Skip to main content
  • Meeting report
  • Open access
  • Published:

Perspectives in immunotherapy: meeting report from the “Immunotherapy Bridge”, Napoli, December 5th 2015

Abstract

Harnessing the immune system and preventing immune escape, the immunotherapy of cancer provides great potential for clinical application, in broad patient populations, achieving both conventional and unconventional clinical responses. After the substantial advances in melanoma, the focus of cancer immunotherapy has expanded to include many other cancers. Targeting immune checkpoints and further mechanisms used by tumors to avoid anticancer immunity, different approaches are under evaluation, including combination therapies.

The first Immunotherapy Bridge meeting focused on various cancer types including melanoma, non-small cell lung cancer, renal cell, breast and ovarian carcinoma, and discussed mechanisms of action of single agents and combination strategies, and the prediction of clinical responses.

Introduction

The potential of immunotherapy to improve clinical outcome in various solid and haematologic malignancies is based on the ability of the immune system to exhibit both innate (i.e., myeloid and lymphoid cells exerting a rapid effector function) and adaptive responses (driven by T- and B-lymphocytes expressing antigen receptors produced by site-specific somatic recombination and endowed with greater specificity retaining antigen memory) [1]. The approval of sipuleucel-T for the treatment of prostate cancer was the first immuno-therapy to be approved, followed by the approval of ipilimumab (anti-CTLA-4), for the treatment of metastatic melanoma and, later, of the anti PD-1 antibodies nivolumab (in melanoma, non-small cell lung cancer, NSCLC, kidney cancer, head & neck cancer), pembrolizumab (in melanoma, NSCLC and Head & Neck cancer), and talimogene laherparepvec (T-VEC) in melanoma (the first oncolytic viral therapy approved for treatment).

The clinical practice with checkpoint inhibition and pathways activation has highlighted a number of important considerations. Firstly, immunotherapy agents may be associated with patterns of response that differ from those seen with other treatment modalities (i.e., chemotherapy) and so their use may require the development of different response criteria [2]. A response to anti-CTLA-4 or anti-PD-1 may be observed after an initial increase in tumor volume and a reduction in tumor burden may be observed after the appearance of new lesions. These observations suggest that treatment should be continued regardless of an increase in volume of existing lesions or the early appearance of new lesions. Even more remarkable, durable responses was observed even after cessation of treatment, suggesting an ability of immunotherapy to reset the equilibrium between host and tumor [3]. As an example, it is now accepted that ipilimumab therapy in advanced melanoma presents a plateau in the survival curve, beginning at approximately 3 years [4]. Nevertheless, current criteria (e.g., RECIST) might not be appropriate to asses these types of responses. Another important consideration is that immunotherapies can act regardless of patient characteristics and tumor characteristics [5, 6]. Various approaches to identify potential biomarkers for immunotherapies are being evaluated. A promising area of research is the use of various immunotherapies in combination with one another and/or with chemotherapy, targeted agents or radiation.

Cancer vaccines revisited

Using a vaccination with CpG 7909 (PF 676), a 23-bp oligodeoxynucleotide TLR-9 agonists, and a human Melan-A peptide antigen (analogue, ELAGIGILTV) emulsified in IFA (Incomplete Freund’s adjuvant), rapid and efficient T cell responses were found in all vaccinated metastatic melanoma patients, with a frequency 10-fold higher than those vaccinated without CpG. The- peak response was 7 to 11 days after booster injection. Furthermore, effector memory (and central) T cells expressing perforin, granzyme B, IFNγ and also CD28-effector cells were generated. Finally, the killing of melanoma cells and IFNγ secretion were observed [7, 8]. More recently robust and sustained CD4 and CD8 T cell responses were found to be induced by vaccination with the long synthetic NY-ESO79–108 peptide, CpG and IFA (Baumgaertner P: Vaccination of melanoma patients with long NY-ESO-1 peptide and CpG-B elicits robust T-cell responses with multiple specificities and a novel DR-7-restricted epitope, Submitted). Of the three polypeptide-based cancer vaccines in advanced clinical testing of a recombinant fusion nearly full length MAGE-A3 protein, failed the first primary endpoint in both lung cancer and melanoma, while HPV-16 long synthetic peptides (LSPs) obtained 50 % complete responses in premalignant lesions, but no impact on overall survival (OS) was found in cervical carcinoma [9], and, finally, IMA 109 (a mix of 9 short naturally presented peptides) followed a similar pattern as a clinical efficacy signal was detected in a randomized phase II clinical trial in renal cell carcinoma [10], but failed the primary efficacy endpoint in the phase III trial. Hurdles in vaccination with polypeptides include, first of all, relatively weak T cell responses (narrow focused and short lived), then inadequate induction of CD8 T cells by LSPs and recombinant proteins (cross priming), and, finally, local immunosuppression. Polypeptide-based vaccines still need to be optimized, however they already showed that (1) LSP (and proteins) depend on cross-presentation for vaccination, (2) that XCL-1 may be used as vaccine component to target antigen to XDCs and (3) that fusion XCL-1 proteins are interesting candidates to achieve consistently strong CD8 T cell responses and protective immunity. Also, combination with nanoparticle delivery is a promising approach. Regarding the translation into clinic, toll-like receptor (TLR) agonists as polypeptide vaccine adjuvants CpG-ODNs and Poly(I:C) have been shown to dramatically increase the antigen-specific effector T cell (Teff):Treg ratio in the lymphoid organs, skewing immune response in favor of a functional anti-tumor effect. In addition, CpG-ODNs and Poly(I:C) rapidly induce a Th1 polarized cytokine milieu, which fits with a reported mechanism for the adjuvant activity of TLR ligands [11]. On the other hand, HER-2-CD1d or CEA-CD1d fusion proteins efficiently function both as vaccine adjuvants and driving Th1 shifts in the tumor microenvironment, partly through transactivation of NK cells, thus harnessing NKT for vaccination [12].

Unlike viral antigen specific T cell responses, molecularly defined subunit vaccines (antigen and adjuvants) may not efficiently induce long lived memory cells. Yet, robust tumor antigen-specific T cell memory responses are needed to sustain durable tumor responses. A deep understanding of the signals involved in instructing T cell memory differentiation should provide clues for the identification of compounds that, added to subunit vaccines, may modulate the quantity and quality of memory T cells. In this regard, inhibition of mTOR signaling by administration of rapamycin during the expansion phase of a primary anti-viral CD8 T cell response augments the size of the memory T cell pool during the contraction phase [13]. The mTOR signaling branches into two major complexes, mTORC1 and mTORC2. Conditional genetic inactivation of mTORC1 in T cells leads to a profound reduction of the primary CD8 T cell response affecting the generation of both effector and memory T cells. In marked contrast, the conditional deletion of a functional mTORC2 complex leads to decreased differentiation of short lived effector cells and a corresponding increase in the formation of memory effector precursor cells. This translates into enhanced recall responses to rechallenge with antigen [14]. This observation provides the rationale to dissect the signaling events downstream of mTORC2 leading to instruction of memory T cell differentiation. Nuclear accumulation of the transcription factor FOXO1 is one of the key events. Future research should reveal the specific transcriptional reprogramming events controlled by FOXO1 which lead to memory formation. Similar signaling events may operate in human CD8 T cells [15].

