Open access peer-reviewed chapter

Cancer Vaccines

Written By

Carmen Murias Henriquez, Hendrik-Tobias Arkenau, Valérie Dutoit and Anna Patrikidou

Submitted: 12 March 2019 Reviewed: 08 August 2019 Published: 13 November 2019

DOI: 10.5772/intechopen.89074

From the Edited Volume

Cancer Immunotherapy and Biological Cancer Treatments

Edited by Hilal Arnouk

Chapter metrics overview

968 Chapter Downloads

View Full Metrics

Abstract

Recent advances in immuno-oncology have allowed for the design of more specific and efficient cancer vaccine approaches. There has been an improvement in molecular biology techniques, as well as a greater understanding of the mechanisms involved in the activation and regulation of T cells and the interplay between the components of the immune system and the escape mechanisms used by cancer cells and the tumour microenvironment. As a result, many interesting developments in therapeutic cancer vaccines are ongoing, with influence on survival still to be proven. The spectrum of tumour antigens that are recognised by T cells is still largely unchartered and, most importantly, dynamically evolving over time, driven by clonal evolution and treatment-driven selection. Vaccine approaches currently in development and tested in clinical studies are based on tumour antigens specifically identified for each tumour type, on tumour cells or dendritic cells, the latter having the potential to be modified to incorporate immunostimulatory genes. However, interplay between the immune system and the tumour and the inhibitory mechanisms developed by tumour cells to subvert immune responses are crucial issues that will need to be targeted in order for efficient therapeutic vaccines to emerge.

Keywords

  • vaccine
  • T cells
  • tumour antigens
  • immune system

1. Introduction

Cancer constitutes one of the biggest burdens in the Western society with lung, breast, prostate and colorectal cancer being the most prevalent. Despite declining rates in the Western societies [1], there have been an estimated 9.6 million deaths by cancer worldwide in 2018 [2, 3], and it is expected that this number will further increase over time.

The molecular nature of human cancers is complex and varies among tumours and individuals. For that reason, the approach towards a more personalised cancer treatment has gained intense interest. Treatment approaches are being increasingly changing from histology- to molecular-based therapies, including targeting the interplay between cancer and the immune system.

In the last few decades, major advances have been made in recognising that an effective immune system—or the lack thereof—plays an important role in cancer development, growth and metastasis. For example, the presence of tumour-infiltrating lymphocytes (TILs) has been identified as a positive prognostic factor in multiple cancer types [4]. Utilising the body’s defences and reactivating the antitumour immune response, initially regarded as a simple paradigm, have created a scientific and therapeutic revolution [5]. After decades of attempts, immunotherapy has achieved a major breakthrough with the sensational successes of immunomodulation with checkpoint blockade (i.e. PD-1/PD-L1 and CTLA-4 inhibitors, among others). In addition to that, the recent development of immune effector cell therapy in the form of chimeric antigen receptor T cells for haematological malignancies has opened exciting horizons for solid tumours as well [6, 7]. Up to now, therapeutic vaccination against cancer, despite few examples like Bacillus Calmette-Guérin (BCG) treatment for superficial bladder cancer [8], the oncolytic virus-based talimogene laherparepvec (T-VEC) [9] and the Sipuleucel vaccine in prostate cancer [10], has not achieved similar results.

Nevertheless, new vaccine development techniques as well as immunotherapy combination strategies shape the pipeline of current trial development and represent a promise and challenge for the future.

1.1 The T-cell response to cancer

The immune system and in particular dendritic cells (DCs) and macrophages are capable, to a variable extent, to recognise damage-associated molecular patterns (DAMPs), thus eliciting an innate immune response. The major DAMP driving innate host antitumour immune responses is tumour-derived DNA, which is detected via the stimulator of interferon gene (STING) pathway and results in type I IFN production [11].

The adaptive immune response begins when cancer antigens are presented to T and B cells by DCs. Although B-cell responses are probably playing a role in antitumour immunity, not much is yet known about them [12]. The following text will therefore mostly refer to antitumour T-cell responses. Tumour antigens are transported via lymphatic vessels to the lymph nodes, where they are captured by lymph node-resident DCs. Alternatively, tissue-resident DCs capture antigens at the tumour site and migrate to induce T-cell responses in the lymph node [13]. DCs present protein antigens in the context of major histocompatibility complex (MHC) class I and II molecules, allowing the stimulation of rare antigen-specific CD8+ or CD4+ T lymphocytes, respectively. Upon antigen encounter, CD8 T cells differentiate into cytotoxic T lymphocytes (CTLs) that have tumour-killing capacities, whereas CD4 T cells will provide CD8 T-cell help [13]. CD4 T cells can also be induced to become FoxP3+ regulatory T cells (Tregs), which are then able to inhibit antitumoural immune responses [14].

A tumour mass is not composed solely of tumour cells, but contains immune cells, stromal cells and vessels, a concept known as the tumour microenvironment. Tumours are organised in various reciprocal, local and systemic relations with myeloid and lymphoid immune cell populations, both being key factors in regulating immune responses to cancer. During progression, tumours are able to modulate the immune response and highjack it to their advantage, in order to invade and grow. Macrophages can be polarised to a pro-tumoural and anti-inflammatory (called M2) phenotype at the tumour’s advantage. In addition, myeloid-derived suppressor cells (MDCSs) accumulate in the tumour microenvironment and are able to suppress antitumour T-cell responses [15, 16].

1.2 The three phases of tumour immunoediting: elimination, equilibrium and escape

The principles of cancer immunoediting have set the basis for understanding the dual host-protective and immuno-sculpting effects of immunity on cancer [17]. During cancer immunoediting, the host immune system influences tumour fate in three phases through activation of innate and adaptive immune mechanisms: elimination, equilibrium and escape.

1.2.1 Elimination

The elimination phase occurs when cancer cells are eradicated by a competent immune system. This is evidenced by immunodeficient mice that have an increased propensity to develop carcinogen-induced and spontaneous cancers than wild-type mice [18]. In addition, tumours that come from immunodeficient mice are more immunogenic than those from immunocompetent mice, as they have not been edited by the immune response. Patients suffering from AIDS [19] or being under immunosuppression are similarly more prone to develop cancer [20, 21]. The role of CD8 T cells has been more extensively studied; however interplay with CD4 T-cell responses is also required in order to have an integrated and efficient response [22, 23].

1.2.2 Equilibrium

The sporadic tumour cells that survive immune destruction will enter into the equilibrium phase where editing arises. Immune pressure is mostly mediated by CD4 and CD8 T cells [24]. Upon tumour editing, more mutations will be acquired, which will favour entry into the escape phase of immunoediting. Importantly, the process of incomplete elimination promotes the generation of tumour cell variants with decreased immunogenicity [23]. The identification of hidden cancer cells in an equilibrium state remains a challenge; however, advances in technology and biomarkers may allow for circulating tumour cells and niches to be investigated further.

1.2.3 Escape

The escape phase represents the final phase of the process, where immunologically sculpted tumours begin to grow progressively, becoming clinically apparent. Tumour escape can result from many different mechanisms including reduced immune recognition, through loss of MHC class I, co-stimulatory molecules or tumour antigens. In addition, the tumour induces many molecules and cells to induce an immunosuppressive tumour microenvironment. Cytokines such as VEGF and TGF-β, immunoregulatory molecules such as indoleamine 2,3-dioxygenase (IDO), programmed death-ligand 1 (PD-L1) and ligands for Tim3 and lymphocyte-activation gene 3 (LAG-3), among others, are induced to supress the incoming CD4 and CD8 T cells. In addition, many cellular components of the tumour microenvironment, such as macrophages and neutrophils, are being redirected in an anti-inflammatory pro-tumoural state [25, 26, 27].

1.3 The principles and means of immunotherapy

Although cancer cells have the unique ability to escape from the immune response, the knowledge that immune cells are able to recognise tumours allows development of therapies that utilise the immune system [28]. Cancer immunotherapies focus on exploiting both the innate and adaptive arms of the immune system. They can be classified into vaccines, monoclonal antibodies (including immune checkpoint inhibitors), recombinant cytokines, small molecules and adoptive T-cell transfer, including chimeric antigen receptor (CAR), TCR and TIL therapy [29, 30, 31, 32].

