was 3.6%.Citation13 Long-term clinical responses have been reported.Citation14–15 Cytokines remain a therapeutic option for mRCC patients with good prognostic factors.

Immunogenicity of ccRCC

The immune system has a dual rule in cancer development. On the one hand, it can identify and control nascent tumor cells, thereby exerting immunosurveillance.Citation16 On the other, it can promote tumor progression through chronic inflammation, immunoselection of poorly immunogenic variants, and suppression of antitumor immunity.Citation17 The balance between activation and inhibition is maintained by immuno-editing. Evidence for immunosurveillance, which relies on T-cell response to tumor-associated antigens (TAA),Citation16 comes from rare cases of spontaneous complete responses observed in the placebo arm of a phase III trial.Citation18 The blood and tumor microenvironment of these cases harbored TAA-specific T-cells; the tumors were highly infiltrated by TAA-specific effector memory CD8 T-cells and CD4 T-cells.

Four classes of TAAs have been identified in RCC

Reactivated embryonic antigens (aka cancer testis antigens), such as MAGE, RAGE, PRAME, SART1, and NY-ESO1. Their expression in RCC is relatively low compared to that in melanoma or squamous cell lung carcinoma. The expression pattern in subtype ccBA of RCC (PRAME, SPANXC, C21orf99, SSX1) confers a worse prognosis than that (FATE1) of subtype ccAB.Citation19

Mutated antigens arising from mutated Von Hippel Landau (VHL) gene (present in >60% of sporadic RCCs) Citation20 or mutated p53 gene.

Tissue-specific antigens (e.g., kidney injury molecule (Kim-1), PAX 2) that are expressed in both normal and malignant renal tissue.

Other antigens overexpressed in many tumors (e.g. carbonic anhydrase IX (CAIX), MUC1 (CA15- 3), HER2, oncofetal antigen 5T4​​, peripilin 2, cyclin D1, c-Met and MMP-7).Citation21

A genomic approach to TAA identification should hasten steps in the development of cancer immunotherapies, i.e., in the evaluation of the advancement of immune-editing, selection of highly immunogenic tumors for personalized therapy, and identification of TAAs that drive addictive oncogenic pathways needed for cancer cell survival.Citation16

TAAs as targets for vaccines

Four criteria have to be met before use of a TAA as a potential target for a vaccine.

Lack of pre-existing immunotolerance. For a vaccine to be effective requires a functional immune system. Vaccines directed against TAAs that might be self-antigens (e.g., HER2) would constitute a means of overcoming immunologic tolerance to self-proteins. However, as cancers are genetically unstable, it might be easier to target TAAs that are neo-antigens through mutation and thus not recognized as “self” by the immune system.

Differential TAA expression between tumor and normal tissue. Immunotherapy toxicity and the spontaneous auto-immune paraneoplastic syndrome are thought to arise from TAA expression in normal tissue. For instance, the neurological toxicity of anti-MAGE-A3 TCR gene therapy (also recognizing MAGE-A9 and A12 epitopes) might be related to unreported MAGE-A12 expression in brain.Citation22 The uncertainty over antigen expression patterns in normal tissue implies a need for systematic monitoring of potential toxicity.

Immunogenicity. TAA induction of specific memory CD8+ T-cell expansion can be assessed by detecting TAA-specific T-cells using ELISPOT or FluoroSpot.Citation23

TAA role in tumorigenesis and tumor cell survival. Because tumors display genomic heterogeneity and constitutive instability, the TAA should preferably be part of an oncogenic addictive pathway in order to avoid selecting a poorly immunogenic variant for the vaccine-induced immune response. In a phase I study of a vaccine based on a VHL-mutated antigen, T-cells from 4 out of 5 evaluable patients were reactive against mutated peptides using IFNɣ-ELISPOTs.Citation24 Unfortunately, the phase II study (NCT00001703) was inconclusive because of poor accrual.

Immune escape mechanisms in ccRCC

The immune cellular response to RCCs is not efficient.

Analysis of CD8+ T-cell clonality has revealed a lower expansion rate in ccRCC than in other solid tumors.Citation25 Only 20% of CD8+ tumor infiltrating lymphocytes (TIL) recognize autologous tumor cells,Citation26 suggesting default in tumor recognition or resistance to TIL cytotoxicity in RCCs. Unlike in other solid tumors, CD8+ TIL in RCC are associated with a poor clinical prognosis.Citation27–28 This highlights the probable importance of immuno-editing in RCC oncogenesis.

Modified expression of several proteins in TILs may contribute to tumor immune escape: (i) decreased expression of CD3 zeta chain (which plays a key role in T-cell receptor signal transduction) is associated with a poor prognosis;Citation29 (ii) upregulated expression of the inhibitory programmed cell death protein PD-1 in as yet unidentified cells is also associated with a poor prognosis;Citation30 (iii) 20–40% of TILs express killer immunoglobulin-like inhibitory receptors (KIR2DL NK-receptors (CD58 a /b) of human leukocyte antigen C (HLA-C).Citation31 KIR can interact with major histocompatibility complex (MHC) Class 1 inhibitors before delivering a signal to the cell.

Like other tumors, therefore, RCCs can develop exhaust mechanisms (loss of HLA molecule expression, immunosuppressive signal production). Moreover, some molecules (CD70, HLA-G, PD-L1, B7-H3, B7-H4) are expressed at higher frequencies in RCC, thereby further affecting the phenotype of CD8+ TIL in RCC compared to other tumors.Citation32-34

State of the art of cancer immunotherapy in RCC

Currently, recombinant cytokines are the only approved immunotherapies for RCC. The potential of other types of cancer immunotherapy is, however, evidenced by the effectiveness of the cancer vaccine sipuleucel-T in prostate cancer.Citation35 Nevertheless, so far, no vaccine has shown proven efficacy in RCC although promising trials are ongoing (see below).

Tumor progression is partly due to escape from immunosurveillance, and targeting immunosuppressive mechanisms is a new thrust in cancer therapy.Citation36-37 Immune checkpoint inhibitors, in particular anti-PD-1/PD-L1 antibodies, have been shown to provide impressive tumor responses in metastatic cancers of different histologies (e.g., ipilimumab in metastatic melanoma).Citation38 PD-L1 is a cell surface glycoprotein belonging to the B7 family of T-cell costimulatory molecule and is overexpressed in many human tumors.Citation39 Activation of the PD-1/PD-L1 pathway induces T-cell anergy. PD-L1 downregulates immune responses by inhibiting both activated and memory T-cells. In a study of 306 patients with localized RCC (median follow-up, 11 y), PD-L1 expression correlated with increased risk of disease progression, cancer-specific death, and overall mortality.Citation40 Five-year cancer-specific survival rates were higher in patients with PD-L1 negative than positive RCCs (83% vs. 42%).