Antigen-specific T cells, MDSCs and survival in melanoma and breast cancer

Monitoring the immune status of cancer patients using peripheral blood assays has several advantages over tumor-infiltrating cells, such as being minimally invasive, repeatable ad libitum, usable to generate functional as well as phenotypic data. The over-riding caveat is that, like any biomarker, it is only relevant if robustly associated with clinical outcome. In a phase II study of a 10-peptide GM-CSF-adjuvant vaccine in renal cell cancer (RCC) with or without the addition of single low-dose cyclophosphamide, Walter and colleagues showed that a T cell response to 3 or more vaccine peptides was indicative of 100 % survival [10]. Observed reductions of peripheral Tregs with a single dose of cyclophosphamide suggests that suppressive cells might be involved in the response to cancer antigens in these patients. Also, the relative abundance of cells with the phenotype CD14 + HLA-DRlow/negative influenced survival in the RCC [10]. Stage IV melanoma patients, on the other hand, possessed higher frequencies of circulating CD4+ Tregs relative to healthy controls, but there was no correlation with survival [16]. In the same study, it was found that the frequencies of CD14 + CD11b + HLA-DR−/low monocytic myeloid-derived suppressor cells (MDSCs) were higher in patients compared to controls and that their levels did correlate negatively with survival. The potential for enhancing the strength of this correlation of MDSC with survival by defining this cell type more accurately was investigated in a pilot study using 45-channel mass spectrometry flow cytometry (CyTOF) on cells preselected for being CD3-, CD19-, CD11b+, CD14+ and CD45+. Comprehensive immune signatures including MDSCs were found in patients relative to controls, which correlated more closely with survival (Wistuba-Hamprecht et al. under revision).

Importantly, the in vitro reactivity of peripheral T cells to tumor-associated antigens (TAA) such as NY-ESO-1 in patients with melanoma with distant metastases is positively associated with a survival benefit [17]. Indicating the complexity of peripheral immune signatures and their correlations with survival, CD8+ T cell reactivity to Melan-A was also associated with survival, but only in the absence of a response of CD4+ T cells producing IL 4 or IL 17 on stimulation with this TAA. Consequently, the presence of CD4+ Melan-A-specific T-cells was in general associated with poorer survival [18]. Similarly, in breast cancer, survival was associated with a CD8+ T cell response to Her-2 peptides and a negative role of both MDSCs and Tregs was demonstrated in the prognosis of these elderly breast cancer patients. The mechanism might be through dampening favorable CD8+ T cell immune responses to tumor-associated antigens [19].

In summary, functional responses of patient peripheral blood to tumor antigens measured in vitro, as well as surface marker phenotyping ex vivo, can predict survival duration. Specificity of the response, nature of the responding T cells and pro- or anti-inflammatory type of response, as well as proportions and types of MDSCs, are all important factors for patient survival in both melanoma and breast cancer. Thus, immune monitoring of peripheral blood identifies predictive biomarkers for patient survival and may provide insights into the mechanisms involved in immune-mediated anti-cancer effects.

Counteracting immune suppression by modulation of checkpoint blockade and inhibitors of CSF-R1

One of the methods used to improve adoptive T cell therapy is the re-stimulation of the injected TIL (Tumor-infiltrating lymphocyte) cells with tumor vaccine expressing same antigen as recognized by TILs to improve life-span. In a phase I clinical trial Poschke et al. [20] combined a dendritic cell (DC) vaccination and TIL infusion in malignant melanoma. The treatment with the two cellular products was found feasible and safe for the eight patients with stage IV melanoma enrolled in the trial. Although the small study size precluded analysis of clinical responses, one patient showed a complete remission and two had stable disease. The analysis of T-cell receptor repertoire in TIL by next-generation sequencing revealed presence of highly dominant clones in most infusion products which could, in the majority of cases, be detected in the circulation by qPCR after T-cell transfer. Another trial combining DC vaccination and adoptive T cell transfer in a two-step approach in patients preconditioned with non-myeloablative chemotherapy is ongoing (EUdrac-CT #2012-000450-63).

Also, a combination of IFN-γ and multiple Toll-like receptor agonists proved superior in activating DCs to tumor-specific activation, thus permitting an improvement of the quality of the DC vaccine. Even the possibility of combining adoptive T cell transfer with silencing checkpoint molecules was explored. Antibodies represent one possibility, but another intriguing option is a new technology by which the checkpoint molecules could be directly silenced in the injected T cells. Hydrophobically modified “self-deliverable” small RNA (sdRNA) can effectively enter cells and results showed that PD-1 silencing in healthy T cells by using sdRNA leads to significant decrease in surface and as well intracellular levels of PD-1 expression in TILs. Activated PD-1 silenced healthy T cells and TILs produced more IFN-γ upon tumor specific stimulation. In addition, PD-1 silenced TILs proved enhanced capacity to kill autologous tumor cells and, finally, the percentage of CD45RA- CCR7- effector memory CD8+ and CD4+ TILs increased after PD-1 silencing. MDSCs are being evaluated as a new target for cancer immunotherapy and one of the unanswered questions is whether checkpoint blockade mediate some of their effects by targeting immune suppressive cells (Tregs, MDSCs, macrophages) or their products. Anti-CTLA4 treatment results in a strong reduction of immuno-suppressive cells (GrMDSC, Tregs) and agents (Arginase, iNOS) in the majority of treated patients [21].

Neuroblastoma is the most common extracranial solid cancer type in childhood, and high-risk patients have poor prognosis. Similar to malignant melanoma, neuroblastoma-driven inflammation contributes to the induction of suppressive myeloid cells that hamper efficient antitumor immune responses. Infiltrating CSF-1R+ myeloid cells predicted poor clinical outcome in patients with neuroblastoma (R2: microarray analysis and visualization platform [22]). In vitro, neuroblastoma-derived factors interfered with early development of myeloid cells and enabled suppressive functions on human monocytes through M-CSF/CSF-1R interaction [23]. In a transgenic mouse model (TH-MYCN) resembling high-risk human neuroblastoma, antagonizing CSF-1R with a selective inhibitor (BLZ945, Novartis) was found to modulate the induction of human and murine suppressive myeloid cells and to efficiently limit tumor progression. Checkpoint inhibitors as monotherapy was found to be insufficient in controlling tumor growth, while combining BLZ945 with PD-1/PD-L1 blocking antibodies resulted in superior tumor control.

Response and resistance to PD-1 pathway blockade: clues from the tumor microenvironment

Monoclonal antibodies blocking anti-PD-1 immune checkpoint showed an unprecedented spectrum of activity versus different cancer types providing a “common denominator” for cancer therapy [24]. On this note, the US food and drug administration (FDA) already approved their usage for the treatment of patients with metastatic melanoma, advanced NSCLC, RCC and Hodgkin’s lymphoma. Great attention is now focused on identifying biomarkers able to predict response or resistance to anti-PD-1 treatment [25]. It has been previously identified that PD-L1 expression on the tumor cell surface is one factor associated with the clinical activity of anti–PD-1 [26]. Notably, a significant number of patients with PD-L1+ expression still do not respond to PD-1 pathway blockade, suggesting that additional intratumoral factors may influence treatment outcomes.