1.3.1 Vaccines

The aim of cancer vaccination is to prime cellular immune response against tumour-specific antigens. Despite its limitations mainly owing to heterogeneous tumour antigen composition and expression and their susceptibility to various mechanisms of immune suppression, it is being intensively developed. Currently revisited with strategies aiming at combinations with other immunotherapies, cancer vaccines will be addressed in detail in this chapter.

1.3.2 Monoclonal antibodies

Antibodies target (a) factors that regulate signal pathways used by cancer cells in division and angiogenesis (such as the VEGF inhibitor bevacizumab) [33]; (b) tumour-associated antigens, activating antibody-dependent cellular cytotoxicity (such as the Her2-directed antibody trastuzumab) [34]; (c) complement-dependent cytotoxicity (such as the anti-20 and anti-EGFR antibodies rituximab and cetuximab); and (d) immune blockade with checkpoint inhibitors such as anti-CTLA4 antibodies (ipilimumab and tremelimumab), anti-PD1 antibodies (nivolumab and pembrolizumab) or anti-PD-L1 antibodies (atezolizumab, durvalumab, and avelumab) [31].

1.3.3 Recombinant cytokines

Immunostimulatory recombinant cytokines promote lymphocyte activation via control of transcriptional and metabolic programmes [35]. An example is recombinant IL-2 (aldesleukin, Proleukin®) that has been used to treat renal cancer and melanoma [36]. Another recombinant cytokine approved by the US Food and Drug Administration (FDA) for the adjuvant treatment in resected melanoma patients is pegylated interferon α-2β (Sylatron®), a member of the IFN cytokine family [37]. Concurrent administration of immunostimulatory cytokines such as IL-2 and granulocyte-macrophage colony-stimulating factor (GM-CSF) may also enhance the efficacy of antibody therapy [38]. Limitations include their antigenicity, poor pharmacokinetics and high toxicity [29].

1.3.4 Small molecules

The use of small molecules in cancer immunotherapy has been increasing, given their ability to target both intracellular and surface targets. Plerixafor is a small molecule that inhibits the binding interaction of stromal cell-derived factor 1 (SDF-1) to the chemokine receptor CXCR4, used as a haematopoietic stem cell mobiliser [39]. This small molecule aims to prevent the development of cancer metastasis in cancer patients, principally in pancreatic ductal adenocarcinoma patients [29]. Another known small molecule called imiquimod, used for the treatment of basal cell carcinoma, is an agonist for toll-like receptor (TLR)-7. Imiquimod-mediated TLR7 activation induces production of proinflammatory cytokines, inhibits Tregs and induces activation of natural killer (NK) cells to eliminate cancer cells [29]. IDO inhibitors are being tested as well in multiple malignancies, but results as monotherapy have been disappointing [40]. Combinations with other immunotherapeutic agents or with chemotherapy/radiation are being currently investigated. A much anticipated combination, however, of the small molecule IDO1 inhibitor with pembrolizumab failed to provide significant benefit in a phase 3 trial in unresectable or metastatic melanoma [41]. Finally, ongoing research evaluates the adenosine signalling with adenosine receptor inhibitors [42].

1.3.5 Adoptive T-cell therapy

The use of cancer patient’s own immune effector cells is a novel cancer immunotherapy, also called adoptive cell therapy. Starting with TILs, it has moved to the generation of artificial T cells that are genetically altered to express an antitumour antibody (CARs) or a selected TCR [30]. These cells are multiplied and subsequently transferred back to the patient, who usually receives conditioning chemotherapy. TIL therapy has shown some clinical evidence of efficacy in the treatment of melanoma [43] and cervical cancer [44], with the LN-145 TIL therapy recently obtaining breakthrough therapy designation by FDA, while its potential is being further investigated. Generation of tumour-specific T cells through expression of a TCR that has shown antitumour properties is ongoing for several malignancies [44]. CAR T cells, which are engineered to express part of a tumour-specific antibody, linked to intracellular T-cell signalling domains are gaining major interest [45]. Two anti-CD19 CAR T-cell therapies have so far received FDA approval in haematological malignancies, notably tisagenlecleucel for diffuse large B-cell lymphoma (DLBCL) and acute lymphoblastic leukaemia and axicabtagene ciloleucel for primary or transformed DLBCL, mediastinal and high-grade B-cell lymphoma [46]. Research on CAR T-cell therapies in solid malignancies is currently ongoing.

Advertisement

2. Cancer vaccines

2.1 Introduction

Cancer vaccination seeks to generate, amplify or skew (or the combination thereof) antitumour immunity. In order to reach such an ambitious goal, many approaches are in development, including the administration of tumour antigens, often with antigen presenting cells or other immune modulators.

Current technological advances in genomics, data science and cancer immunotherapy enable the fast mapping of alterations within a genome, as well as the rational selection of vaccine targets and on-demand production of a therapy that has been customised to a patient’s individual tumour. With the development of vaccination being promoted by emerging innovations in the digital era, vaccinating patients according to their individual tumour mutational profile may become the first truly personalised treatment for cancer.

It is important to distinguish vaccines that are designed to prevent cancer from the ones that are designed to treat cancer. The mode of action of the HPV vaccine for the prevention of cervical and other HPV-associated cancers [9] and of hepatitis B virus (HBV) vaccine for the prevention of HBV infection that carries a risk of development of hepatocellular carcinoma [47] is the prevention of infection itself. Their action is based on the generation of antiviral antibodies and has led to a net reduction in the incidence of these cancers in vaccinated individuals [48]. The development of therapeutic cancer vaccines has been more challenging. The nature of the antigen, which, in the case of therapeutic cancer vaccines, is derived from self-antigens against which the host has been tolerised and the presence of a hostile tumour microenvironment are key limiting factors.

2.2 Therapeutic vaccines

2.2.1 FDA-approved vaccines

Three therapeutic cancer vaccines have been approved by the FDA. The Bacillus Calmette-Guérin (BCG, TheraCys®, TICE®) vaccine, based on a live attenuated strain of Mycobacterium bovis, was the first approved cancer vaccine for use in non-muscle invasive bladder carcinoma following transurethral resection. It showed a prolongation in disease-free survival (DFS) of 30 months in patients with bladder carcinoma in situ (CIS) and of 22.5 months in patients with Ta/T1 urothelial carcinoma compared to 4.9 months in bladder CIS and 10.5 months in Ta/T1 patients treated with topical doxorubicin [49].

Sipuleucel-T (Provenge) is an autologous DC vaccine for patients with minimally symptomatic or asymptomatic metastatic castrate-resistant prostate cancer (mCRPC). Patient’s DCs are being injected with a recombinant fusion protein, PA2024, which consists of a tumour antigen, the prostate acid phosphatase (PAP) and GM-CSF, before reinfusion. The phase 3 IMPACT study, a double-blind, placebo-controlled, phase 3 trial of 512 mCRPC patients randomised to receive either three infusions of Sipuleucel-T or placebo 2 weeks apart, demonstrated statistically significant improvement of 4.1 months in median overall survival (OS) (25.8 months in the Sipuleucel-T group compared to 21.7 months in the placebo group) [10]. However, this study elicited significant criticism in regard with the observed—albeit modest—OS benefit without correlation with a progression-free survival (PFS) benefit or a T-cell response, the lack of association between survival benefit and T-cell proliferation responses, the fact that T-cell proliferative responses to the chimeric antigen (PA2024) did not cross-react to the physiological human PAP and hence the absence of alternative mechanisms to explain the survival benefit [50].

The third approved vaccine, called talimogene laherparepvec (T-VEC or Imlygic), is an oncolytic herpes virus 1-based vaccine for advanced melanoma. In this vaccine, two viral genes governing neurovirulence and blockade of antigen presentation are deleted, and the virus is modified to produce GM-CSF to enhance immunogenicity [8]. T-VEC was approved based on data published on the phase 3 OPTiM trial. The vaccine virus, injected intralesionally, infects both the cancer and normal cells but can only replicate within cancer cells. The OPTiM trial showed more durable response rate (>6 months) with T-VEC than GM-CSF alone, as well as higher overall response rate and a longer median OS (23.3 months compared to 18.9 months with GM-CSF alone) in patients with stage IIIB, IIIC or IV M1a melanoma [51].