Early trials of anti-PD1 and anti-PD-L1 antibodies in mRCC patients have reported a 30% ORR and 20–25% prolonged response rate.Citation41–42 The anti-PD1 antibody pidilizumab (CT011) has a particularly good safety profile and lasting clinical activity.Citation63 Two ongoing phase III trials in mRCC patients are comparing first-line sunitinib to the anti-PD1 antibody nivolumab either alone (NCT01668784) or combined with ipilimumab. ORRs in early phase trials are low and highlight the need for a standard method of determining PD-L1 expression to improve prediction.Citation44

Another promising new approach is adoptive cell therapy using engineered T-cell Chimeric Antigen Receptor (CAR).Citation45

Clinical trial results for therapeutic RCC vaccines

We reviewed 6 clinical trials of RCC-specific therapeutic vaccines administered in an adjuvant or a metastatic setting: three completed phase III trials (Reniale,Citation46–47 Trovax,Citation48–50 and Vitespen Citation51) and three phase II trials (TG4010,Citation52 AGS003,Citation53–54 and IMA901 Citation21), as well as a meta-analysis of dendritic cell (DC)-based vaccines in mRCC Citation55 (). The publications were retrieved from a Medline search (from january 2004 to november 2014).

Table 1. Completed clinical trials of therapeutic vaccines in renal cell carcinoma (localized: RCC or metastatic: mRCC)

Reniale® is an autologous RCC-tumor lysate cell-based vaccine. It was administered post nephrectomy in an adjuvant setting in a phase III trial that had tumor progression risk (defined as progression or death) as primary endpoint. The goal of a significant reduction in risk was not met. However, the subgroup of patients with pT3 tumors showed significantly improved progression-free survival (PFS) compared to placebo (5-year PFS: 71.2% vs 65.4%, p = 0.02; 10-year PFS: 53.6% vs 36.2%, p = 0.022) as did patients with Fuhrman grade 3 tumors (5-year PFS: 71.9% vs 60.3%, p = 0.008). There was no significant improvement in patients with pT2 or grade 2 tumors. Strong criticism relating to methodological bias (choice of primary endpoint, between-arm imbalance) meant that drug approval was not granted despite the promising results.Citation46-47

TroVax® (MVA-5T4) is a therapeutic vaccine targeting a heavily glycosylated 5T4 antigen chiefly expressed in human placental trophoblasts but also expressed in various human cancer cells. This TAA is overexpressed in most RCCs.Citation56 The TRIST phase III trial of MVA-5T4 in combination with IFNα, IL2 or sunitinib as first-line mRCC therapy did not meet its objective of a significant increase in OS.Citation48 However, patients with a vaccine-induced antigen-specific immune response did show higher OS, and those with a good MSKCC prognostic score receiving MVA-5T4/IL2 had significantly better OS than patients on placebo/IL2 ; (Hazard Ratio (HR): 0·54 [0·30–0·98], p = 0·046). As MVA-5T4 targets a single TAA, each patient has to be evaluated for tumor TAA expression and for any preexisting specific immune response.

Vitespen (formerly Oncophage®) consists of a heat-shock protein (HSP) -glycoprotein 96 peptide complex derived from autologous tumor. Because HSPs bind strongly to peptides, they provide a tumor antigenic fingerprint. Vitespen showed a specific CD8+T-cell immune response and NK-cell expansion in 50% of patients in a preliminary study.Citation57 However, in the phase III trial, it did not meet its primary endpoint of a significant increase in relapse-free survival (37.7% vs. 39.8% in the observation arm (p = 0.5), median follow-up= 1.9 y).Citation51 In a planned subgroup analysis (stage I or II disease), a non-significant reduction in disease recurrence was observed (Hazard Ratio (HR): 0·576 [0·324–1·023], p = 0·056). Vitespen has been approved in Russia for intermediate-risk patients in an adjuvant setting.

TG4010 is a modified vaccinia virus expressing MUC1. MUC1 is overexpressed in RCC and is associated with a poor prognosis.Citation58 No objective clinical response was observed in the phase II study evaluating TG4010 efficacy and tolerability, alone or in combination with cytokines, as first-line mRCC therapy. Stable disease for >6 months was reported in 5/27 of evaluable patients (18%) with TG4010 alone and 6/20 of patients (30%) with TG4010 plus cytokines. MUC1-specific CD8+ T-cell responses were associated with longer OS.Citation50

AGS-003 is a DC-based vaccine in which mature DCs are electroporated with amplified total tumor mRNA and CD40-ligand (CD40L). CD40L expression on the DC surface is thought to induce a costimulation signal, counteract T-cell anergy induced by inhibitory receptors like PD-1, and reestablish a balance in favor of T-cell activation. AGS-003 was tested in combination with sunitinib in a phase II study in poor or intermediate risk mRCC patients eligible for nephrectomy. Median PFS was 11.2 month; median OS was 30.2 months. No additive toxicity was reported other than grade 1 local reactions.Citation53 Peripheral blood mononuclear cells (PBMCs) showed decreased Treg levels and increased levels of CD28+ memory cytotoxic T-cells (CTLs) that were positively correlated with improved PFS.Citation59 A phase III trial is ongoing.

IMA-901® is a 10 tumor-associated peptide (TUMAP) pool combined with granulocyte macrophage colony-stimulating factor (GM-CSF) injected after a single low dose of cyclophosphamide. The phase I and II trials had two objectives.Citation21 The first was to identify strong immunogenic TUMAPs (using the XPRESIDENT platform to screen for overexpressed genes corresponding to HLA ligands) and specific anti-TUMAPS T-cells in blood. Nine HLA-A*02-restricted and one HLA-DR-restricted TUMAPs were selected (see footnote to ). The studies showed that multi-anti-TUMAP T-cell responses were associated with better disease control and that lower pre-vaccine infiltration of Tregs was correlated with a better T-cell response. On the other hand, preexisting type 2 (CD15+IL-4Ra+) and type 4 (CD14+HLA-DR-/lo) myeloid derived suppressor cells (MDSC) seemed to worsen prognosis. The second objective was to identify an agent (single low cyclophosphamide dose) capable of depleting Tregs. A vaccine vector was not used for reasons of cost, safety, and simplicity of clinical use. An efficient immune response was achieved, with disease control rates of 31% and 14% after cytokines or anti-angiogenic agents TKIs, respectively. A Phase III trial is ongoing.

DC-vaccines

DCs are potent antigen-presenting cells. Trials of small mRCC patient numbers have evaluated DC-based vaccines to boost specific antitumor responses but with few significant results. A meta-analysis of 12 trials reported an ORR of 12.7% and a clinical benefit of 48%. The trials differed in DC subtype (9 used mature and 3 immature monocyte-derived DCs) and type of antigen (6 tumor lysate, 2 peptide pulsing, 1 either cell lysate or peptide, 2 mRNA coincubation, 1 cell fusion with autologous tumor cells). The cellular immune response and DC dose (mean= 38.7×10*6 cells) were the main predictors of response.Citation55

Ongoing clinical trials of therapeutic RCC vaccines

We selected five ongoing trials: two ongoing phase III trials, one Phase II trial and two Phase I trials (). The trials were retrieved from a NCT Database search in November 2014 on Clinicaltrial.gov.

Table 2. Ongoing clinical trials of therapeutic vaccines in renal cell carcinoma

IMA-901

In the IMPRINT phase III trial, 340 mRCC patients with HLA-A*02 haplotype were randomized to a combination of first-line sunitinib and IMA-901 (with single dose cyclophosphamide and GM-CSF as adjuvant) or sunitinib alone. Recruitment is closed.

AGS-003

The ADAPT phase III trial is comparing the combination AGS-003 plus sunitinib versus sunitinib alone, in newly diagnosed unfavorable-risk mRCC patients. Recruitment will stop once 450 patients have been included.