In melanoma, by comparing PD-L1+ vs. PD-L1(−) tumors, it has been shown an overexpression of genes involved in CD8+ T-cell activation (e.g., CD8A, IFNG) and immunosuppression pathways (e.g., PD-1, LAG3 and IL10) in PD-L1+ tumors, thus suggesting the coordinate expression of multiple immunological factors mediating therapy response to anti PD-1 blockade [27]. However, a deep molecular analysis performed on formalin-fixed, paraffin-embedded (FFPE) pretreatment tumor biopsies expressing PD-L1 (PD-L1+) and derived from patients with RCC showed that none of the immune molecules previously found to be significantly overexpressed in PD-L1+ melanomas was associated with anti-PD-1 treatment response. On the contrary, additional immunologic factors and novel metabolic molecules were found to be associated with treatment resistance in RCC. Particularly, genes involved in metabolic and solute transport functions, also found in kidney cancer cell lines, were found associated with treatment failure of patients with PD-L1+ RCC. Conversely tumors from responding patients overexpressed immune markers such as BACH2, a regulator of CD4+ T cell differentiation, and CCL3 involved in leukocyte migration [28]. Thus, the intratumoral balance between metabolic and immunologic gene expression might determine the effective response to anti-PD-1 blockade in RCC. This is not surprising if we consider that RCC has been characterized as a metabolic disease, with the signature up-regulation of factors adapting to hypoxia and functioning to meet the bioenergetic demands of cellular proliferation [29]. These evidences suggested that the general approach used to identifying markers predicting clinical response to anti-PD-1-targeted therapies, which has mostly focused on modulatory receptors and ligands (e.g., PD-1, PD-L2, LAG-3) and T cell infiltrates, must be revaluated. Indeed a deeper level of investigation might be warranted for individual tumor types to which these new therapies are being applied.

Understanding and combining CD137-targeted immunotherapy

Hepatocellular carcinoma (HCC) is typically an inflammation-associated cancer that can be immunogenic [30]. Association of hepatitis C and hepatitis B infection with upregulation of PD-1 has been reported [31, 32], but it is associated with poor outcomes [33]. Immune checkpoint inhibition (anti-CTLA-4) has shown encouraging activity in an early clinical trial in HCC [34] and also PD-1 blockade with nivolumab may boost host immunity against HCC and improve clinical outcomes. Preliminary data from a phase I/II study on safety and antitumor activity of nivolumab show durable though partial response and an OS rate at 6 months of 72 % [35].

CD137 is a surface glycoprotein involved in T cell co-stimulation, with functions in T cell proliferation, inhibition of apoptosis, cytotoxic activity enhancement and cytokine production. Treatment with anti-CD137 agonist monoclonal antibodies can overcome tumor antigen tolerance [36]. On the other hand, mouse experimentation provided both a rational and a recipe for immunotherapy combinations: (1) procedures aimed at enhancing the number of primed nonanergic T cells in the organism by means of vaccines, adoptive transfer, or procedures to enhance the immunogenicity of a tumor lesion, and (2) strategies to de-repress or stimulate artificially the ongoing immune response, for the most part considering immunostimulatory monoclonal antibodies [37]. Based on this, co-infusion of both agonist antibodies and T cells has been shown to attain marked synergistic effects that result from more focused and intense cytolytic activity visualized under in vivo microscopy and from more efficient entrance of T cells into the tumor through the vasculature. These several levels of dynamic interaction between adoptive T cell therapy and CD137 offer much opportunity to raise the efficacy of current cancer immunotherapies [38]. In particular, anti-CD137 infusion may enhance adoptive immunotherapy by CTL by a dual mechanism, enhancing endogenous effector functions as well as focusing and prolonging the efficacy of therapeutically transferred lymphocytes [39]. New studies, including cross-priming of CD137 with DC and combination of immunostimulatory monoclonal antibodies anti-hCD137 (urelumab) with anti-hPD-1 (nivolumab) are presenting promising results.

Combination immunotherapy with autologous stem cell transplantation, protein immunization, and PBMC reinfusion in myeloma patients

Multiple myeloma is rich in tumor antigens of the cancer-testis (CT) family conferring spontaneous immunogenicity, with CT7 (MAGE-C1) and MAGE-A3/6 already known to correlate with plasma-cell proliferation [40] and with NY-ESO-1 eliciting spontaneous humoral and T-cell immunity [41]. Antibody responses to CT antigens were found after treatment with allogeneic stem cell transplant in multiple myeloma patients, which might help to achieve long-lasting remissions [42]. Autologous Stem Cell Transplant (ASCT) is a standard of care for some multiple myeloma patients but not curative. ASCT might be used as a platform for adoptive immunotherapy, considering that myeloablative therapy may provide immunologic benefits, such as tumor antigen release for cross-presentation and priming, lymphopenia-induced proliferation of T cells, and elimination of suppressor populations (Tregs, MDSCs). Nevertheless, ASCT also results in durable immunosuppression, as evidenced by frequent CMV reactivation. Combining vaccination and ASCT was therefore investigated for CT antigen MAGE-A3, taking into account potential benefits and risks of MAGE-A3 targeting in adoptive transfer, reported to lead to clinical tumor regression in 5 out of 9 metastatic cancer patients (7 with metastatic melanoma, 1 with synovial sarcoma, 1 with esophageal cancer) but also neurological toxicity associated with lymphocytic brain infiltration, probably due to cross-reactivity with an epitope from MAGE-A12 expressed in human brain [43]. A pilot study of MAGE-A3 antigen-specific cancer immunotherapeutic (recMAGE-A3 protein + AS15) as consolidation for multiple myeloma patients undergoing ASCT has recently completed accrual, and was safe and well tolerated. Using mass cytometry for phenotypic analysis of immune reconstitution, preliminary results show an inversion of CD4 / CD8 ratio and a B cell dip and rebound after ASCT. Nevertheless, antigen-specific assays (ELISA, ELISPOT, Intracellular cytokine staining) for MAGE-A3 showed induction of a strong antibody and Th1-biased CD4 T cell response in all patients. In conclusion, a combined approach of vaccination with ASCT is feasible and overcomes typical immunosuppression in multiple myeloma patients. Absence of MAGE-A expression observed in some patients upon disease relapse suggests potential immune selective pressure, and calls for combination strategies to limit antigen escape.

Anti-cancer immunity despite T cell “exhaustion”

The three main stumbling blocks for anti-cancer T cells are (1) low T cell numbers and low TCR affinity (with ACT and vaccination as possible solutions), (2) inefficient priming/boosting (with checkpoint blockade or vaccination as possible solutions), and (3) T cell suppression in the TME (with checkpoint blockade, radiation, or intratumoral injection of “adjuvants” as possible solutions) [44].

Tumor antigen specific T cells in the TME only produce low levels of cytokines, thus their effector functions are relatively weak. For the first time in any species, the Speiser group analyzed T cells from a TME by gene expression array [45], demonstrating that the molecular profile significantly resembled the one of T cell “exhaustion” that was first defined in chronic mouse and then human infections [46]. During chronic infection, T cells progressively lose their functional properties. T cell exhaustion is also accompanied by a progressive increase in the amount and diversity of inhibitory receptors expressed. Altered inflammation and changes in immunoregulatory cytokines can have important roles. Ultimately, if the severity and/or duration of the infection is high and/or prolonged, virus-specific T cells can be completely eliminated, leading to loss of protective T cell responses. The severity of T cell exhaustion correlates with inhibitory receptor expression, viral (and antigen) load, loss of CD4+ T cell help, and infection duration. Similar observations were made for metastatic melanoma, the disease studied by the group. They showed this by molecular profiling of CD8+ T cells not only in humans, but also in an autochthonous mouse melanoma model, finding several corresponding molecular hits [45, 47, 48]. They also demonstrated a remarkable coexistence of fully functional effector cells in circulation and exhausted cells in the tumor environment. An important hit, i.e., a gene that was significantly overexpressed in TIL was Maf, an AP-1 family member. Subsequent experiments demonstrated that Maf is a first in-class driver of exhaustion [47, 48], as Maf overexpression reproduces and enhances a large number of molecular and functional features of exhausted cells, and deletion of Maf reverses exhaustion as T cells become strongly functional and eliminate cancers.