2.2.2 Mode of action of therapeutic vaccines

The mode of action of most therapeutic vaccines involves development of cell-mediated immunity directed against tumour antigens; such antigens ought to ideally not be expressed in normal cells or have restricted normal expression, be of high expression on cancer cells, be highly immunogenic and be necessary for cancer cell survival [32]. Tumour antigens can be delivered as peptides, proteins, DNA or viral vectors or tumour cells themselves. They are usually administered with an adjuvant (see Section 2.5), in order to potentiate the immune response. Tumour antigens can also be generated via antigen spreading, which is the exposure of novel antigens after an initial antitumour response [52].

2.3 Tumour antigens

2.3.1 Tumour-associated antigens (TAAs)

TAAs are self-antigens commonly expressed in a specific tumour type among different patients. They are derived from non-mutated proteins that are overexpressed in tumour cells as compared to normal cells. The first TAAs were discovered after the cloning of gene-encoding proteins that generated epitopes recognised by tumour reactive TILs [53]. The first gene discovered that was reported to encode a tumour antigen recognised by T cells was MAGE-1 [53]. Since the discovery of MAGE-1, a large number of TAAs have been described, and they are classified into shared TAAs and unique TAAs [54], the latter being present only in individual patients.

Shared TAAs can be classified in three main groups, cancer/testis antigens, overexpressed antigens and differentiation antigens [54].

Cancer testis antigens (CT) are a large family of TAAs expressed in human tumours of different histological origins but not in normal adult tissues, with the exception of immune-privileged cells such as testis and placenta [55]. These antigens result from the reactivation of genes that are normally silent in adult tissues but that are transcriptionally activated in tumours. This quasi-exclusive tumour-restricted expression pattern (sparing normal germ cells that do not express HLA class I molecules) as well as their high prevalence make them ideal vaccine candidates [55]. They have been identified and tested in many human clinical trials [56]; however, there is usually very little knowledge about their specific function, especially with regard to tumour transformation. CT antigens include, among others, the MAGE-A, MAGE-B, MAGE-C, NY-ESO and SSX-2 families.

Overexpressed antigens are expressed at a higher level in tumour cells than in normal tissues. Expression of these antigens at variable levels in normal cells conveys the risk of autoimmune attack upon vaccination, but a large number of clinical trials have used these antigens with up to now few side effects [57]. Some examples of this group of antigens are tumour suppressor proteins such as p53 and the antiapoptotic proteins hTERT and Mucin 1 (MUC-1).

Tissue-specific (cell lineage) differentiation antigens are shared between the tumour and the normal tissue of origin, albeit with variable specificity. They include carcinoembryonic antigen (CEA), prostate-specific antigen (PSA), HER2/neu and melanoma lineage antigens such as gp-100, Melan-A/Mart-1 and tyrosinase, expressed in melanoma [58]. As for overexpressed antigens, they are endowed with a risk for autoimmune reactions.

The advantage of TAAs is that they are frequently expressed by the majority of patients and can therefore be used to treat many patients. The disadvantage of TAAs is the fact that some of them retain a level of expression in normal tissues, entailing the potential risk of autoimmune damage upon efficient vaccination. In addition, as TAAs derive from self-antigens, specific T cells have undergone negative selection, leaving only T cells with low avidity of antigen recognition, which are not able to generate strong immune responses.

2.3.2 Tumour-specific antigens (TSAs)

TSAs are antigens resulting from point mutations. They represent neoantigens mostly expressed by individual tumours. TSAs are tumour-specific, and it is usually viewed that their immunogenicity is not restricted by central tolerance, which is true when the mutated epitope is different enough from the wild-type one. Additionally, induced T-cell responses are not expected to result in autoimmune toxicity [59]. Moreover, neoantigens may be more resistant to immune selection, as they are critical for the oncogenic process and, therefore, essential for keeping the neoplastic state. In contrast, the fact that TSAs are patient-specific prevents broad vaccination and requires identification in a patient-specific manner. However, recent availability of sequencing technologies and epitope prediction algorithms allows for a rapid identification of potential neoantigens. Methods of in silico prediction of neo-epitope candidates with a high potential for neoantigen generation potentially present in multiple patients. These neoantigens hold the potential for development of “off the shelf” T-cell therapies, aiming to complement individualised, patient- and tumour-specific precision medicine approaches [32]. Nevertheless, it should be kept in mind that only a small percentage of mutations are being presented on MHC molecules at the tumour cell surface, making verification of the presence of a neo-epitope at the tumour cell surface a prerequisite [60, 61].

2.4 Types of vaccines

2.4.1 Peptide vaccines

Peptide vaccines consist in the delivery of MHC class I- or class II-restricted peptide epitopes derived from tumour antigens with the intent of activating CD8+ and CD4+ T cells. As peptides are not immunogenic per se, they need to be injected with an adjuvant [62]. GM-CSF, Montanide and TLR agonists, among others, have shown clinical benefit in small- and larger-scale clinical trials [63, 64, 65]. Peptide vaccines have the limitation of being applicable only to patients that have the HLA allele the peptide is restricted to. In addition, most vaccines are made of MHC class I-restricted peptides, therefore not eliciting CD4 T-cell help [66, 67]. In order to overcome this issue, the addition of non-tumour-specific peptides has been used, but limited data is available on the improvement provided by such heterologous helper peptides [68]. Overall, the numerous clinical trials performed in different tumour types have not provided satisfactory results yet [69].

Using multiple peptides derived from different TAAs targeting several antigens at once could overcome such tumour escape mechanisms. This multipeptide approach has demonstrated in in vitro and in vivo studies that multiple peptides do not compete for MHC presentation, inducing a multi-specific T-cell response [70, 71, 72]. The use of synthetic peptides with improved DC-targeting mechanisms, such as integrating pattern recognition receptors or TLRs [73], the conjugation of synthetic peptides to a DC-targeting antibody [74] or the encapsulation of long peptides in structures such as nanoparticles, liposomes or nano-hydrogel systems to enhance T-cell priming by DCs [75, 76] are some of the strategies under investigation towards a more efficient processing and presentation pathway that would lead to greater T-cell activation.

In two trials (a phase 1 and a randomised phase 2) combining single-dose pre-vaccine cyclophosphamide with IMA901, a renal cell carcinoma (RCC) peptide vaccine containing 10 antigens (9 HLA class I-binding and 1 HLA class II-binding) adjuvanted with GM-CSF in HLA-A02+ subjects showed that there was an improvement in survival with amplification of antigenic response and a reduction in suppressive circulatory T cells and MDSCs, with a disease control rate (DCR) at 6 months of 31% (95% CI: 3–35%) [77]. Mouse models also support combinations of multipeptide vaccines and chemotherapy. However, the addition of IMA901 to first-line sunitinib (an anti-angiogenic tyrosine kinase inhibitor) failed to show improvement in metastatic renal cancer, owing to low-level immune responses [78], despite the fact that sunitinib has been shown to decrease the number of Tregs in mice and patients with RCC [79, 80], as well as MDSCs in patients with RCC [81].

Owing to their tumour specificity, much effort has been made in order to exploit neoantigens for vaccine development. Such neoantigen-directed vaccines have been developed for melanoma, using either synthetic RNAs containing up to 10 predicted neoantigens or long peptides targeting up to 20 neoantigens [82, 83]. In these trials, neo-epitopes were chosen to bind HLA class I [82] or HLA class I and II molecules [83] and showed activation of CD4+ and CD8+ T cells in response to vaccination.

Peptide vaccines can also be helpful in the prevention of the progression of a premalignant lesion to cancer. The MUC-1 peptide vaccine has been tested as a prevention of progression of colon adenoma to colorectal cancer [84]. MUC-1 was highly immunogenic in about half of the patients evaluated. Moreover, response to the vaccine correlated with prevaccination levels of circulating MDSCs, as nonresponders had a significantly higher percentage of MDSCs (p < 0.05); interestingly, no such association was observed for regulatory T cells.

NeuVax is a peptide vaccine that has been developed for early-stage node-positive low or intermediate HER2-expressing breast cancer after standard of care treatment [85]. The vaccine is composed of a peptide isolated from HER2/neu proto-oncogene combined with GM-CSF. Final results of a phase 1/2 clinical trial showed a non-significant improvement in 5-year DFS of 89.7% in the vaccine group versus 80.2% in the control group (p = 0.08); the improvement in DFS was even greater in the sub-group of optimally dosed patients (94.6%; p = 0.05 versus the control group) [86]. The vaccine is now being tested in an ongoing phase 3 trial (NCT01479244).