DC-RCC is a tumor vaccine in which patient-derived tumor cells are fused with autologous DCs. In a phase I trial of 16 mRCC patients, DC-RCC vaccination post-cytoreductive nephrectomy was associated with antitumor immunity in all 16 patients and good tolerance, with 7 patients achieving a partial response or prolonged stable disease.Citation60 The efficacy of DC vaccination might be enhanced by targeting effector cell inhibition induced by the tumor microenvironment. The anti-PD1 antibody pidilizumab (CT011), which promotes Th1 polarization of vaccine-induced T-cells and decreases Treg number, has shown promising results when combined ex vivo with an autologous DC/myeloma fusion vaccine.Citation61 The combination DC-RCC/CT011 is being studied in a phase II trial (NCT01441765) in patients who have undergone nephrectomy or cytoreductive metastasis surgery.

AdGMCAIX is another DC-based vaccine. A fusion gene construct of GM-CSF and CAIX is transduced by a replication deficient adenovirus into autologous DC. CAIX is a hypoxia-induced protein. Its expression in ccRCC is ubiquitous because of the mutational loss of VHL that is invariable in ccRCC (> 95%) but rare in normal cells.Citation62 It is used to activate a specific immune response whereas GM-CSF is used to boost antitumor immunity. AdGMCAIX has shown activity in preventive and therapeutic preclinical modelsCitation63 and is under study in a phase I trial of mRCC patients (NCT01826877). All six patients included had a good tolerance profile. Four patients were evaluable for response, one had progressive disease, two had stable disease lasting < 6 months, and one still had stable disease at 6 months. Issues pertaining to the conduct of the study have temporarily stopped patient accrual.Citation64

DC-CIK

Autologous DCs pulsed with tumor lysate in order to generate a specific antitumor response have been combined with an infusion of Cytokine Induced Killer (CIK) cells (a heterogeneous population of non-MHC restricted CD3+ CD56+ T-cells) on the premise that the vaccine might enhance CIK cell activity and the CIK cells might enhance the vaccine-induced antitumor immune response.Citation65 In a phase I trial (NCT00862303) of 28 patients with mRCC, the ORR was 39% and the disease control rate was 75%. No serious adverse event was reported.Citation66

Strategies for the development of therapeutic RCC vaccines

No vaccine-based treatment has been approved in RCC therapy because of lack of proven efficacy, but promising trials are ongoing.

Optimal choice of TAA

With the exception of cancer cell-derived vaccines (based on tumor lysate or fusion cells) which embrace all antigens expressed by tumor cells, most therapeutic vaccines target only one or a few TAAs. Choosing a single TAA provides a stronger directional immune response but downsides are variable TAA expression on tumor cells and tumor heterogeneity among patients. For instance, the IMA-901 vaccine is exclusively destined for HLA-A*02 patients, i.e., about 50% of Caucasians. It is thus essential to test each tumor for the TAA targeted by the potential vaccine. Choosing a TAA involved in an addictive oncogenesis pathway can help prevent secondary resistance to treatment by down-regulation of antigen expression.

Choice of TAA vector

A potent anti-TAA immune response is elicited by viral delivery systems, such as the modified vaccinia virus Ankara (MVA) or poxvirus, as they abate self-tolerance. However, to avoid an anti-vector immune response that might curb the efficiency of repeat administrations of the same recombinant vaccine, either a strategy using different viral vectors for the prime and boost is usedCitation67 or the peptide of interest is loaded ex vivo directly on DCs (e.g., sipuleucel-T). The latter method has a theoretical benefit even if the exact nature of the cells used as DC precursors (mononuclear?) may not be known. An option to be considered in DC-based therapies is use of non-replicative vectors to avoid the intrinsic immunogenicity of live vectors and to reduce treatment complexity and cost. Non-replicative vectors have been shown to be effective in preclinical models.Citation68 In practice, TAA-specific immune responses and clinical efficacy should be compared using different vectors but it may also be feasible to do away altogether with vectors for peptide vaccines (see IMA-901). This would lower cost and might even be safer.

Choice of adjuvant to boost immune response

Choice of TAA and vector is followed by the difficult choice of best adjuvant to boost and drive the immune response. The ideal adjuvant should trigger an effective immune response and ensure strong immunogenicity. A variety of agents can be defined as adjuvants (e.g., water-in-oil emulsions, GM-CSF, toll-like receptor (TLR) agonists).

Several water-in-oil emulsions were tested after Freund incomplete adjuvant (FIA) was abandoned. Like FIA, Montanide ISA 51 - an experimental adjuvant for protein and multi-peptide vaccines such as the dual-adjuvant telomerase vaccine GX301 is composed of a light mineral oil and a mannide monooleate emulsifier but the emulsifier is highly refined and the emulsifier-to-oil ratio is lower than in FIA. The resultant emulsion is more consistent and controllable, and is effective and in general well tolerated in humans. The main reported adverse reactions are transient local reactions, including local swelling and pain with or without fever.Citation69

GM-CSF is used as a local adjuvant for IMA-901, and is expressed within a fusion protein in the sipuleucel-T and AdGMCAIX-transduced autologous DC-based vaccines. GM-CSF plays a critical role in DC maturation and T-cell proliferation and activation, and increases DC-mediated responses to tumor cells.Citation70 When used as an adjuvant, it might, however, increase MDSCs in the tumor micro-environment and in blood. This deleterious effect remains, however, controversial and its clinical relevance is doubtful.Citation71

In the absence of a "danger signal", tolerization may be induced instead of a proper antitumor immune response. Danger-associated molecular patterns (DAMPs) can reverse DC inhibition in the tumor microenvironment, increase the efficiency of antigen presentation to cytotoxic T-cells (CTLs), and prevent tolerization to tumor antigens. DAMPs interact with TLRs, and TLR agonists are in increasing use in cancer therapy.Citation72 The one in most common use is poly-ICLC, a TLR3 agonist, which has shown clinical benefit, either alone or in combination, in a variety of cancers.Citation73 Imiquimod, a TLR7 agonist, has been approved for the treatment of superficial basal skin carcinoma and pre-cancerous lesions. Tasquinimod, a TLR4 agonist with anti-angiogenic activity, targets S100A9 protein and affects regulatory myeloid cell function.Citation74 A phase II trial of tasquinimod in several cancers including mRCC is ongoing (NCT01743469).

Identification of biomarkers

Several potential biomarkers have emerged during trials of vaccines against RCC.

Hematologic impairment (low hemoglobin level, high neutrophil count and low lymphocyte count) predicted a poor response to TroVax.Citation49–50 The neutrophil-lymphocyte ratio (NLR) has also shown prognostic value in mRCC (HR = 1.59 [1.10-2.31], p = 0.014 for NLR >3.3).Citation75 An Immune Response Surrogate (IRS) predictive score for vaccine efficiency (anti-5T4 antibody, hemoglobin, and hematocrit) has been proposed but needs validation.Citation49

Among potential serum biomarkers, serum apolipoprotein A-1 (ApoA1) and chemokine (C-C motif) ligand 17 (CCL17) predicted immune response to TAA and OS in the IMA-901 trial. Both factors were significant positive pre­dictors of immune response (p = 0.016, p = 0.032, respectively) and multipeptide responses (p < 0.0001, p = 0.0028, respectively). High levels of these biomarkers identified patient populations with significantly longer OS (p < 0.007, p < .011, respectively) but only in the cyclophosphamide arm of the trial.