Another aspect studied by the group is that exhausted cells still have some functional competence, indicating that exhaustion is not an optimal term, T cell “dysfunction” might be better, or “long term effector T cell population”, because these cells are heterogeneous and likely contain short and (very) long lived cell subsets [49]. In fact, absence of “exhausted” cells renders diseases worse, both in chronic infections and in various cancers. Therefore, the group proposes that exhausted cells are specialized with respect to differentiation and activation, adapted to the disease in which these T cells stably adjust their effector capacity to the conditions of chronic infection or cancer. This phenotype is optimized to cause minimal tissue damage while still mediating a critical level of pathogen control. This concept provides a rationale for emerging therapies that enhance T cell activity in chronic infection and cancer e.g., by blocking inhibitory receptors [49]. Clearly, many dysfunctional cells can regain function within short time, as revealed by strong cytotoxicity by those T cells [50].

The group also studied T cell responses from melanoma patients treated with the anti-CTLA-4 antibody Ipilimumab. Long-term observations suggest that responder patients progressively had less Tregs in blood, and particularly also in tumors. Furthermore, responders had higher levels of non-classical CD16+ monocytes compared with non responder patients. As CD16 binds Ipilimumab, experiments were performed to determine whether these CD16+ monocytes could mediate antibody-dependent cell- mediated cytotoxicity (ADCC). Indeed, this was the case, leading to specific killing of Tregs due to their constitutive CTLA-4 expression at high levels [51]. Another paper, a collaborative work performed in the lab of Pia Kvistborg, Ton Schumacher et al., reports the analysis of melanoma patients’ PBMC with an HLA-A2 tetramer panel comprising 145 tumor antigens. The study shows that anti–CTLA-4 treatment induced a significant increase in the number of detectable CD8 T cell specificities. Thus, the therapy induced considerable broadening of the immune response, demonstrating that Ipilimumab may not primarily act on effector cells but rather on other immune cells, likely Tregs and DCs that can mobilize many novel T cell populations that help fighting cancer [52]. Another paper describes that melanoma patients treated with ipilimumab experienced a significant increase in the number of detectable CD8 T cell specificities. Thus, the therapy induced considerable broadening of the immune response, demonstrating that ipilimumab may not primarily act on already activated effector T cells but rather on other immune cells, likely Tregs and DCs that can activate many novel T cell populations that help fighting cancer [52]. This study was a collaborative work performed in the lab of Pia Kvistborg, Ton Schumacher et al., who developed and applied an HLA-A2 tetramer panel detecting CD8 T cells with 145 different antigen specificities.

Predictor of response to radiation and immunotherapy

An in vitro model recently showed that radiotherapy (RT) produces both a radiation dose-dependent induction and chemotherapeutic enhancement of immunogenic cell death (ICD) as well as induction of ICD by the combination of radiation and certain chemotherapy agents [53]. These findings provide preliminary evidence that ICD stimulated by either high-dose radiotherapy alone, or concurrent chemo-radiation regimens, may contribute to the establishment of a peritumoral proimmunogenic milieu [53]. In combination with different forms of immunotherapy, RT stimulates the release of antigens and also the signals in the tumor microenvironment that allow T cells to permeate the surviving tumor, and contribute to cell killing so that the tumor is converted into an in situ vaccine. Since 2004, the combination of DC growth factors in combination with RT were found to induce abscopal effects in a syngeneic mouse model [54]. These findings were translated to the clinic, where a combination of radiotherapy with granulocyte-macrophage colony-stimulating factor produced objective abscopal responses in 11 out of 41 patients with metastatic solid tumors refractory to treatment [55]. The results of this clinical trial also highlighted the importance of role of host’s “immune integrity” in the process, with more patients responding among those with a neutrophil to lymphocyte ratio <4. A similar translation of preclinical to clinical results was demonstrated with a combination of immune checkpoint blockade and radiotherapy. In the 4 T1 syngeneic mouse model of metastatic mammary carcinoma, it was shown that RT to a peripheral tumor and CTLA-4 blockade achieved both reduction of lung metastases and improved survival compared either modality alone [56]. Consistently, in a patient with chemotherapy-refractory metastatic adenocarcinoma of the lung, an abscopal response was seen 2 ½ months after the start of treatment with ipilimumab and fractionated RT, supporting the belief that a combination of RT and immunotherapy might prove to be a useful strategy to improve the outcomes of some cancer patients with metastases that are historically known to have dismal prognoses [57].

RT alone is seldom capable of inducing rejection of aggressive poorly immunogenic tumors. RT ability to induce tumor rejection is dependent on the balance of positive and negative signals that pre-exist or are induced by RT itself. Barriers to the potential of radiation to convert a tumor into an in situ vaccine might be associated with classical immunosuppressive microenvironment of established tumors or be RT-induced immunosuppressive effects. One of the immunosuppressive signals generated by RT is the activation transforming growth factor (TGF) β [58]. Because TGFβ regulates many processes that can affect tumor growth, including angiogenesis, a comprehensive assessment of the major changes by gene expression analysis was performed. The top 500 genes selectively upregulated in RT + TGF beta blockade treatment group were identified using the Pavlidis Template Matching Method, defining a majority of genes involved in the immune response. The concept was investigated in a trial of RT with fresolimumab in metastatic breast cancer, that failed to show responses with the combination. The role of adaptive immune resistance was in limiting tumor response to radiotherapy and TGFβ blockade was investigated in a mouse model: increased PDL-1 and PDL-2 expression on tumor and myeloid cells was demonstrated, compared to controls. PD-1 blockade extended survival and delayed tumor recurrence in mice treated with radiation and TGFβ blockade [59], suggesting that PD-1 blockade may be required to unleash the effect of combining TGFβ blockade and RT. In summary, results of RT and immunotherapy combinations from preclinical experiments in syngeneic models of different murine tumors were consistently confirmed in the clinic. RT remains a powerful adjuvant to multiple immunotherapy strategies.

Checkpoint inhibitors in metastatic non-small cell lung cancer

With limited current options for the treatment of advanced NSCLC and phase III studies of vaccines that to date have failed to meet their primary endpoint, great attention is being paid to checkpoint inhibitors. A double-blind, phase 2 randomized controlled trial in treatment-naïve patients with advanced NSCLC, proved that phased ipilimumab plus chemotherapy improved both irPFS (immune-related PFS) and PFS. Overall rates of grade 3 and 4 immune-related adverse events were 15 % (control 6 %) [60]. Nivolumab monotherapy in a phase II single arm trial found meaningful activity (15 % ORR) and a manageable safety profile (17.1 % grade 3/4 adverse events) in previously treated patients with advanced, refractory, squamous NSCLC [61]. Nivolumab was tested versus chemotherapy in a multicenter, open-label, phase III, randomized trial in previously-treated patients with advanced or metastatic squamous NSCLC, and overall survival (mOS 9.2 vs 6.0), response rate, and progression-free survival were significantly better with nivolumab, regardless of PD-L1 expression level [62]. The corresponding trial in metastatic non-squamous NSCLC (multicenter, open-label, phase III, randomized trial in previously-treated patients) was stopped earlier due to efficacy and nivolumab was associated with even greater efficacy (e.g., mOS 12.2 vs 9.4) than docetaxel across all end points in subgroups defined according to prespecified levels of tumor-membrane expression of the PD-1 ligand. Treatment-related adverse events of grade 3 or 4 were reported in 10 % of patients in the nivolumab group, as compared with 54 % of those in the docetaxel group [63].