CDX-110 is a peptide vaccine also known as the rindopepimut vaccine. It is a 14-mer peptide covering the EGFRvIII mutation (the commonest form of EGFR mutation in human glioblastoma multiforme (GBM), detected in 23–33% of tumours) [87], and it is linked to the adjuvant keyhole limpet hemocyanin (KLH) to stimulate a specific immune response against EGFRvIII expressing tumour cells. This vaccine has been evaluated in three phase 2 clinical trials (ACTIVATE, ACT II and ACT III trials) for newly diagnosed GBM and one phase 2 (ReACT trial) for recurrent GBM. The ACTIVATE trial demonstrated that patients with EGFRvIII-specific humoral responses had an improved median OS as compared to patients not displaying immune responses (47.7 months vs. 22.8 months OS) [88]. The ACT II and ACT III clinical trials demonstrated longer PFS and OS than with historically matched controls [88, 89]. Deceivingly, the phase 3 trial ACT IV for newly diagnosed GBM was terminated for futility at the second preplanned interim analysis (HR: 0.99 for rindopepimut versus control, 95% CI: 0.74–1.31) [90]. A lack of benefit was also observed in the intention-to-treat population. The study confirmed earlier-phase trial findings of rindopepimut-induced EGFRvIII-specific antibody responses in the majority of patients; however, the fact that loss of EGFRvIII was observed in patients receiving or not the vaccine suggests that this target is unstable and therefore not a suitable antigen for immunotherapy. The phase 2 ReACT trial, evaluating the combination of bevacizumab and rindopepimut for recurrent GBM, showed that the vaccine induced robust anti-EGFRvIII antibodies in the majority of patients. The primary endpoint of PFS at 6 months was improved for the rindopepimut arm, albeit non-significantly (28% vs. 16%, p = 0.12), with a similar outcome for OS and duration of response. Rapid anti-EGFRvIII antibody generation was shown to be associated with prolonged OS in the rindopepimut arm [91]. A major criticism for this study, which could explain the non-significant results, is that the EGFRvIII status was principally decided on diagnostic tumour specimens, despite the known fact that EGFRvIII expression is lost in half of tumours upon recurrence.

2.4.2 DC vaccines

In order to improve peptide presentation in vivo, cancer vaccines using DC have been developed. DC-based vaccines are safe and immunogenic, and they have the ability to promote clinically significant tumour regression in some patients [92, 93, 94]. Clinical trials performed with DC-based vaccines usually involve an individualised patient vaccination approach with single clinical trial arms, which makes it difficult to evoke firm conclusions about their efficacy. Several cells such as monocytes and CD34+ progenitor cells, antigens including complex tumour lysates and synthetic MHC class I-restricted peptides have all been used in different trials [95]. Some promising and important clinical trials involving DC vaccines have been published. Sipuleucel-T (Provenge) is one of the three FDA-approved vaccines (see Section 2.2.1). In addition to the IMPACT trial that led to FDA approval, 42 men with localised prostate cancer received Sipuleucel-T in a phase 2 study prior to radical prostatectomy [96]. Increased incidence of T cells was observed in the post-operative prostate gland histology compared to preoperative biopsies. Currently, clinical trials are investigating combination of Sipuleucel-T with other approved drugs, such as abiraterone acetate, enzalutamide, radium-223, ipilimumab and atezolizumab (NCT01487863, NCT01981122, NCT02463799, NCT01832870, NCT01804465, and NCT3024216).

A clinical trial that used DCs loaded with a MUC-1-derived peptide and heterologous pan DR epitope (PADRE) peptides (universal CD4 T-cell helper peptides) delivered subcutaneously in patients with RCC has shown encouraging objective clinical responses and immunologic responses [97]. A phase 1/2 clinical trial used autologous WT-1 (Wilms’ tumour 1, a shared TAA) mRNA-loaded DCs in patients with acute myeloid leukaemia (AML) in remission after standard of care, with the aim of eradicating or controlling residual disease. This study showed clinical responses correlating with increased WT-1-specific CD8+ T-cell frequencies, as well as elevated levels of post-vaccine-activated NK cells [98]. Another study used patient-derived AML cells fused with autologous DCs vaccination in post-chemotherapy remission AML patients, achieving a marked rise in circulating T cells recognising whole AML cells and leukaemia-specific antigens that persisted for more than 6 months, which was associated with prolonged survival [99].

DCVax is a DC vaccine that has been developed for GBM. Two phase 1/2 studies tested the vaccine, which collectively recruited 39 patients, 20 of whom had newly diagnosed GBM and the remaining had recurrent high-grade glioma [100, 101]. For the newly diagnosed patients, the median OS with the addition of DC vaccine to the standard of care chemoradiation was 36 months. Long-term survival was also reported for some patients; 33% of patients reached or exceeded a 4-year survival, 27% reached an OS of 6 years, and two patients achieved a 10-year survival. The first report of the DCVax 2:1 randomised phase 3 trial in newly diagnosed GBM unfortunately does not allow interpretation as it is endowed with methodological flaws [102].

Some findings have suggested that the current DC vaccines can be optimised in order to get improved clinical outcomes. The discovery that the overexpression of CD40L in human DCs produces an increased stimulation of the T-cell response to tumour antigens such as gp100 and Melan-A is promising [103]. Additionally, DC function can be enhanced by stimulating antigen-specific Th1 and CTL responses through modulation of other co-stimulatory or co-inhibitory molecules, such as PD-1, CTLA4, CD28, OX40, etc. [104, 105]. On the contrary, suppressing the ubiquitin-editing enzyme A20 or the scavenger receptor SRA/CD204 in human DC helps in the development of IFN-α-producing Th1 cells and antigen-specific CD8+ T cells [106, 107]. These developments suggest that there is promising data for the future in DC-based cancer vaccines.

2.4.3 Tumour cell vaccines

Tumours concentrate a high number of genetic modifications in somatic cells and therefore carry a large number of potential antigens. For that reason, vaccination with whole tumour cells has been an interesting strategy, with the limitation that they need to be patient-tailored. Autologous tumour cell vaccines have been evaluated in several cancer types such as lung cancer [108], melanoma [109, 110], RCC [111], prostate cancer [112] and colorectal cancer [113, 114]. In order to prepare the vaccine, a large amount of tumour tissue needs to be collected, which impedes its application in some tumour types or some individuals.

MVX-ONCO-1 is an autologous tumour cell vaccine containing irradiated tumour cells from a patient and a capsule implanted with a genetically modified allogeneic cell line that continuously releases the adjuvant GM-CSF [115]. Results of the first-in-human phase 1 trial testing of this vaccine reported an excellent safety profile, the main toxicity being a discomfort at the implantation site (20%) [116]. Over 50% of patients (8/15) experienced either partial response (PR) or stable disease (SD) including disappearance of lung metastases, with interesting activity in head and neck squamous cell carcinoma (HNSCC) and chordoma [117]. A phase 2 trial is ongoing in HNSCC (NCT02999646).

Canvaxin was the first allogeneic whole-cell vaccine to be developed and consisted of three melanoma cell lines in combination with BCG as adjuvant [118]. It showed promising results in a phase 2 clinical trial [119, 120], but failed in the randomised phase 3 trial [121]. Although the reasons for the lack of efficiency remain to be determined, it is possible that the induced immune response was not able to control the disease. To potentiate induction of immune response, tumour antigens utilised in vaccines should be linked with potent immunological adjuvants [122]. Such examples are tumour vaccines that have been modified genetically to express co-stimulatory molecules and/or cytokines. Such an example is the GVAX vaccine, an allogeneic whole-cell vaccine modified with the GM-CSF gene, which has been evaluated for recurrent prostate cancer [123, 124], breast cancer [125] and pancreatic cancer [126, 127], but impact on patient survival remains to be proven.

In order to improve the immunogenicity of allogeneic tumour cells, cell lines have been engineered to secrete antisense oligonucleotides to inhibit expression of immunosuppressive cytokines, such as TGF-β. The tumour vaccine Lucanix (Belagenpumatucel-L) has been designed using this strategy to target metastatic NSCLC and has shown significant improvement in OS in two phase 2 clinical trials [128, 129]. However, the phase 3 clinical trial in stage III/IV patients did not demonstrate prolongation of OS in the whole cohort of patients, a survival benefit being however observed in several subgroups of patients [130].