Pretherapy immune response (i.e., amount of TAA-specific CTL detected by ELISPOT) seems to be a good predictor of response to immunotherapy.

Immunosuppressive cells such as Tregs (CD45+CD3+CD4+FoxP3+CD25hiCD127low), MDSC type 4 (CD14+HLA-DR-/lo) or type 5 (CD11b+CD14-CD15+) cells and Th17 cells (IL-17+CD4+Tcells) are elevated in several cancers and may predict metastatic progression.Citation76 TILs should be more informative than PBMCs as they reflect tumor immune response better but caution is needed as technicalities (e.g., isolation methods) might generate artifacts.

In short, there is an urgent need for effective immune-monitoring both pretreatment in order to select patients and specifically counteract the immunosuppressive pathways involved and also during treatment for early prediction of clinical response to therapeutic vaccines.

Counteracting the immunosuppressive environment

After boosting the immune response with adjuvants, it is necessary to loosen the brake on the immunosuppressive environment from Tregs, MDSCs, overexpression of immune checkpoint proteins and other mechanisms.

Several approaches to deplete or block Treg function have been successfully tested in preclinical models and in humans.Citation77–80

For instance, a low cyclophosphamide dose decreased Tregs in a preclinical modelCitation77–78 and in the phase I-II trials of IMA-901 in mRCC.

Anti-angiogenic agents are the current standard of care for mRCC. They inhibit cancer-related immunosuppression thus justifying combination with immunotherapy. Some induce a less immunosuppressive tumor microenvironment by decreasing Tregs and MDSCs.Citation81 However, it is unclear whether they affect VEGF-dependent DC maturation or Treg proliferation.Citation82 In a prospective study of 28 mRCC patients receiving first-line sunitinib, the number of Tregs (defined as CD3+CD4+CD25(hi) Foxp3+) in blood and tumor fell after each sunitinib cycle. OS was significantly longer in patients with a decrease in Treg after two or three cycles of sunitinib (p < 0.05).Citation81 Treg depletion has also been observed with axitinib.Citation83 In murine models, combining anti-angiogenic agents with immunotherapy enhanced the effectiveness of immunostimulation. Treg number may thus be a predictor of anti-angiogenic response.

There is an inverse association between angiogenesis and PD-L1 expression in primary RCC.Citation84 The mechanism of escape from immunosurveillance might depend on tumor biology. Increased VEGF expression decreases immune infiltration and might thus lessen the adaptative pressure on the tumor which would thus not need to express PD-L1. PD-L1 expression, which often reflects a preexisting immune response, may be an adaptation mechanism to immune selection pressure but, depending upon the tumor, might also be due to early oncogenic events.Citation85

Vaccination results in upregulation of immune checkpoint receptors (e.g., PD1 receptor and T-cell membrane protein 3 (TIM3) that suppresses Th1 cell activation) and induction of T-cell anergy.Citation86 Studies are being conducted on immune checkpoint inhibitors combined with anti-angiogenic agents or DC-based vaccines in mRCC. Synergy between an anti-PD-L1 antibody and a vaccine against human papillomavirus was shown.Citation87

Assessing clinical response to immunotherapy

Most therapeutic vaccines are considered failures because direct clinical tumor regression, as evaluated by radiological response according to RECIST criteria (Response Evaluation Criteria in Solid Tumors), is rare. A survival benefit can be observed in the absence of an objective response, as demonstrated by the efficacy of sipuleucel-T in prostate cancer.Citation88 Moreover, the immune checkpoint inhibitor, ipilimumab, showed distinct radiological response patterns in metastatic melanoma (long stabilization, pre-response flare-up, and delayed response). An early response might not always be a good surrogate for survival. New radiological criteria – the immune response related criteria (irRC) – have been proposed for solid tumor response to immunotherapy but have not undergone validationCitation89 A simple threshold percentage reduction in tumor burden was successfully used as a criterion in a trial of nivolumab plus ipilimumab in malignant melanoma.Citation90 Until the clinical relevance of new end-points is established and a robust surrogate indicator is developed, OS must remain the endpoint in immunotherapy trials.

Concluding Remarks

Reactivating the immune response after an escape from immunosurveillance is challenging. A shrewd choice of TAA and vector, as well as the evaluation of pretreatment specific immune response, are necessary to target an immunogenic tumor but are not sufficient to halt escape mechanisms. In short, boosting the immune response with vaccines without counteracting immunosuppressive mechanisms is not enough. Avenues that are being explored are cyclophosphamide-induced Treg depletion and immunomodulation by immune checkpoint inhibitors and anti-angiogenic agents. Therapeutic vaccines combined with immunomodulators that will boost antigen presentation and DC maturation, offset the immunosuppressive micro-environment and orient Th1 polarization of the immune response are being investigated in attempts to achieve an efficient antitumor immune response with significant clinical benefit.

However, an efficient immune response might be hard to achieve in the absence of immune infiltration and in a context of an extrinsic resistance mechanism. The absence of immune infiltration and PD-L1 overexpression, not because of immune pressure but as a result of loss of a tumor suppressor gene (phosphatase and tensin homolog (PTEN)) in RCC, are documented adverse prognostic factors. The impact of the immune micro-environment on immunotherapy outcome should thus be assessed and used to stratify patients in clinical trials. Such an approach could lead to the development of more relevant biomarkers and to the translation of the latest advances in immunotherapy to kidney cancer.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Funding

This work has received funding from Canceropôle Ile de France, Institut National du Cancer (INCa) , Ligue Nationale contre le Cancer, Association de Recherche contre le Cancer (ARC), Sites de Recherche Intégrée sur le Cancer (SIRIC - project: Cancer Research for Personalized Medicine (CARPEM)), Agence Nationale de la Recherche (ANR - program: RPIB), LabEx Immuno-Oncology.