The phase I study of pembrolizumab in monotherapy in patients with advanced NSCLC, it had an acceptable side-effect profile and showed antitumor activity in patients (median duration of response exceeds 1 year). Also, PD-L1 expression in at least 50 % of tumor cells correlated with improved efficacy of pembrolizumab [64]. A multicenter, open-label, phase II/III, randomized trial in previously-treated patients with PD-L1-positive NSCLC, demonstrated that pembrolizumab prolongs overall survival (mOS 10.4 with pembrolizumab 2 mg/kg, 12.7 with pembrolizumab 10 mg/kg, 8.5 with docetaxel) and has a favorable benefit-to-risk profile in this selected population [65]. In the multicenter, open-label, phase 3, randomized trial in treatment-naïve patients with PD-L1+ advanced or metastatic NSCLC, pembrolizumab showed a manageable safety profile and promising antitumor activity [66]. Preliminary data from a phase II trial of pembrolizumab in untreated brain metastases in NSCLC showed antitumoral activity (33 % brain metastasis response rate) and good tolerability [67]. In a multicenter, open-label, phase II, randomized trial in previously-treated patients with NSCLC, a pattern of improved survival that correlates with PD-L1 expression and a good safety profile were reported.

PD1/PDL1 Inhibition in metastatic renal cell carcinoma

The phase III study of nivolumab versus everolimus demonstrated an overall survival benefit for patients with advanced RCC treated with nivolumab, (mOS 25.0 vs 19.6) and was generally well tolerated with a low rate of grade 3/4 events. Prespecified subgroup analyses did not identify a population which did not benefit from nivolumab in terms of overall survival (including patients with PD-L1 negative tumors). On the other hand, a clinically meaningful and statistically significant improvement in quality of life was seen with nivolumab versus everolimus for the duration of the study [68]. The role of treatment beyond progression which was described initially in a phase II dose ranging study in RCC, is still being explored at this time since some patients experience pseudoprogression on PD1 inhibition [69]. The dual immune checkpoint blockade with nivolumab /ipilimumab therapy is showing an encouraging activity with unprecedented ORR of about 40 % [70]. The combination of nivolumab with the vascular endothelial growth factor (VEGF) pathway inhibitors sunitinib or pazopanib in mRCC has shown promising activity as well, but was associated with significant liver toxicity [71]. The association of immunotherapy with more selective VEGF pathway inhibitors was investigated in a phase Ib study with MPDL3280A and bevacizumab with good safety and efficacy outcomes [72]. Phase II and III trials combining immunotherapy and VEGF pathway inhibitors are currently ongoing and will most likely transform first line therapy in the near future. However, a number of questions (e.g., regarding who benefits the most, biomarkers availability, dosing schedule/cost saving opportunities) still need to be answered.

Immune checkpoint inhibition for breast cancer

Breast cancer is the most common malignancy diagnosed in women worldwide. Approximately 15 % of breast cancers are triple-negative. As triple-negative breast cancers (TNBC) lack expression of ER, PR, and HER2, they do not benefit from available targeted therapies. TNBC is associated with a younger age at diagnosis, an advanced stage at diagnosis, and a worse clinical outcome. The rationale for investigating immunotherapy for TNBC comes from a number of observations made over the past several years. For one, we know that ER negative tumors have a higher density of tumor infiltrating lymphocytes (TILs) than ER positive tumors [73], which can in turn facilitate and immune response. Mittendorf and colleagues found that TNBCs have increased PD-L1 expression, which can suppress T cell function [74]. Gene expression profiling has revealed at least six distinct subtypes of TNBC, including an immunomodulatory subtype which is characterized by elevated expression of genes involved in T cell function [75]. And finally, the genomic instability and high mutation rates in TNBCs can produce neoantigens that induce an immune response [76]. Based on these observations, immune checkpoint inhibition was studied in TNBC. The phase Ib multi-cohort KEYNOTE-12 study enrolled patients with PD-L1 positive TNBC. The 32 patients enrolled were treated with single-agent pembrolizumab at 10 mg/kg every 2 weeks, which demonstrated an acceptable safety profile (events of a potentially immune-mediated nature, regardless of attribution, included pruritus, hepatitis, and hypothyroidism), and was associated with prolonged responses in 18.5 % of patients enrolled in the study [77]. Preliminary data from a phase Ia study of the PD-L1 inhibitor atezolizumab demonstrated a similar safety and tolerability profile, as well as early signs of durable clinical activity with a RR of 19 % [78]. PD-1 or PD-L1 blockade in TNBC is associated with a response rate approaching 20 %, and while somewhat lower than the RR in melanoma (30-40 % range), is comparable to monotherapy response rates in other solid tumors. The responses are durable and immune checkpoint inhibitors are safe and tolerable, with the majority of side effects being mild and easily managed. Future work is aimed at building on these promising monotherapy responses with the combination of different checkpoint inhibitors or with radio/chemo-therapy.

The role of immunotherapy in ovarian cancer

Despite advances in combinatorial chemotherapy regimens, current therapeutic options for ovarian cancer (OC) patients are inadequate and the majority of patients will relapse and die of disease, with a 5-year survival of approximately 30 %. The strong presence of TILs is strongly, positively associated with improved survival, the activity of TILs (CD3+ and CD8+ cells) as antitumor effectors and the identification of tumor-associated antigens (TAAs) that up-regulate in tumor tissue and ascites of OC patients (Her2/neu, Folate Receptor α, p53, CA125, members of the cancer-testis antigen family like MAGE –A4 and NY-ESO-1), all account for the evidence of OC immunogenicity.

The immunohistochemical analysis of frozen specimens from advanced-stage ovarian carcinomas showed significant differences in the distributions of progression-free survival and overall survival according to the presence or absence of intratumoral T cells. Patients whose tumors contained T cells had a median duration of progression-free survival of 22.4 months and a median duration of overall survival of 50.3 months, as compared with 5.8 and 18.0 months, respectively, among patients whose tumors did not contain T cells [79]. Later, intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio were found to be associated with favorable prognosis in OC (mOS 55 vs 26 months) [80]. When taking into account selected published studies that evaluated the association between TIL and patient survival, the prognostic value of TILs persisted among populations with diverse histologic tumor subtypes, regardless of the stage or grade of disease [81]. The Cancer Genome Atlas findings on OC showed that there are few recurrent mutated genes, with four transcript subtypes and the presence of extensive defects homologous recombination with mutual exclusivity [82]. Considering that BRCA 1/2 mutated OC are known to be associated with improved survival and that the presence of TILs (CD3+ and CD8+) and high CD8+/CD4+ ratio are associated with favorable OS in OC, a pilot study explored the differences in the presence of TILs and PD-1 and PD-L1 expression between BRCA mutated and non mutated. The results showed a significantly higher CD8+/CD4+ ratio in BRCA1/2-mutated tumors and peritumoral CD3+ T-cells significantly increased in BRCA1/2-mutated compared to non-BRCA1/2-mutated ovarian cancers. An elevated number of CD3+ and CD8+ in BRCA1/2-mutated OC may provide an additional explanation for the improved clinical outcomes associated with these mutations. In patients with platinum-resistant OC receiving nivolumab, a phase II trial on a very limited number of patients found that nivolumab at 1 mg/kg was well tolerated and has encouraging clinical efficacy [83].

In a phase Ib, open-label expansion trial in patients with previously treated, recurrent or refractory ovarian cancer, avelumab demonstrated an acceptable safety profile and to be clinically active [84]. Activity of avelumab was higher in patients with platinum sensitive recurrence compared to the resistant and in patients with less line of previous chemotherapy. The interim results from a phase Ib study with pembrolizumab in patients with PD-L1 positive advanced OC, has also demonstrated antitumor activity in heavily pretreated patients and a manageable safety and toxicity profile [85]. The first phase 2 and 3 trials are ready to start also in ovarian cancer addressing the role of immunotherapy in all treatment settings of the disease. In the next future it will be also important to identify the dominant immunosuppressive pathways in the different OC subtypes that will allow patients to be optimally matched with therapies. Also, combination strategies should be explored with standard and emerging therapies.