BiovaxID is a patient-specific therapeutic cancer vaccine composed of the patient clonal immunoglobulin molecule idiotype vaccine conjugated to the adjuvant KLH. In a phase 2 clinical trial, the administration of BiovaxID together with GM-CSF in patients diagnosed with follicular lymphoma in complete remission with minimal residual disease demonstrated induction of tumour-specific cellular and humoral immune responses, which translated into clinical benefit, with a median DFS of 8 years and an OS rate of 95% at 9 years [131]. A randomised, controlled phase 3 trial in patients achieving remission after chemotherapy showed a median DFS after randomisation of 44.2 months for the vaccine arm versus 30.6 months for control arm [132]. However, other phase 3 trials failed to demonstrate increase in survival for patients receiving the vaccine [133, 134].

The HyperAcute vaccines are made of tumour cell lines that have been genetically engineered to express the α(1,3)-galactosyltransferase enzyme in order to induce an hyperacute reaction with complement- and antibody-dependant cytotoxicity [135]. They have been tested in several malignancies including melanoma, pancreatic and prostate cancer [136, 137, 138] and showed encouraging results improving OS. This vaccine was further evaluated in two phase 3 clinical trials. The IMPRESS study evaluated the vaccine with or without gemcitabine/chemoradiation in resected pancreatic cancer patients but failed to achieve its primary endpoint, with no observed statistically significant difference between the treatment and control groups. The PILLAR trial for borderline resectable (stage II) and advanced unresectable (stage III) pancreatic adenocarcinoma patients, combining the vaccine with FOLFIRINOX or gemcitabine/nab-paclitaxel and chemoradiation, is currently ongoing.

2.4.4 Heat shock protein vaccines

Heat shock proteins (HSPs) are a group of intracellular protein chaperones. Their function is to protect cells from protein misfolding, dysfunction and cell apoptosis, and they have been implicated in the activation of innate and adaptive immunity [139]. Therapeutic HSPs vaccines utilise HSPs as a source of tumour-associated antigens and involve isolation and purification of HSPs from a patient’s tumour with subsequent reinfusion of the complex. The advantages of this type of vaccines are, similarly to tumour vaccines, that they do not require a pre-identification of tumour antigens and provide several targets at the same time.

GBM are natural inducers of HSP expression, making them an interesting target for HSP vaccines [139]. A phase 2 trial testing the HSPPC-96 vaccine in recurrent GMB patients showed a 90.2% 6-month OS and a 29.3% 12-month OS, with an interesting observation of an adverse effect of lymphopenia on the vaccination outcome [140]. Adjuvant vaccination following standard treatment by surgery and chemoradiation in patients with newly diagnosed GBM showed a median OS of 23.8 months [141]. Interestingly, this phase 2 trial showed better outcome (median OS: 44.7 months) in patients with low PD-L1-expressing myeloid cells than patients with high PD-L1 myeloid expression (median OS: 18 months) [141]. Nevertheless, a phase II randomised study (Alliance A071101) evaluating the combination of HSPPC-96 vaccine with bevacizumab versus bevacizumab alone in patients with recurrent GBM failed to demonstrate a survival benefit [142]. HSP-based vaccine has also been tested in various malignancies [143].

2.4.5 Viral vectors

Delivery of tumour antigens can be achieved using viral vectors. The advantage of virus-based vaccines is that human immune system has evolved to react efficiently against them with innate and adaptive responses, inducing long-lasting immunity. The most common viruses from which viral vaccines vectors have been developed are poxviruses, adenoviruses and alphaviruses [144]. A potentially restraining factor using viral vectors is the fact that the induced antiviral immune response will neutralise the vector, limiting efficacy of repeated vaccination with the same vector. In order to overcome this, heterologous prime-boost vaccination is used, where initial delivery of a tumour antigen with one virus vector is followed by a boost with the same tumour antigen delivered with another virus vector [145]. Using viral vector also offers the possibility to insert genes coding for adjuvants such as GM-CSF and IL-2.

As an example, the TRICOM vaccine platform exploits heterologous prime-boost vaccination where priming is achieved using a vaccinia vector encoding a chosen TAA and boosting using a fowlpox-derived vector encoding the same TAA. In addition, it incorporates three co-stimulatory molecules for immune activation and has been used in several trials in various malignancies. In men with CRPC, the PROSTVAC vaccine phase 3 trial, using PSA as antigen, failed to positively influence OS [146], although phase 2 trials were encouraging [147, 148]. An analysis of immune response to the PROSTVAC vaccine on pooled data from several clinical trials conducted similarly reported that 68% of the tested patients exhibited evidence of cross-priming with immune responses mounted against TAAs not found in the vaccine, for example, MUC-1, PSMA, PAP and PSCA, a phenomenon known as antigen spreading [149]. Other applications of the TRICOM vaccine in breast and ovarian cancer [150], solid carcinomas [151, 152], colorectal carcinoma [153] or advanced cancers [154] have been tested in phase 1 trials using various antigens and virus vectors, and further studies are planned.

Another example is BN-CV301, a poxvirus-based vaccine that codes for the MUC-1 and CEA TAAs. The phase 1 clinical trial showed no dose-limiting toxicity; the vaccine produced one PR in one patient and prolonged SD in multiple patients, especially in KRAS gastrointestinal cancer mutant patients [155].

Similarly, a first-in-human trial of the LV305 vaccine, a vaccine using DCs transduced with a lentivirus expressing the NY-ESO-1 antigen, demonstrated a favourable safety profile with grade 1/2 event such as fatigue (49%), injection (46%) and myalgia (21%); induction of anti-NY-ESO-1-specific CD4+ and CD8+ responses were observed, with a DCR of 56.4% in all patients and 62% in sarcoma patients [159].

2.4.6 Oncolytic virus vaccines

Oncolytic viruses are a particular category of viruses that have the characteristic of infecting both healthy and tumour cells, but of selectively replicating only in the latter. They therefore kill tumour cells, additionally inducing activation of innate and adaptive immune responses through immunogenic tumour cell death [156]. As for viral vectors, they also offer the possibility to express cytotoxic or immunomodulatory molecules. The herpes virus vaccine called T-VEC, engineered to selectively replicate in tumour cells and to secrete GM-CSF, has been approved by the FDA for intratumoural administration for stage IIIB/C-IV melanoma based on the phase 3 OPTiM trial [51, 157], as mentioned above (see Section 2.2.1). A recently reported series of off-trial uses of T-VEC in early advanced melanoma (stages IIIB/C-IVM1a) showed a CR rate of 61.5% and a PR rate of 26.9, with a DCR of 92.3% [158].

T-VEC is also being tested in other malignancies. In HNSCC, T-VEC was used in combination with standard chemoradiation for untreated unresectable stage III/IV disease in a phase 1/2 trial. At a median follow-up of 29 months, PFS was 76%, very importantly demonstrating the safety and feasibility of this combination approach [159]. The initial design of the phase 3 trial was subsequently modified in view of the introduction of pembrolizumab in the standard of care management of HNSCC and was redesigned as a phase 1b trial randomising patients to pembrolizumab with or without T-VEC delivered to involved cervical nodes (MASTERKEY-232, NCT2626000). This trial showed a manageable safety profile, with however 24/36 (66.7%) patients experiencing serious adverse events, including one vaccine-related death. The overall response rate was 16.7%, the majority of which was in patients with PD-L1-positive tumours, and the DCR was 38.9% (again mostly in PD-L1-positive tumours) [160].

2.5 Vaccine adjuvants

Vaccination “per se” can activate antigen-specific T cells. However, when the antigen is in the form of peptides, proteins or even tumour cells, they are usually not strong enough to induce an immune response that leads to tumour eradication. The reason for this is that these antigens come without pathogen-associated molecular pattern (PAMPs) that can be recognised by innate immune cells. Most cancer vaccines are therefore combined with adjuvants, which, in addition to eliciting an innate immune response, have the role of protecting the antigen from degradation, ensuring prolonged release and promoting antigen uptake by DCs. The efficiency and choice of the adjuvant heavily influences the vaccine efficacy.