References

  • Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. N Engl J Med 1996; 335:865-75; PMID:8778606; http://dx.doi.org/10.1056/NEJM199609193351207.
  • Flanigan RC, Mickisch G, Sylvester R, Tangen C, Van Poppel H, Crawford ED. Cytoreductive nephrectomy in patients with metastatic renal cancer: a combined analysis. J Urol 2004; 171:1071-76; PMID:14767273; http://dx.doi.org/10.1097/01.ju.0000110610.61545.ae.
  • Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007; 356:125-34; PMID:17215530; http://dx.doi.org/10.1056/NEJMoa060655.
  • Motzer RJ, Michaelson MD, Redman BG, Hudes GR, Wilding G, Figlin RA, Ginsberg MS, Kim ST, Baum CM, DePrimo SE et al. Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma. J Clin Oncol 2006; 24:16-24; PMID:16330672; http://dx.doi.org/10.1200/JCO.2005.02.2574.
  • Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 2010; 28:1061-68; PMID:20100962; http://dx.doi.org/10.1200/JCO.2009.23.9764.
  • Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, Michaelson MD, Gorbunova VA, Gore ME, Rusakov IG et al. Comparative effectiveness of axitinib vs. sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 2011 Dec 3; 378(9807):1931-9; http://dx.doi.org/10.1016/S0140-6736(11)61613-9.
  • Escudier B, Bellmunt J, Négrier S, Bajetta E, Melichar B, Bracarda S, Ravaud A, Golding S, Jethwa S, Sneller V. Phase III trial of bevacizumab plus interferon alfa-2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. J Clin Oncol 2010; 28(13):2144-50; PMID:20368553; http://dx.doi.org/10.1200/JCO.2009.26.7849.
  • Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007; 356(22):2271-81; PMID:17538086; http://dx.doi.org/10.1056/NEJMoa066838.
  • Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008; 372(9637):449-56; PMID:18653228; http://dx.doi.org/10.1016/S0140-6736(08)61039-9.
  • Albiges L, Oudard S, Negrier S, Caty A, Gravis G, Joly F, Duclos B, Geoffrois L, Rolland F, Guillot A et al. Complete remission with tyrosine kinase inhibitors in renal cell carcinoma. J Clin Oncol 2012; 30(5):482-7; PMID:22231040; http://dx.doi.org/10.1200/JCO.2011.37.2516.
  • Vacchelli E, Aranda F, Obrist F, Eggermont A, Galon J, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunostimulatory cytokines in cancer therapy. Oncoimmunology 2014; 3:e29030; PMID:25083328; http://dx.doi.org/10.4161/onci.29030.
  • Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood 2006; 107:2409-14; PMID:16304057; http://dx.doi.org/10.1182/blood-2005-06-2399.
  • Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database Syst Rev 2005; CD001425; PMID:15674877; http://dx.doi.org/10.1002/14651858.
  • Rosenberg SA. IL-2: the first effective immunotherapy for human cancer. J Immunol 2014; 192:5451-58; PMID:24907378; http://dx.doi.org/10.4049/jimmunol.1490019.
  • Tartour E, Mathiot C, Fridman WH. Current status of interleukin-2 therapy in cancer. Biomed Pharmacother 1992; 46, 473-84; PMID:1306361; http://dx.doi.org/10.1016/0753-3322(92)90005-R.
  • Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases-elimination, equilibrium and escape. Curr Opin Immunol 2014; 27:16-25; PMID:24531241; http://dx.doi.org/10.1016/j.coi.2014.01.004.
  • Vesely MD, Schreiber RD. Cancer immunoediting: antigens, mechanisms, and implications to cancer immunotherapy. Ann N Y Acad Sci 2013; 1284:1-5; PMID:23651186; http://dx.doi.org/10.1111/nyas.12105.
  • Gleave ME, Elhilali M, Fradet Y, Davis I, Venner P, Saad F, Klotz LH, Moore MJ, Paton V, Bajamonde A. Interferon gamma-1b compared with placebo in metastatic renal-cell carcinoma. Canadian Urologic Oncology Group. N Engl J Med 1998; 338:1265-71; PMID:9562580; http://dx.doi.org/10.1056/NEJM199804303381804.
  • Yao J, Caballero OL, Yung WKA, Weinstein JN, Riggins GJ, Strausberg RL, Zhao Q. Tumor subtype-specific cancer-testis antigens as potential biomarkers and immunotherapeutic targets for cancers. Cancer Immunol Res 2014; 2:371-79; PMID:24764584; http://dx.doi.org/10.1158/2326-6066.CIR-13-0088.
  • Linehan WM, Pinto PA, Srinivasan R, Merino M, Choyke P, Choyke L, Coleman J, Toro J, Glenn G, Vocke C et al. Identification of the genes for kidney cancer: opportunity for disease-specific targeted therapeutics. Clin. Cancer Res 2007; 13:671s-79s; PMID:17255292; http://dx.doi.org/10.1158/1078-0432.CCR-06-1870.
  • Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, Staehler M, Brugger W, Dietrich PY, Mendrzyk R et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med 2012; 18(8):1254-61; PMID:22842478; http://dx.doi.org/10.1038/nm.2883.
  • Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, Dudley ME, Feldman SA, Yang JC, Sherry RM et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 2013; 36:133-51; PMID:23377668; http://dx.doi.org/10.1097/CJI.0b013e3182829903.
  • Chauvat A, Benhamouda N, Gey A, Lemoine FM, Paulie S, Carrat F, Gougeon M-L, Rozenberg F, Krivine A, Cherai M et al. Clinical validation of IFNγ/IL-10 and IFNγ/IL-2 FluoroSpot assays for the detection of Tr1 T cells and influenza vaccine monitoring in humans. Hum Vaccin Immunother 2014; 10:104-13; PMID:24084262; http://dx.doi.org/10.4161/hv.26593.
  • Rahma OE, Ashtar E, Ibrahim R, Toubaji A, Gause B, Herrin VE, Linehan WM, Steinberg SM, Grollman F, Grimes G et al. A pilot clinical trial testing mutant von Hippel-Lindau peptide as a novel immune therapy in metastatic renal cell carcinoma. J Transl Med 2010; 8, 8; http://dx.doi.org/10.1186/1479-5876-8-8.
  • Sittig SP, Køllgaard T, Grønbæk K, Idorn M, Hennenlotter J, Stenzl A, Gouttefangeas C, Thor Straten P. Clonal expansion of renal cell carcinoma-infiltrating T lymphocytes. Oncoimmunology 2013; 2:e26014; PMID:24228230; http://dx.doi.org/10.4161/onci.26014.
  • Markel G, Cohen-Sinai T, Besser MJ, Oved K, Itzhaki O, Seidman R, Fridman E, Treves AJ, Keisari Y, Dotan Z et al. Preclinical evaluation of adoptive cell therapy for patients with metastatic renal cell carcinoma. Anticancer Res 2009; 29:145-54; PMID:19331144.
  • Remark R, Alifano M, Cremer I, Lupo A, Dieu-Nosjean M-C, Riquet M, Crozet L, Ouakrim H, Goc J, Cazes A et al. Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: influence of tumor origin. Clin Cancer Res 2013; 19:4079-91; PMID:23785047; http://dx.doi.org/10.1158/1078-0432.CCR-12-3847.
  • Nakano O, Sato M, Naito Y, Suzuki K, Orikasa S, Aizawa M, Suzuki Y, Shintaku I, Nagura H, Ohtani H. Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res 2001; 61:5132-36; PMID:11431351.