Conclusions

Based on the characteristics of the immune system to exhibit innate and adaptive responses to tumors and to maintain a memory of such responses, the “modern” immunotherapy with checkpoint inhibitors got remarkable results mainly in melanoma patients with the possibility to reach long-term benefit. This results were translated into different malignancies, and there are a lot of clinical studies still ongoing. However, important results include improvement in OS for NSCLC and RCC; these data have already well established in clinical practice. Moreover, promising results have been also obtained in head and neck, Hodgkin lymphoma, breast and ovarian cancer, neuroblastoma, and myeloma.

Data from ongoing clinical trials and further researches will provide more information about mechanisms of activation and resistance, as well as combination approaches. A critical issue will be also to provide biomarkers which could be predictive of response.

Abbreviations

ADCC:

Antibody-dependent cell- mediated cytotoxicity

AE:

Adverse event

ASCT:

Authologous stem cell transplantation

BRCA:

Breast related cancer antigens

CCL:

Chemokine ligand

CEA:

Carcino-embryonic antigen

CSF-R1:

Colony stimulating factor 1 receptor

CT:

Cancer testis

CTL:

Cytotoxic T lymphocyte

CTLA-4:

Cytotoxic T lymphocyte antigen 4

DC:

Dendritic cell

ELISA:

Enzyme-linked immunosorbent assay

ELISPOT:

Enzyme-linked immunospot

ER:

Estrogen receptor

FDA:

Food and Drug Administration

FOXO1:

Forkhead box protein O1

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

HCC:

Hepatocellular carcinoma

HER-2:

Human epidermal growth factor receptor 2

HLA:

Human leucocyte antigens

ICD:

Immunogenic cell death

IFA:

Incomplete Freund’s Adjuvant

IFNγ:

Interferon gamma

irPFS:

Immune-related progression-free survival

LAG-3:

Lymphocyte-activation gene 3

MAGE-A3:

Melanoma AntiGen Encoding-antigen 3

MDSC:

Myeloid-derived suppressor cell

mTOR:

Mammalian target of rapamycin

NK:

Natural killer

NKT:

Natural killer T cells

NSCLC:

Non-small-cell lung cancer

OC:

Ovarian cancer

ORR:

Overall response rate

OS:

Overall survival

PBMC:

Peripheral blood mononuclear cell

PCR:

Polymerase chain reaction

PD-1:

Programmed cell death protein 1

PD-L1:

Programmed death ligand 1

PFS:

Progression-free survival

RCC:

Renal cell cancer

RT:

Radiotherapy

sdRNA:

Seed region ribonucleic acid

TAA:

Tumor-associated antigen

TGF:

Transforming growth factor

TIL:

Tumor-infiltrating lymphocyte

TLR:

Toll-like receptor

TME:

Tumor microenvironment

TNBC:

Triple-negative breast cancer

T-reg:

Regulatory T cells

T-VEC:

Talimogene laherparepvec

VEGF:

Vascular endothelial growth factor

References

  1. Daniele B, Sangro B, Petrylak D, Calabrò F, Cartenì G, Montesarchio V, et al. The 2nd meeting of the Campania Society of Oncology Immunotherapy (SCITO): focus on hepatocellular carcinoma, kidney and bladder cancer. J ImmunoTher Cancer. 2016;4:1.

  2. Wolchok JD, Hoos A, O’Day S, Weber JS, Hamid O, Lebbe C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15(23):7412–20.

    Article  CAS  PubMed  Google Scholar 

  3. Lipson EJ, Sharfman WH, Drake CG, Wollner I, Taube JM, Anders RA, et al. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin Cancer Res. 2013;19(2):462–8.

    Article  CAS  PubMed  Google Scholar 

  4. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 2015;33(17):1889–94.

    Article  CAS  PubMed  Google Scholar 

  5. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, et al. Overall survival and long-term safety of nivolumab (Anti-Programmed Death 1 Antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol. 2015;33(18):2004–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Speiser DE, Lienard D, Rufer N, Rubio-Godoy V, Rimoldi D, Lejeune F, et al. Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909. J Clin Invest. 2005;115(3):739–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Speiser DE, Baumgaertner P, Voelter V, Devevre E, Barbey C, Rufer N, et al. Unmodified self antigen triggers human CD8 T cells with stronger tumor reactivity than altered antigen. Proc Natl Acad Sci U S A. 2008;105(10):3849–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. van Poelgeest MI, Welters MJ, van Esch EM, Stynenbosch LF, Kerpershoek G, van Persijn van Meerten EL, et al. HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy of patients with advanced or recurrent HPV16-induced gynecological carcinoma, a phase II trial. J Transl Med. 2013;11:88.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, 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.

    Article  CAS  PubMed  Google Scholar 

  11. Perret R, Sierro SR, Botelho NK, Corgnac S, Donda A, Romero P. Adjuvants that improve the ratio of antigen-specific effector to regulatory T cells enhance tumor immunity. Cancer Res. 2013;73(22):6597–608.

    Article  CAS  PubMed  Google Scholar 

  12. Corgnac S, Perret R, Zhang L, Mach JP, Romero P, Donda A. iNKT/CD1d-antitumor immunotherapy significantly increases the efficacy of therapeutic CpG/peptide-based cancer vaccine. J Immunother Cancer. 2014;2(1):39.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, et al. mTOR regulates memory CD8 T-cell differentiation. Nature. 2009;460(7251):108–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhang L, Tschumi BO, Lopez-Mejia IC, Oberle SG, Meyer M, Samson G, et al. Mammalian target of rapamycin complex 2 controls CD8 T cell memory differentiation in a Foxo1-dependent manner. Cell Rep. 2016;14(5):1206–17.

    Article  CAS  PubMed  Google Scholar 

  15. Scholz G, Jandus C, Zhang L, Grandclement C, Lopez-Mejia IC, Soneson C, et al. Modulation of mTOR signalling triggers the formation of stem cell-like memory T cells. EBioMedicine. 2016;4:50–61.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Weide B, Martens A, Zelba H, Stutz C, Derhovanessian E, Di Giacomo AM, et al. Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin Cancer Res. 2014;20(6):1601–9.

    Article  CAS  PubMed  Google Scholar 

  17. Weide B, Zelba H, Derhovanessian E, Pflugfelder A, Eigentler TK, Di Giacomo AM, et al. Functional T cells targeting NY-ESO-1 or Melan-A are predictive for survival of patients with distant melanoma metastasis. J Clin Oncol. 2012;30(15):1835–41.

    Article  CAS  PubMed  Google Scholar 

  18. Zelba H, Weide B, Martens A, Derhovanessian E, Bailur JK, Kyzirakos C, et al. Circulating CD4+ T cells that produce IL4 or IL17 when stimulated by melan-A but not by NY-ESO-1 have negative impacts on survival of patients with stage IV melanoma. Clin Cancer Res. 2014;20(16):4390–9.

    Article  CAS  PubMed  Google Scholar 

  19. Bailur JK, Gueckel B, Derhovanessian E, Pawelec G. Presence of circulating Her2-reactive CD8 + T-cells is associated with lower frequencies of myeloid-derived suppressor cells and regulatory T cells, and better survival in older breast cancer patients. Breast Cancer Res. 2015;17:34.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Poschke I, Lovgren T, Adamson L, Nystrom M, Andersson E, Hansson J, et al. A phase I clinical trial combining dendritic cell vaccination with adoptive T cell transfer in patients with stage IV melanoma. Cancer Immunol Immunother. 2014;63(10):1061–71.