Adjuvants that act as delivery systems are classified into virosomes, liposomes, the saponin QS-21, mineral salts and the water-in-oil emulsion Montanide (an incomplete Freund’s adjuvant analogue). Montanide is used in many trials of peptide vaccines and is generally well tolerated [61]. Aluminium is mostly used for antiviral vaccines such as the HPV vaccine as it promotes humoral rather than cellular responses [61]. Immunostimulatory complexes (ISCOMs) are ring-like structures containing lipids and saponin and can incorporate the antigen for optimal presentation for DCs. GM-CSF, which is employed to recruit and activate DCs at the injection site, is also being used in a large number of trials [61].

Innate immune stimulatory adjuvants are dominated by TLR ligands, but STING ligands, C-type lectin receptor (CLR) ligands and RIG-like receptor (RLR) ligands are also being tested [161]. TLR ligands induce a strong activation of DCs, and currently tested molecules include agonists to TLR2, TRL3 (e.g. the dsRNA analogue poly-ICLC), TLR7/8 (e.g. imiquimod) and TLR9 (e.g. the bacterial dinucleotide DNA CpGs). Many trials using CpGs have demonstrated its potential to improve T-cell responses, but it is now difficult to have access to it. Imiquimod is approved for the treatment of basal cell carcinoma and is used in combination with vaccines in several trials [61]. The TLR4 agonist glucopyranosyl lipid A (GLA) is currently used as adjuvants in peptide vaccines, such as with the NY-ESO-1 antigen [166]. Use of poly-ICLC is increasing, mostly for GBM vaccine trials, at it has proposed to favour T-cell homing to the brain [162].

Although many of the above-mentioned adjuvants are promising, the fear that using them alone would not induce strong enough immune response has led to development of combination strategies. Montanide is commonly used to protect the antigen in combination with a TLR ligand to promote inflammation. However, combining several immunostimulatory adjuvants such as two or more TLR ligands is being tested. Many more combination can be envisaged as long as safety is preserved.

2.6 Vaccine combinations

Vaccines, when efficiently designed, have the ability to induce strong T-cell responses. However, this does not imply that these T cells will be allowed to function at the tumour site, for several reasons. These include, among others, the immunosuppressive tumour microenvironment and the induction of immune checkpoint molecules on T cells. In an attempt to target these mechanisms, many combinations of vaccines with other immunotherapeutic strategies are currently in development. Checkpoint inhibitors, agonist antibodies and immunostimulatory cytokines can increase tumour cell immune destruction. Moreover, combining with radiotherapy, hormonotherapy and chemotherapy may also be synergistic.

2.6.1 Vaccines + checkpoint inhibitors

2.6.1.1 Vaccine + anti-CTLA-4 antibodies

CTLA-4 is expressed on T cells after activation as part of the normal regulation process of immune responses. However, in the case of antitumour responses, function of T cells need to be sustained, which is prevented by CTLA-4 expression [163]. To prevent that, two anti-CTLA-4 monoclonal antibodies, ipilimumab and tremelimumab, are currently in various stages of clinical development in combination with vaccines.

As examples, the PROSTVAC vaccine was tested with ipilimumab in mCRPC in a phase 1 escalation clinical trial. As a result, 14 of the 24 chemotherapy-naïve patients had reduction in PSA. Median OS was 31.3 months, which was longer than PROSTVAC alone [164]. This vaccine is currently being tested in combination with other checkpoint inhibitors (NCT2506114, NCT02933255, and NCT03532217).

GVAX was studied in combination with ipilimumab in 28 mCRPC patients in a phase 1 trial. Around 39% grade 3/4 irAEs were seen (most common: hypophysitis, alveolitis and hepatitis). About 25% had >50% decline in PSA, while 53.5% had SD radiologically [165]. GVAX has also been combined with ipilimumab in 30 pancreatic adenocarcinoma patients, versus ipilimumab alone [166]. The combination arm showed that three patients had extended SD and seven patients had a reduction in their tumour marker.

2.6.1.2 Vaccines + PD-1/PD-L1 inhibitors

PD-1 is a protein expressed on T cells, some B cells and NK cells, and binding of its ligands PD-L1 and PD-L2 results in cell inhibition [163]. PD1 ligands can be expressed not only by tumour cells but also by other cells of the tumour microenvironment, and PD-L1 has been shown to be induced as a result of T-cell activity [167]. The blocking of this interaction is being tested with the aim to allow prolonged T-cell activity to take place, and several anti-PD1 (pembrolizumab and nivolumab, among others) and anti-PD-L1 (atezolizumab, avelumab and durvalumab) antibodies have been developed.

Among others, combination of nivolumab with a multipeptide vaccine has been evaluated for the adjuvant treatment of high-risk melanoma. Results were promising, showing a median PFS of 47.1 months compared to historical median of 5–7.2 months with other approaches [168].

Pembrolizumab has been combined with a DNA vaccine encoding PAP in mCRPC patients. PSA responses were more important in the cohort receiving concurrent than sequential treatment. PSA declines were associated with the development of PAP-specific Th1-biased T-cell immunity and CD8+ T-cell infiltration in metastatic tumour biopsy specimens. No confirmed CR or PR was observed; however, 4/5 patients treated concurrently had measurable decreases in tumour volume at 12 weeks [169].

A multitude of studies are currently testing vaccines combinations with checkpoint inhibitors for different malignancies.

2.6.2 Vaccines + tyrosine kinase inhibitors

Tyrosine kinase inhibitors (TKIs) have been used for the treatment of several solid tumours and haematological malignancies. There is preclinical and clinical data proposing that TKIs have an “off-target” effect on immune cells that restraint and/or intensify the antitumour response [170].

A phase 3 trial evaluating the combination of sunitinib with a modified vaccinia Ankara-based vaccine encoding the tumour-associated antigen 5T4 (MVA-5T4) was not able to demonstrate benefit in OS, although patients with good-risk tumours responded better to the combination [171].

Based on the positive results of a phase 3 trial evaluating the epidermal growth factor (EGF) vaccine CIMAvax-EGF as switch maintenance therapy versus placebo for previously chemo-treated advanced NSCLC patients [172], a phase 1b study evaluating the CIMAvax-EGF vaccine in combination with EGFR TKI in EGFR-mutated NSCLC tumours (EPICAL trial) is currently ongoing (NCT03623750).

2.6.3 Vaccines + endocrine treatment

Endocrine treatment is important in hormonally driven tumours like prostate and breast cancer. Patients treated with letrozole, an aromatase inhibitor used for the adjuvant treatment of hormone-responsive breast cancer, were found to have less Tregs in the tumour microenvironment [172]. In addition, androgen deprivation therapy in prostate cancer patients generates an immunostimulatory microenvironment increasing the number of effector T cells [173, 174].

A post hoc analysis of a phase 3 randomised trial of the Sialyl Tn-KLH vaccine in women with metastatic breast cancer indicated an improved clinical outcome with the addition of concomitant endocrine therapy, with prolonged time to progression and OS [175]. The order of sequential treatment seemed to be important; a combination crossover study of nilutamide with a PSA-encoding poxvirus-based vaccine in non-metastatic CRPC suggested improved OS when the vaccine was administered before the hormonotherapy [176].

These combinations are attractive therapy options for hormonosensitive cancers because vaccines are minimally toxic and can easily be incorporated into standard of care regimens.

2.6.4 Vaccines + chemotherapy

Chemotherapy agents are known to induce reduction in both CD4+ and CD8+ T cells, however still allowing for immune responses to occur [177]. Several chemotherapeutic agents such as gemcitabine, taxanes, topoisomerase inhibitors, platinum compounds and 5-FU have been shown to produce immunomodulatory effects [177, 178].

The OPT-822 vaccine in combination with cyclophosphamide was tested in a phase 2/3 study in metastatic breast cancer versus cyclophosphamide plus placebo. The vaccination arm failed to show a PFS or interim OS benefit in the overall study population; however, they were significantly improved in the 50% of patients that developed an immune response to the vaccination [179].