  • Tartour E, Latour S, Mathiot C, Thiounn N, Mosseri V, Joyeux I, D'Enghien CD, Lee R, Debre B, Fridman WH. Variable expression of CD3-zeta chain in tumor-infiltrating lymphocytes (TIL) derived from renal-cell carcinoma: relationship with TIL phenotype and function. Int J Cancer 1995 63:205-12; PMID:7591205; http://dx.doi.org/10.1002/ijc.2910630210.
  • Thompson RH, Dong H, Lohse CM, Leibovich BC, Blute ML, Cheville JC, Kwon ED. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin Cancer Res 2007b; 13:1757-61; http://dx.doi.org/10.1158/1078-0432.CCR-06-2599.
  • Gati A, Guerra N, Giron-Michel J, Azzarone B, Angevin E, Moretta A, Chouaib S, Caignard A. Tumor cells regulate the lytic activity of tumor-specific cytotoxic t lymphocytes by modulating the inhibitory natural killer receptor function. Cancer Res 2001; 61:3240-44; PMID:11309272.
  • Wang QJ, Hanada K-I, Robbins PF, Li YF, Yang JC. Distinctive features of the differentiated phenotype and infiltration of tumor-reactive lymphocytes in clear cell renal cell carcinoma. Cancer Res 2012; 72:6119-29; PMID:23071066; http://dx.doi.org/10.1158/0008-5472.CAN-12-0588.
  • Dunker K, Schlaf G, Bukur J, Altermann WW, Handke D, Seliger B. Expression and regulation of non-classical HLA-G in renal cell carcinoma. Tissue Antigens 2008; 72:137-48; PMID:18721274; http://dx.doi.org/10.1111/j.1399-0039.2008.01090.x.
  • Crispen PL, Sheinin Y, Roth TJ, Lohse CM, Kuntz SM, Frigola X, Thompson RH, Boorjian SA, Dong H, Leibovich BC et al. Tumor cell and tumor vasculature expression of B7-H3 predict survival in clear cell renal cell carcinoma. Clin Cancer Res 2008; 14:5150-57; PMID:18694993; http://dx.doi.org/10.1158/1078-0432.CCR-08-0536.
  • Kantoff PW, Higano CS, Shore ND et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010; 363(5):411-22; PMID:20818862; http://dx.doi.org/10.1056/NEJMoa1001294.
  • Tartour E, Sandoval F, Bonnefoy J-Y, Fridman WH. Cancer immunotherapy: recent breakthroughs and perspectives. Med Sci (Paris) 2011; 27:833-41; PMID:22027420; http://dx.doi.org/10.1051/medsci/20112710011.
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144(5):646-74; PMID:21376230; http://dx.doi.org/10.1016/j.cell.2011.02.013.
  • Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC et al. Improved survival with ipilimumab in patients with metastatic melanoma.N Engl J Med 2010; 363(8):711-23; PMID:20525992; http://dx.doi.org/10.1056/NEJMoa1003466.
  • Azuma T, Yao S, Zhu G, Flies AS, Flies SJ, Chen L. B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood 2008; 111:3635-43; PMID:18223165; http://dx.doi.org/10.1182/blood-2007-11-123141.
  • Thompson RH, Dong H, Kwon ED. Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy. Clin Cancer Res 2007a; 13:709s-15s; http://dx.doi.org/10.1158/1078-0432.CCR-06-1868.
  • Brahmer JR, Tykodi SS, Chow LQM, Hwu W-J, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366:2455-65; PMID:22658128; http://dx.doi.org/10.1056/NEJMoa1200694.
  • Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366:2443-54; PMID:22658127; http://dx.doi.org/10.1056/NEJMoa1200690.
  • Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010; 28:3167-75; PMID:20516446; http://dx.doi.org/10.1200/JCO.2009.26.7609.
  • Granier C, Roussel H, De Guillebon E, Ladoire S, Gey A, Combe P, Vano Y, Fabre E, Oudard S, Badoual C, Tartour E. Biomarkers from the tumor microenvironment to predict clinical response to checkpoint inhibitors.J Oncopathol In Press. 2014.
  • Aranda F, Vacchelli E, Obrist F, Eggermont A, Galon J, Hervé Fridman W, Cremer I, Tartour E, Zitvogel L, Kroemer G et al. Trial watch: adoptive cell transfer for anticancer immunotherapy. Oncoimmunology 2014; 3:e28344; PMID:25050207; http://dx.doi.org/10.4161/onci.28344.
  • Jocham D, Richter A, Hoffmann L, Iwig K, Fahlenkamp D, Zakrzewski G, Schmitt E, Dannenberg T, Lehmacher W, von Wietersheim J et al. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: phase III, randomised controlled trial. Lancet 2004; 363, 594-99; PMID:14987883; http://dx.doi.org/10.1016/S0140-6736(04)15590-6.
  • May M, Kendel F, Hoschke B, Gilfrich C, Kiessig S, Pflanz S, Seidel M, Brookman-Amissah S. Adjuvant autologous tumour cell vaccination in patients with renal cell carcinoma. Overall survival analysis with a follow-up period in excess of more than 10 years. Urologe A 2009; 48:1075-83; PMID:19562320; http://dx.doi.org/10.1007/s00120-009-2044-y.
  • Amato RJ, Hawkins RE, Kaufman HL, Thompson JA, Tomczak P, Szczylik C, McDonald M, Eastty S, Shingler WH, de Belin J et al. Vaccination of metastatic renal cancer patients with MVA-5T4: a randomized, double-blind, placebo-controlled phase III study. Clin Cancer Res 2010; 16:5539-47; PMID:20881001; http://dx.doi.org/10.1158/1078-0432.CCR-10-2082.
  • Said R, Amato RJ. Identification of pre- and post-treatment markers, clinical, and laboratory parameters associated with outcome in renal cancer patients treated with MVA-5T4. Front Oncol 2013; 3:185; PMID:23875174; http://dx.doi.org/10.3389/fonc.2013.00185.
  • Harrop R, Treasure P, de Belin J, Kelleher M, Bolton G, Naylor S, Shingler WH. Analysis of pre-treatment markers predictive of treatment benefit for the therapeutic cancer vaccine MVA-5T4 (TroVax). Cancer Immunol Immunother 2012; 61(12):2283-94. Epub 2012 Jun 13; http://dx.doi.org/10.1007/s00262-012-1302-9.
  • Wood C, Srivastava P, Bukowski R, Lacombe L, Gorelov AI, Gorelov S, Mulders P, Zielinski H, Hoos A, Teofilovici F et al. An adjuvant autologous therapeutic vaccine (HSPPC-96; vitespen) versus observation alone for patients at high risk of recurrence after nephrectomy for renal cell carcinoma: a multicentre, open-label, randomised phase III trial. Lancet 2008; 372:145-54; PMID:18602688; http://dx.doi.org/10.1016/S0140-6736(08)60697-2.
  • Oudard S, Rixe O, Beuselinck B, Linassier C, Banu E, Machiels J-P, Baudard M, Ringeisen F, Velu T, Lefrere-Belda M-A et al. A phase II study of the cancer vaccine TG4010 alone and in combination with cytokines in patients with metastatic renal clear-cell carcinoma: clinical and immunological findings. Cancer Immunol Immunother 2011; 60:261-71; PMID:21069322; http://dx.doi.org/10.1007/s00262-010-0935-9.
  • Amin A, Dudek A, Logan T et al. A Phase II study testing the safety and activity of AGS-003 as an immunotherapeutic in subjects with newly diagnosed advanced stage renal cell carcinoma (RCC) in combination with sunitinib. J Clin Oncol 2010; 28(15 Suppl.), Abstract: 4588.