    Article  CAS  PubMed  Google Scholar 

  21. Pico de Coana Y, Poschke I, Gentilcore G, Mao Y, Nystrom M, Hansson J, et al. Ipilimumab treatment results in an early decrease in the frequency of circulating granulocytic myeloid-derived suppressor cells as well as their Arginase1 production. Cancer Immunol Res. 2013;1(3):158–62.

    Article  CAS  PubMed  Google Scholar 

  22. R2: genomics analysis and visualization platform. Available from: http://hgserver1.amc.nl/cgi-bin/r2/main.cgi?&species=hs. Accessed 19 Jul 2016.

  23. Mao Y, Eissler N, Blanc KL, Johnsen JI, Kogner P, Kiessling R. Targeting suppressive myeloid cells potentiates checkpoint inhibitors to control spontaneous neuroblastoma. Clin Cancer Res. 2016;22(15):3849-59.

  24. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275–87.

    Article  CAS  PubMed  Google Scholar 

  26. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Taube JM, Young GD, McMiller TL, Chen S, Salas JT, Pritchard TS, et al. Differential expression of immune-regulatory genes associated with PD-L1 display in melanoma: implications for PD-1 pathway blockade. Clin Cancer Res. 2015;21(17):3969–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ascierto ML, McMiller TL, Berger AE, Danilova L, Anders RA, Netto GJ, et al. Transcriptional signatures associated with lack of response to anti-PD-1 therapy in patients with renal cell carcinoma. Cancer Immunol Res. 2016;4(9):726-33.

  29. Linehan WM, Srinivasan R, Schmidt LS. The genetic basis of kidney cancer: a metabolic disease. Nat Rev Urol. 2010;7(5):277–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hato T, Goyal L, Greten TF, Duda DG, Zhu AX. Immune checkpoint blockade in hepatocellular carcinoma: current progress and future directions. Hepatology. 2014;60(5):1776–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xu P, Chen YJ, Chen H, Zhu XY, Song HF, Cao LJ, et al. The expression of programmed death-1 in circulating CD4+ and CD8+ T cells during hepatitis B virus infection progression and its correlation with clinical baseline characteristics. Gut Liver. 2014;8(2):186–95.

    Article  CAS  PubMed  Google Scholar 

  32. Barathan M, Gopal K, Mohamed R, Ellegard R, Saeidi A, Vadivelu J, et al. Chronic hepatitis C virus infection triggers spontaneous differential expression of biosignatures associated with T cell exhaustion and apoptosis signaling in peripheral blood mononucleocytes. Apoptosis. 2015;20(4):466–80.

    Article  CAS  PubMed  Google Scholar 

  33. Zeng Z, Shi F, Zhou L, Zhang MN, Chen Y, Chang XJ, et al. Upregulation of circulating PD-L1/PD-1 is associated with poor post-cryoablation prognosis in patients with HBV-related hepatocellular carcinoma. PLoS One. 2011;6(9):e23621.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sangro B, Gomez-Martin C, de la Mata M, Inarrairaegui M, Garralda E, Barrera P, et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol. 2013;59(1):81–8.

    Article  CAS  PubMed  Google Scholar 

  35. El-Khoueiry AB, Melero I, Crocenzi SB, Welling TH, Yau TC, Yeo W. Phase I/II safety and antitumor activity of nivolumab in patients with advanced hepatocellular carcinoma (HCC): CA209-040. J Clin Oncol. 2015;33(Suppl):Abstr LBA101.

    Google Scholar 

  36. Melero I, Murillo O, Dubrot J, Hervas-Stubbs S, Perez-Gracia JL. Multi-layered action mechanisms of CD137 (4-1BB)-targeted immunotherapies. Trends Pharmacol Sci. 2008;29(8):383–90.

    Article  CAS  PubMed  Google Scholar 

  37. Melero I, Martinez-Forero I, Dubrot J, Suarez N, Palazon A, Chen L. Palettes of vaccines and immunostimulatory monoclonal antibodies for combination. Clin Cancer Res. 2009;15(5):1507–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Weigelin B, Bolanos E, Rodriguez-Ruiz ME, Martinez-Forero I, Friedl P, Melero I. Anti-CD137 monoclonal antibodies and adoptive T cell therapy: a perfect marriage? Cancer Immunol Immunother. 2016;65(5):493–7.

    Article  CAS  PubMed  Google Scholar 

  39. Weigelin B, Bolanos E, Teijeira A, Martinez-Forero I, Labiano S, Azpilikueta A, et al. Focusing and sustaining the antitumor CTL effector killer response by agonist anti-CD137 mAb. Proc Natl Acad Sci U S A. 2015;112(24):7551–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jungbluth AA, Ely S, DiLiberto M, Niesvizky R, Williamson B, Frosina D, et al. The cancer-testis antigens CT7 (MAGE-C1) and MAGE-A3/6 are commonly expressed in multiple myeloma and correlate with plasma-cell proliferation. Blood. 2005;106(1):167–74.

    Article  CAS  PubMed  Google Scholar 

  41. van Rhee F, Szmania SM, Zhan F, Gupta SK, Pomtree M, Lin P, et al. NY-ESO-1 is highly expressed in poor-prognosis multiple myeloma and induces spontaneous humoral and cellular immune responses. Blood. 2005;105(10):3939–44.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Atanackovic D, Arfsten J, Cao Y, Gnjatic S, Schnieders F, Bartels K, et al. Cancer-testis antigens are commonly expressed in multiple myeloma and induce systemic immunity following allogeneic stem cell transplantation. Blood. 2007;109(3):1103–12.

    Article  CAS  PubMed  Google Scholar 

  43. Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36(2):133–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Baitsch L, Fuertes-Marraco SA, Legat A, Meyer C, Speiser DE. The three main stumbling blocks for anticancer T cells. Trends Immunol. 2012;33(7):364–72.

    Article  CAS  PubMed  Google Scholar 

  45. Baitsch L, Baumgaertner P, Devevre E, Raghav SK, Legat A, Barba L, et al. Exhaustion of tumor-specific CD8(+) T cells in metastases from melanoma patients. J Clin Invest. 2011;121(6):2350–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–9.

    Article  CAS  PubMed  Google Scholar 

  47. Verdeil G, Fuertes Marraco SA, Murray T, Speiser DE. From T cell “exhaustion” to anti-cancer immunity. Biochim Biophys Acta. 2016;1865(1):49–57.

    CAS  PubMed  Google Scholar 

  48. Giordano M, Henin C, Maurizio J, Imbratta C, Bourdely P, Buferne M, et al. Molecular profiling of CD8 T cells in autochthonous melanoma identifies Maf as driver of exhaustion. EMBO J. 2015;34(15):2042–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Speiser DE, Utzschneider DT, Oberle SG, Munz C, Romero P, Zehn D. T cell differentiation in chronic infection and cancer: functional adaptation or exhaustion? Nat Rev Immunol. 2014;14(11):768–74.

    Article  CAS  PubMed  Google Scholar 

  50. Mahnke YD, Devevre E, Baumgaertner P, Matter M, Rufer N, Romero P, et al. Human melanoma-specific CD8(+) T-cells from metastases are capable of antigen-specific degranulation and cytolysis directly ex vivo. Oncoimmunology. 2012;1(4):467–530.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Romano E, Kusio-Kobialka M, Foukas PG, Baumgaertner P, Meyer C, Ballabeni P, et al. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112(19):6140–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph-Pietras D, et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med. 2014;6(254):254ra128.

    Article  PubMed  Google Scholar 

  53. Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology. 2014;3:e28518.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004;58(3):862–70.