IMA950 is a multipeptide GBM-specific vaccine composed of tumour-associated MHC class I- and II-restricted peptides [179]. The vaccine has been combined with standard chemoradiotherapy and adjuvant temozolomide in patients with newly diagnosed GBM in two reported trials. A phase 1 study of IMA950 adjuvanted with GM-CSF showed that the primary immunogenicity endpoint of observing multi-antigen responses in at least 30% of patients was reached. PFS was 74% at 6 months and 31% at 9 months [180]. The second clinical trial was a phase 1/2 trial of the IMA950 vaccine adjuvanted with poly-ICLC in high-grade gliomas; CD8 T-cell responses to a single or multiple peptides were observed in 63.2% and 36.8% of patients, respectively, while median OS was 19 months, comparing favourably to classical chemoradiation results [181]. A phase 1/2 trial evaluating the combination of the IMA950 vaccine with pembrolizumab in recurrent GBM is currently ongoing (NCT03665545).

2.6.5 Vaccines + radiotherapy

The concept of synergy between vaccines and radiotherapy attracts growing interest in cancer therapy. One of the hypotheses to explain this is that radiation can not only elicit a tumour-specific immune response locally but also at distant sites, therefore acting as an in situ vaccine, eliciting both local and systemic responses [182]. Many trials have tested and are currently testing vaccines and radiotherapy, and hope is that they will provide important information on how to optimise cancer vaccines.

Advertisement

3. Conclusions

Vaccine immunotherapy currently shows a prolific activity in early phase trials and an expanding pipeline, with however few successes in late phase trials, despite encouraging or promising early results, resulting in a limited number of approved drugs with modest therapeutic benefit. Furthermore, there have been therapeutic vaccine studies reported in the early or mid-2000s, without further translation or progression to later trial phases.

As our understanding of the potential of immunotherapy expands so does the list of research questions that will need to be answered before this approach can be translated for effective clinical use. Can the thus far limited success, reflected by the very few approved drugs, be attributed to suboptimal or inadequate trial design? What is the optimal endpoint for vaccine trials? How long would we need to treat patients with immune modulatory therapies? What is the best combination of approaches? What is the optimal sequence strategy?

It is evident that, in order to proceed in the next stage of therapeutic vaccine development, paradigm changes ought to probably be made towards more optimal utilisation of resources and therapeutic potential. We need a clearly defined clinical readout for therapeutic response, and we need a blueprint for successful translation. We might need to consider that the concept of using vaccines in stage IV disease is not the correct way forward, but rather bringing vaccines in earlier disease stages and developing adjuvant or maintenance strategies. In this context, OS might not be the correct endpoint to use, but disease-free or relapse-free survival might be more appropriate. Our understanding of the evolution of immune escape is still incomplete, and additional work must be done to identify those patients who will benefit most from immunotherapy and to develop novel strategies.