  • Amin A, Dudek A, Logan T et al. Prolonged survival with personalized immunotherapy (AGS-003) in combination with sunitinib in unfavorable risk metastatic RCC (mRCC). J Clin Oncol 2013; 31(Suppl. Six), Abstract 357.
  • Draube A, Klein-González N, Mattheus S, Brillant C, Hellmich M, Engert A, von Bergwelt-Baildon M. Dendritic cell based tumor vaccination in prostate and renal cell cancer: a systematic review and meta-analysis. PLoS ONE 2011; 6:e18801; PMID:21533099; http://dx.doi.org/10.1371/journal.pone.0018801.
  • Griffiths RW, Gilham DE, Dangoor A, Ramani V, Clarke NW, Stern PL, Hawkins RE. Expression of the 5T4 oncofoetal antigen in renal cell carcinoma: a potential target for T-cell-based immunotherapy. Br J Cancer 2005; 93:670-7; PMID:16222313; http://dx.doi.org/10.1038/sj.bjc.6602776.
  • Tamura Y, Peng P, Liu K, Daou M, Srivastava PK. Immunotherapy of tumors with autologous tumor-derived heat shock protein preparations. Science 1997; 278:117-20; PMID:9311915; http://dx.doi.org/10.1126/science.278.5335.117.
  • Aubert S, Fauquette V, Hémon B, Lepoivre R, Briez N, Bernard D, Van Seuningen I., Leroy X, Perrais M. MUC1, a new hypoxia inducible factor target gene, is an actor in clear renal cell carcinoma tumor progression. Cancer Res. 2009; 69:5707-15; PMID:19549898; http://dx.doi.org/10.1158/0008-5472.CAN-08-4905.
  • Figlin RA, Nicolette CA, Amin A et al. Monitoring T-cell responses in a Phase II study of AGS-003, an autologous dendritic cell-based therapy in patients with newly diagnosed advanced stage renal cell carcinoma in combination with sunitinib. J Clin Oncol 2011; 29(Suppl.), Abstract 2532.
  • Avigan D, Rosenblatt J, Vasir B et al. Vaccination with DC/RCC fusions following cytoreductive nephrectomy. J Clin Oncol 2010; 28(15s): abstract 2511.
  • Rosenblatt J, Glotzbecker B, Mills H, Vasir B, Tzachanis D, Levine JD, Joyce RM, Wellenstein K, Keefe W, Schickler M et al. PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccine. J Immunother 2011; 34, 409-18; PMID:21577144; http://dx.doi.org/10.1097/CJI.0b013e31821ca6ce.
  • Grabmaier K, A de Weijert MC, Verhaegh GW, Schalken JA, Oosterwijk E. Strict regulation of CAIX(G250/MN) by HIF-1alpha in clear cell renal cell carcinoma. Oncogene 2004; 23:5624-31; PMID:15184875; http://dx.doi.org/10.1038/sj.onc.1207764.
  • Birkhäuser FD, Koya RC, Neufeld C, Rampersaud EN, Lu X, Micewicz ED, Chodon T, Atefi M, Kroeger N, Chandramouli GVR et al. Dendritic cell-based immunotherapy in prevention and treatment of renal cell carcinoma: efficacy, safety, and activity of Ad-GM·CAIX in immunocompetent mouse models. J Immunother 2013; 36:102-11; http://dx.doi.org/10.1097/CJI.0b013e31827bec97.
  • Kabbinavar FF, Zomorodian N, Belldegrun AS et al. A phase I, open label, dose escalation and cohort expansion study to evaluate the safety and immune response to autologous dendritic cells (DC) transduced with AdGMCA9 (DC-AdGMCAIX) in patients with mRCC. J Clin Oncol 2014; 32(4s): abstract 490.
  • Märten A, Ziske C, Schöttker B, Renoth S, Weineck S, Buttgereit P, Schakowski F, von Rücker A, Sauerbruch T, Schmidt-Wolf IG. Interactions between dendritic cells and cytokine-induced killer cells lead to an activation of both populations. J Immunother 2001; 24:502-10; http://dx.doi.org/10.1097/00002371-200111000-00007.
  • Wang D, Zhang B, Gao H, Ding G, Wu Q, Zhang J, Liao L, Chen H. Clinical research of genetically modified dendritic cells in combination with cytokine-induced killer cell treatment in advanced renal cancer. BMC Cancer 2014; 14:251; PMID:24720900; http://dx.doi.org/10.1186/1471-2407-14-251.
  • Sandoval F, Terme M, Nizard M, Badoual C, Bureau M-F, Freyburger L, Clement O, Marcheteau E, Gey A, Fraisse G et al. Mucosal imprinting of vaccine-induced CD8+ T cells is crucial to inhibit the growth of mucosal tumors. Sci Transl Med 2013; 5:172ra20; PMID:23408053; http://dx.doi.org/10.1126/scitranslmed.3004888.
  • Adotevi O, Vingert B, Freyburger L, Shrikant P, Lone Y-C, Quintin-Colonna F, Haicheur N, Amessou M, Herbelin A, Langlade-Demoyen P et al. B subunit of Shiga toxin-based vaccines synergize with α-galactosylceramide to break tolerance against self antigen and elicit antiviral immunity. J Immunol 2007; 179:3371-79; PMID:17709554; http://dx.doi.org/10.4049/jimmunol.179.5.3371.
  • Fenoglio D, Traverso P, Parodi A, Tomasello L, Negrini S, Kalli F, Battaglia F, Ferrera F, Sciallero S, Murdaca G et al. A multi-peptide, dual-adjuvant telomerase vaccine (GX301) is highly immunogenic in patients with prostate and renal cancer. Cancer Immunol. Immunother 2013; 62:1041-52; http://dx.doi.org/10.1007/s00262-013-1415-9.
  • Hercus TR, Thomas D, Guthridge MA, Ekert PG, King-Scott J, Parker MW, Lopez AF The granulocyte-macrophage colony-stimulating factor receptor: linking its structure to cell signaling and its role in disease. Blood 2009; 114:1289-98; PMID:19436055; http://dx.doi.org/10.1182/blood-2008-12-164004.
  • Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mari­ani L, Parmiani G, Rivoltini L et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of mela­noma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 2007; 25:2546-53; PMID:17577033; http://dx.doi.org/10.1200/JCO.2006.08.5829.
  • Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012; 1:894-907; PMID:23162757; http://dx.doi.org/10.4161/onci.20931.
  • Salazar AM, Erlich RB, Mark A, Bhardwaj N, Herberman RB. Therapeutic in situ autovaccination against solid cancers with intratumoral poly-ICLC: case report, hypothesis, and clinical trial. Cancer Immunol Res 2014; 2:720-24; PMID:24801836; http://dx.doi.org/10.1158/2326-6066.CIR-14-0024.
  • Källberg E, Vogl T, Liberg D, Olsson A, Björk P, Wikström P, Bergh A, Roth J, Ivars F, Leanderson T. S100A9 interaction with TLR4 promotes tumor growth. PLoS ONE 2012; 7:e34207; http://dx.doi.org/10.1371/journal.pone.0034207.
  • Pichler M, Hutterer GC, Stoeckigt C, Chromecki TF, Stojakovic T, Golbeck S, Eberhard K, Gerger A, Mannweiler S, Pummer K et al. Validation of the pre-treatment neutrophil-lymphocyte ratio as a prognostic factor in a large European cohort of renal cell carcinoma patients. Br J Cancer 2013; 108:901-07; PMID:23385728; http://dx.doi.org/10.1038/bjc.2013.28.