    Article  PubMed  Google Scholar 

  55. Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol. 2015;16(7):795–803.

    Article  CAS  PubMed  Google Scholar 

  56. Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005;11(2 Pt 1):728–34.

    CAS  PubMed  Google Scholar 

  57. Golden EB, Demaria S, Schiff PB, Chachoua A, Formenti SC. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol Res. 2013;1(6):365–72.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst. 2013;105(4):256–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Vanpouille-Box C, Diamond JM, Pilones KA, Zavadil J, Babb JS, Formenti SC, et al. TGFbeta is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res. 2015;75(11):2232–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Lynch TJ, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Oncol. 2012;30(17):2046–54.

    Article  CAS  PubMed  Google Scholar 

  61. Rizvi NA, Mazieres J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16(3):257–65.

    Article  CAS  PubMed  Google Scholar 

  62. Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373(2):123–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39.

    Article  CAS  PubMed  Google Scholar 

  64. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.

    Article  PubMed  Google Scholar 

  65. Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–50.

    Article  CAS  PubMed  Google Scholar 

  66. Mok T, Wu YL, Sadowski S, Zhang J, Rangwala R, Kush D, et al. 195TiP: Pembrolizumab (MK-3475) versus platinum-based chemotherapy for PD-L1+ NSCLC in a phase 3, randomized, open-label study: KEYNOTE-042. J Thorac Oncol. 2016;11(4 Suppl):S142.

    Article  CAS  PubMed  Google Scholar 

  67. Goldberg SB, Gettinger SN, Mahajan A, Herbst R, Chiang A, Tsiouris AJ, editors. A phase II trial of pembrolizumab for untreated brain metastases from non-small cell lung cancer. 16th ed. Denver: WCLC; 2015.

    Google Scholar 

  68. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803–13.

    Article  CAS  PubMed  Google Scholar 

  69. George S, Motzer RJ, Hammers HJ, Redman BG, Kuzel TM, Tykodi SS, et al. Safety and efficacy of nivolumab in patients with metastatic renal cell carcinoma treated beyond progression: a subgroup analysis of a randomized clinical trial. JAMA Oncol. 2016;2(9):1179-86.

  70. Hammers HJ, Plimack ER, Infante JR, Ernstoff MS, Rini BI, McDermott DF. Phase I study of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma (mRCC). J Clin Oncol. 2014;32:5s (suppl; abstr 4504).

    Article  Google Scholar 

  71. Amin A, Plimack ER, Infante JR, Ernstoff MS, Rini BI, McDermott DF. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) in combination with sunitinib or pazopanib in patients (pts) with metastatic renal cell carcinoma (mRCC). J Clin Oncol. 2014;32:5s (suppl; abstr 5010).

    Article  Google Scholar 

  72. Lieu C, Bendell J, Powderly JD, Pishvaian MJ, Hochster H, Eckhardt SG, et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with bevacizumab and/or chemotherapy in patients with locally advanced or metastatic solid tumours. Ann Oncol. 2014;25 Suppl 4:Iv361–72.

    Google Scholar 

  73. Loi S, Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol. 2014;25(8):1544–50.

    Article  CAS  PubMed  Google Scholar 

  74. Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res. 2014;2(4):361–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121(7):2750–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Wang Y, Waters J, Leung ML, Unruh A, Roh W, Shi X, et al. Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature. 2014;512(7513):155–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol. 2016;34(21):2460-7.

  78. Emens LA, Braiteh FS, Cassier P, Delord JP, Eder JP, Fasso M, et al., editors. Inhibition of PD-L1 by MPDL3280A leads to clinical activity in patients with metastatic triple-negative breast cancer (TNBC). Philadelphia: American Association for Cancer Research Annual Meeting; 2015.

    Google Scholar 

  79. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348(3):203–13.

    Article  CAS  PubMed  Google Scholar 

  80. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102(51):18538–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol. 2012;124(2):192–8.

    Article  PubMed  Google Scholar 

  82. Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474(7353):609–15.

    Article  Google Scholar 

  83. Hamanishi J, Mandai M, Ikeda T, Minami M, Kawaguchi A, Matsumura N, et al. Durable tumor remission in patients with platinum-resistant ovarian cancer receiving nivolumab. J Clin Oncol. 2015;33(Suppl):Abstr 5570.

    Google Scholar 

  84. Disis ML, Patel MR, Pant S, Infante JR, Lockhart AC, Kelly K, et al. Avelumab (MSB0010718C), an anti-PD-L1 antibody, in patients with previously treated, recurrent or refractory ovarian cancer: a phase Ib, open-label expansion trial. J Clin Oncol. 2015;33(Suppl):Abstr 5509.

    Google Scholar 

  85. Varga AA, Ott PA, Mehnert JM, Berton-Rigaud D, Johnson EA, et al. Antitumor activity and safety of pembrolizumab in patients (pts) with PD-L1 positive advanced ovarian cancer: Interim results from a phase Ib study. J Clin Oncol. 2015;33(Suppl):5510.

    Google Scholar 

Download references

Acknowledgement

The meeting was supported by Fondazione Melanoma Onlus and the Society of ImmunoTherapy of Cancer (SITC). A special thanks to 3P Solution of Napoli for their support and cooperation in organizing the meeting and especially to Lucia Politi.

Funding

Not applicable.

Availability of data and materials

Not applicable.

Authors’ contributions

PAA prepared the manuscript collaboratively with input of MLA, SF, SG, HH, VH, RK, IM, RN, GP, SP, PR, DS, BAF, FMM. All authors read and approved the final manuscript.

Competing interests

PAA has/had consultant and advisory role for Bristol Myers Squibb, Merck Sharp & Dohme, Roche-Genenetech, Novartis, Ventana Medical Systems, Inc, Amgen, Array, and . He received research fund from Bristol Myers Squibb, Roche-Genetech, Ventana, and Array.

MLA has no competing interest.

SF has no competing interest.

RK is in the Scientific Advisory Board for the companies Immunicum AB, Idogen Inc, Glactone Pharma AB and CLS AB, and has received compensation for arranging courses for Bristol Myers Squibb and a research grant from Moderna Therapeutics.

SG has/had consultant and advisory role for Third Rock Ventures, Boehringer-Ingelheim, B4CC, and OncoMed. He received research funds from Janssen R&D and Immune Design, unrelated to the topic presented.

VH she participated to advisory boards of BI, AZ, Roche, Pfizer, Amgen, Merck, BMS.

HH participated to advisory board for BMS and Exelixis. He received also clinical trial support from BMS.

IM Advisor: BMS, Roche, AstraZeneca, Boehringer Ingelheim. Grants: BMS, Pfizer.

RN is consultant for Genentech, Novartis, Puma Biotechnology. She received also research funding from Celgene, Corcept Therapeutics, Merck.

GP reports receiving honoraria from Pfizer, Cellgene, Clasado and Astellas.

SP received honoraria from Roche, AstraZeneca, Pfizer, Pharmamar.

PR He is a member of the Scientific Advisory Board of Immatics biotechnologies, Transgene SA and Neximmune and receives funding from Roche Pharma Research and Early Development (2016).

DS has no competing interest.

BAF has/had advisory role for PerkinElmer. He received also research support from Definiens, PerkinElmer, Ventana/Roche, BMS, Viralytics.

FMM has no competing interest.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Paolo A. Ascierto.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ascierto, P.A., Ascierto, M.L., Formenti, S. et al. Perspectives in immunotherapy: meeting report from the “Immunotherapy Bridge”, Napoli, December 5th 2015. j. immunotherapy cancer 4, 62 (2016). https://doi.org/10.1186/s40425-016-0168-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40425-016-0168-3

Keywords