  • Derhovanessian E, Adams V, Hähnel K, Groeger A, Pandha H, Ward S, Pawelec G. Pretreatment frequency of circulating IL-17+ CD4+ T-cells, but not Tregs, correlates with clinical response to whole-cell vaccination in prostate cancer patients. Int. J. Cancer 2009; 125:1372-79; http://dx.doi.org/10.1002/ijc.24497
  • Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, Solary E, Le Cesne A, Zitvogel L, Chauffert B. 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; 56:641-48; PMID:16960692; http://dx.doi.org/10.1007/s00262-006-0225-8.
  • Lutsiak MEC, Semnani RT, De Pascalis R, Kashmiri SVS, Schlom J, Sabzevari H. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood 2005; 105:2862-68; PMID:15591121; http://dx.doi.org/10.1182/blood-2004-06-2410.
  • Pere H, Tanchot C, Bayry J, Terme M, Taieb J, Badoual C, Adotevi O, Merillon N, Marcheteau E, Quillien VR et al. Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer. Oncoimmunology 2012; 1:326-33; PMID:22737608; http://dx.doi.org/10.4161/onci.18852.
  • Jacobs JFM, Punt CJA, Lesterhuis WJ, Sutmuller RPM, Brouwer HM-LH, Scharenborg NM, Klasen IS, Hilbrands LB, Figdor CG, de Vries IJM et al. Dendritic cell vaccination in combination with anti-CD25 monoclonal antibody treatment: a phase I/II study in metastatic melanoma patients. Clin Cancer Res 2010; 16, 5067-78; PMID:20736326; http://dx.doi.org/10.1158/1078-0432.CCR-10-1757.
  • Adotevi O, Pere H, Ravel P, Haicheur N, Badoual C, Merillon N, Medioni J, Peyrard S, Roncelin S, Verkarre V et al. A decrease of regulatory T cells correlates with overall survival after sunitinib-based antiangiogenic therapy in metastatic renal cancer patients. J Immunother 2010; 33(9):991-8; PMID:20948437; http://dx.doi.org/10.1097/CJI.0b013e3181f4c208.
  • Tartour E, Pere H, Maillere B, Terme M, Merillon N, Taieb J, Sandoval F, Quintin-Colonna F, Lacerda K, Karadimou A et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy.Cancer Metastasis Rev 2011; 30(1):83-95; PMID:21249423; http://dx.doi.org/10.1007/s10555-011-9281-4.
  • Bose A, Lowe DB, Rao A, Storkus WJ. Combined vaccine+axitinib therapy yields superior antitumor efficacy in a murine melanoma model. Melanoma Res 2012; 22(3):236-43; PMID:22504156; http://dx.doi.org/10.1097/CMR.0b013e3283538293.
  • Joseph RW, Parasramka M, Eckel-Passow JE, Serie D, Wu K, Jiang L, Kalari K, Thompson RH, Huu Ho T, Castle EP et al. Inverse association between programmed death ligand 1 and genes in the VEGF pathway in primary clear cell renal cell carcinoma. Cancer Immunol Res 2013; 1:378-85; PMID:24778130; http://dx.doi.org/10.1158/2326-6066.CIR-13-0042.
  • Badoual C, Combe P, Gey A, Granier C, Roussel H, De Guillebon E, Oudard S, Tartour E. Signification et intérêt clinique de l'expression de PD-1 et PDL-1 dans les tumeurs. Medecine sciences 2013a; 29:10-12; http://dx.doi.org/10.1051/medsci/2013296005.
  • Fourcade J, Sun Z, Pagliano O, Chauvin J-M, Sander C, Janjic B, Tarhini AA, Tawbi HA, Kirkwood JM, Moschos S et al. PD-1 and Tim-3 regulate the expansion of tumor antigen-specific CD8+ T cells induced by melanoma vaccines. Cancer Res 2014; 74:1045-55; PMID:24343228; http://dx.doi.org/10.1158/0008-5472.CAN-13-2908.
  • Badoual C, Hans S, Merillon N, Van Ryswick C, Ravel P, Benhamouda N, Levionnois E, Nizard M, Si-Mohamed A, Besnier N et al. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res 2013; 73:128-38; PMID:23135914; http://dx.doi.org/10.1158/0008-5472.CAN-12-2606.
  • Sheikh N, Petrylak D, Kantoff PW, Dela Rosa C, Stewart FP, Kuan LY et al. Sipuleucel-T immune parameters correlate with survival: an analysis of the randomized phase 3 clinical trials in men with castration-resistant prostate cancer. Cancer Immunol Immunother 2013; 62(1):137-47; PMID:22865266; http://dx.doi.org/10.1007/s00262-012-1317-2.
  • Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbé C, Maio M, Binder M, Bohnsack O, Nichol G et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res 2009; 15:7412-20; PMID:19934295; http://dx.doi.org/10.1158/1078-0432.CCR-09-1624.
  • Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon R-A, Reed K et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013; 369:122-33; PMID:23724867; http://dx.doi.org/10.1056/NEJMoa1302369.
  • Schmidt SM, Schag K, Müller MR, Weinschenk T, Appel S, Schoor O, Weck MM, Grünebach F, Kanz L, Stevanovic S et al. Induction of adipophilin-specific cytotoxic T lymphocytes using a novel HLA-A2-binding peptide that mediates tumor cell lysis. Cancer Res 2004; 64:1164-70; PMID:14871853; http://dx.doi.org/10.1158/0008-5472.CAN-03-2538.
  • Sukov WR, Ketterling RP, Lager DJ, Carlson AW, Sinnwell JP, Chow GK, Jenkins RB, Cheville JC. CCND1 rearrangements and cyclin D1 overexpression in renal oncocytomas: frequency, clinicopathologic features, and utility in differentiation from chromophobe renal cell carcinoma. Hum Pathol 2009; 40:1296-1303; PMID:19386349; http://dx.doi.org/10.1016/j.humpath.2009.01.016.
  • Saino M, Maruyama T, Sekiya T, Kayama T, Murakami Y. Inhibition of angiogenesis in human glioma cell lines by antisense RNA from the soluble guanylate cyclase genes, GUCY1A3 and GUCY1B3. Oncol Rep 2004; 12:47-52; PMID:15201957.
  • Salagierski M, Verhaegh GW, Jannink SA, Smit FP, Hessels D, Schalken JA. Differential expression of PCA3 and its overlapping PRUNE2 transcript in prostate cancer. Prostate 2010; 70:70-78; PMID:19760627; http://dx.doi.org/10.1002/pros.21040.
  • Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin A-M, Figlin RA, Hutson TE, Sternberg CN, Amado RG et al. Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials. Lancet Oncol 2012; 13, 827-37; PMID:22759480; http://dx.doi.org/10.1016/S1470-2045(12)70241-3.
  • Silini A, Ghilardi C, Figini S, Sangalli F, Fruscio R, Dahse R, Pedley RB, Giavazzi R, Bani M. Regulator of G-protein signaling 5 (RGS5) protein: a novel marker of cancer vasculature elicited and sustained by the tumor's proangiogenic microenvironment. Cell Mol Life Sci 2012; 69:1167-78; PMID:22130514; http://dx.doi.org/10.1007/s00018-011-0862-8.
  • Fang YJ, Lu ZH, Wang GQ, Pan ZZ, Zhou ZW, Yun JP, Zhang MF, Wan DS. Elevated expressions of MMP7, TROP2, and survivin are associated with survival, disease recurrence, and liver metastasis of colon cancer. Int J Colorectal Dis 2009; 24:875-84; PMID:19421758; http://dx.doi.org/10.1007/s00384-009-0725-z.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.