References

  1. 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA: A Cancer Journal for Clinicians. 2018;68(1):7-30
  2. 2. Bray F et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2018;68(6):394-424
  3. 3. Global Cancer Observatory. Available from: http://gco.iarc.fr/
  4. 4. Fridman WH et al. The immune contexture in cancer prognosis and treatment. Nature Reviews. Clinical Oncology. 2017;14(12):717-734
  5. 5. Couzin-Frankel J. Breakthrough of the year 2013. Cancer Immunotherapy. Science. 2013;342(6165):1432-1433
  6. 6. Castellarin M et al. Driving cars to the clinic for solid tumors. Gene Therapy. 2018;25(3):165-175
  7. 7. Pettitt D et al. CAR-T cells: A systematic review and mixed methods analysis of the clinical trials landscape. Molecular Therapy. 2018;26(2):342-353
  8. 8. Morales A, Eidinger D, Bruce AW. Intracavitary Bacillus Calmette-Guerin in the treatment of superficial bladder tumors. The Journal of Urology. 1976;116(2):180-183
  9. 9. Conry RM et al. Talimogene laherparepvec: First in class oncolytic virotherapy. Human Vaccines & Immunotherapeutics. 2018;14(4):839-846
  10. 10. Kantoff PW et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New England Journal of Medicine. 2010;363(5):411-422
  11. 11. Woo SR, Corrales L, Gajewski TF. Innate immune recognition of cancer. Annual Review of Immunology. 2015;33:445-474
  12. 12. Largeot A et al. The B-side of cancer immunity: The underrated tune. Cell. 2019;8(5):449
  13. 13. Steinman RM. Decisions about dendritic cells: Past, present, and future. Annual Review of Immunology. 2012;30:1-22
  14. 14. Zhu J, Paul WE. CD4 T cells: Fates, functions, and faults. Blood. 2008;112(5):1557-1569
  15. 15. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nature Reviews Immunology. 2012;12:253
  16. 16. Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell. 2005;7(3):211-217
  17. 17. Mittal D et al. New insights into cancer immunoediting and its three component phases—Elimination, equilibrium and escape. Current Opinion in Immunology. 2014;27:16-25
  18. 18. Donnou S et al. Murine models of B-cell lymphomas: Promising tools for designing cancer therapies. Advances in Hematology. 2012;2012:13
  19. 19. Boshoff C, Weiss R. AIDS-related malignancies. Nature Reviews Cancer. 2002;2(5):373-382
  20. 20. Weaver JL. Establishing the carcinogenic risk of immunomodulatory drugs. Toxicologic Pathology. 2012;40(2):267-271
  21. 21. Dunn GP et al. Cancer immunoediting: From immunosurveillance to tumor escape. Nature Immunology. 2002;3(11):991-998
  22. 22. Yuan J et al. Integrated NY-ESO-1 antibody and CD8+ T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab. Proceedings of the National Academy of Sciences. 2011;108(40):16723-16728
  23. 23. Ostroumov D et al. CD4 and CD8 T lymphocyte interplay in controlling tumor growth. Cellular and Molecular Life Sciences. 2018;75(4):689-713
  24. 24. Koebel CM et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007;450(7171):903-907
  25. 25. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: Integrating immunity's roles in cancer suppression and promotion. Science. 2011;331(6024):1565-1570
  26. 26. Vesely MD et al. Natural innate and adaptive immunity to cancer. Annual Review of Immunology. 2011;29(1):235-271
  27. 27. Gajewski TF et al. Molecular profiling to identify relevant immune resistance mechanisms in the tumor microenvironment. Current Opinion in Immunology. 2011;23(2):286-292
  28. 28. Miller JF, Sadelain M. The journey from discoveries in fundamental immunology to cancer immunotherapy. Cancer Cell. 2015;27(4):439-449
  29. 29. Adams JL et al. Big opportunities for small molecules in immuno-oncology. Nature Reviews Drug Discovery. 2015;14:603
  30. 30. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348(6230):62-68
  31. 31. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350-1355
  32. 32. Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. npj Vaccines. 2019;4(1):7
  33. 33. Keating GM. Bevacizumab: A review of its use in advanced cancer. Drugs. 2014;74(16):1891-1925
  34. 34. Weiner LM, Dhodapkar MV, Ferrone S. Monoclonal antibodies for cancer immunotherapy. The Lancet. 2009;373(9668):1033-1040
  35. 35. Berraondo P et al. Cytokines in clinical cancer immunotherapy. British Journal of Cancer. 2019;120(1):6-15
  36. 36. Mortara L et al. Anti-cancer therapies employing IL-2 cytokine tumor targeting: Contribution of innate, adaptive and immunosuppressive cells in the anti-tumor efficacy. Frontiers in Immunology. 2018;9:2905-2915
  37. 37. Mishra P, Nayak B, Dey RK. PEGylation in anti-cancer therapy: An overview. Asian Journal of Pharmaceutical Sciences. 2016;11(3):337-348
  38. 38. Barroso-Sousa R, Ott PA. Transformation of old concepts for a new era of cancer immunotherapy: Cytokine therapy and cancer vaccines as combination partners of PD1/PD-L1 inhibitors. Current Oncology Reports. 2018;20(12):1
  39. 39. Uy GL, Rettig MP, Cashen AF. Plerixafor, a CXCR4 antagonist for the mobilization of hematopoietic stem cells. Expert Opinion on Biological Therapy. 2008;8(11):1797-1804
  40. 40. Zhu MMT, Dancsok AR, Nielsen TO. Indoleamine dioxygenase inhibitors: Clinical rationale and current development. Current Oncology Reports. 2019;21(1):2
  41. 41. Yap TA et al. First-in-human study of KHK2455, a long-acting, potent and selective indoleamine 2,3-dioxygenase 1 (IDO-1) inhibitor, in combination with mogamulizumab (Moga), an anti-CCR4 monoclonal antibody, in patients (pts) with advanced solid tumors. Journal of Clinical Oncology. 2018;36(15_suppl):3040-3040
  42. 42. Leone RD, Emens LA. Targeting adenosine for cancer immunotherapy. Journal for Immunotherapy of Cancer. 2018;6(1):57
  43. 43. Rohaan MW et al. Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: A viable treatment option. Journal for Immunotherapy of Cancer. 2018;6(1):102
  44. 44. Bonini C, Mondino A. Adoptive T-cell therapy for cancer: The era of engineered T cells. European Journal of Immunology. 2015;45(9):2457-2469
  45. 45. Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discovery. 2013;3(4):388-398
  46. 46. Brown CE, Mackall CL. CAR T cell therapy: Inroads to response and resistance. Nature Reviews Immunology. 2019;19(2):73-74
  47. 47. Gomes C, Wong RJ, Gish RG. Global perspective on hepatitis B virus infections in the era of effective vaccines. Clinics in Liver Disease. 2019;23(3):383-399
  48. 48. Stanley M. Tumour virus vaccines: Hepatitis B virus and human papillomavirus. Philosophical Transactions of the Royal Society B: Biological Sciences. 2017;372(1732):20160268
  49. 49. Lamm DL et al. A randomized trial of intravesical doxorubicin and immunotherapy with Bacille Calmette-Guérin for transitional-cell carcinoma of the bladder. New England Journal of Medicine. 1991;325(17):1205-1209
  50. 50. Huber ML et al. Interdisciplinary critique of Sipuleucel-T as immunotherapy in castration-resistant prostate cancer. JNCI: Journal of the National Cancer Institute. 2012;104(4):273-279
  51. 51. Andtbacka RHI et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. Journal of Clinical Oncology. 2015;33(25):2780-2788
  52. 52. Ribas A et al. Determinant spreading and tumor responses after peptide-based cancer immunotherapy. Trends in Immunology. 2003;24(2):58-61
  53. 53. van der Bruggen P et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254(5038):1643-1647
  54. 54. Coulie PG et al. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nature Reviews Cancer. 2014;14(2):135-146
  55. 55. Fratta E et al. The biology of cancer testis antigens: Putative function, regulation and therapeutic potential. Molecular Oncology. 2011;5(2):164-182
  56. 56. Wei X et al. Cancer-testis antigen peptide vaccine for cancer immunotherapy: Progress and prospects. Translational Oncology. 2019;12(5):733-738
  57. 57. Bezu L et al. Trial watch: Peptide-based vaccines in anticancer therapy. OncoImmunology. 2018;7(12):e1511506
  58. 58. Vigneron N. Human tumor antigens and cancer immunotherapy. BioMed Research International. 2015;2015:17
  59. 59. Castle JC et al. Exploiting the mutanome for tumor vaccination. Cancer Research. 2012;72(5):1081-1091
  60. 60. Finn OJ, Rammensee H-G. Is it possible to develop cancer vaccines to neoantigens, what are the major challenges, and how can these be overcome?: Neoantigens: Nothing new in spite of the name. Cold Spring Harbor Perspectives in Biology. 2018;10(11):a028829-a028836
  61. 61. Gouttefangeas C, Rammensee H-G. Personalized cancer vaccines: Adjuvants are important, too. Cancer Immunology, Immunotherapy. 2018;67(12):1911-1918
  62. 62. Khong H, Overwijk WW. Adjuvants for peptide-based cancer vaccines. Journal for Immunotherapy of Cancer. 2016;4(1):56
  63. 63. Kirkwood JM et al. Immunogenicity and antitumor effects of vaccination with peptide vaccine+/−granulocyte-monocyte colony-stimulating factor and/or IFN-alpha2b in advanced metastatic melanoma: Eastern Cooperative Oncology Group Phase II Trial E1696. Clinical Cancer Research. 2009;15(4):1443-1451
  64. 64. Slingluff CL et al. A randomized phase II trial of multiepitope vaccination with melanoma peptides for cytotoxic T cells and helper T cells for patients with metastatic melanoma (E1602). Clinical Cancer Research. 2013;19(15):4228-4238
  65. 65. Pollack IF et al. Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas. Journal of Clinical Oncology. 2014;32(19):2050-2058
  66. 66. Parmiani G et al. Cancer immunotherapy with peptide-based vaccines: What have we achieved? Where are we going? JNCI: Journal of the National Cancer Institute. 2002;94(11):805-818
  67. 67. Guo C et al. Therapeutic cancer vaccines: Past, present, and future. Advances in Cancer Research. 2013;119:421-475
  68. 68. Antony PA et al. CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. The Journal of Immunology. 2005;174(5):2591-2601
  69. 69. van der Burg SH. Correlates of immune and clinical activity of novel cancer vaccines. Seminars in Immunology. 2018;39:119-136
  70. 70. Jr CLS et al. Immunologic and clinical outcomes of vaccination with a multiepitope melanoma peptide vaccine plus low-dose interleukin-2 administered either concurrently or on a delayed schedule. Journal of Clinical Oncology. 2004;22(22):4474-4485
  71. 71. Slingluff CL et al. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clinical Cancer Research. 2007;13(21):6386-6395
  72. 72. Jr CLS et al. Randomized multicenter trial of the effects of melanoma-associated helper peptides and cyclophosphamide on the immunogenicity of a multipeptide melanoma vaccine. Journal of Clinical Oncology. 2011;29(21):2924-2932
  73. 73. Hos BJ et al. Approaches to improve chemically defined synthetic peptide vaccines. Frontiers in Immunology. 2018;9:884-891
  74. 74. Swee LK et al. Sortase-mediated modification of αDEC205 affords optimization of antigen presentation and immunization against a set of viral epitopes. Proceedings of the National Academy of Sciences. 2013;110(4):1428-1433
  75. 75. Rahimian S et al. Polymeric nanoparticles for co-delivery of synthetic long peptide antigen and poly IC as therapeutic cancer vaccine formulation. Journal of Controlled Release. 2015;203:16-22
  76. 76. Varypataki EM et al. Synthetic long peptide-based vaccine formulations for induction of cell mediated immunity: A comparative study of cationic liposomes and PLGA nanoparticles. Journal of Controlled Release. 2016;226:98-106
  77. 77. Walter S et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nature Medicine. 2012;18(8):1254-1261
  78. 78. Rini BI et al. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): A multicentre, open-label, randomised, controlled, phase 3 trial. The Lancet Oncology. 2016;17(11):1599-1611
  79. 79. Hipp MM et al. Sorafenib, but not sunitinib, affects function of dendritic cells and induction of primary immune responses. Blood. 2008;111(12):5610-5620
  80. 80. Finke JH et al. Sunitinib reverses Type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clinical Cancer Research. 2008;14(20):6674-6682
  81. 81. Ko JS et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clinical Cancer Research. 2009;15(6):2148-2157
  82. 82. Ott PA et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217
  83. 83. Sahin U et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222
  84. 84. Kimura T et al. MUC1 vaccine for individuals with advanced adenoma of the colon: A cancer immunoprevention feasibility study. Cancer Prevention Research. 2012;6(1):18-26
  85. 85. Clifton GT et al. Clinical development of the E75 vaccine in breast cancer. Breast Care. 2016;11(2):116-121
  86. 86. Mittendorf EA et al. Final report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patients. Annals of Oncology. 2014;25(9):1735-1742
  87. 87. Pelloski CE et al. Epidermal growth factor receptor variant III status defines clinically distinct subtypes of glioblastoma. Journal of Clinical Oncology. 2007;25(16):2288-2294
  88. 88. Sampson JH et al. Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro-Oncology. 2010;13(3):324-333
  89. 89. Schuster J et al. A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: The ACT III study. Neuro-Oncology. 2015;17(6):854-861
  90. 90. Weller M et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. The Lancet Oncology. 2017;18(10):1373-1385
  91. 91. Reardon DA et al. IMCT-08 ReACT: Long-term survival from a randomized phase II study of rindopepimut (CDX-110) plus bevacizumab in relapsed glioblastoma. Neuro-Oncology. 2015;17(suppl_5):v109-v109
  92. 92. Pizzurro GA, Barrio MM. Dendritic cell-based vaccine efficacy: Aiming for hot spots. Frontiers in Immunology. 2015;6:91
  93. 93. Nestle FO et al. Vaccination of melanoma patients with peptide- or tumorlysate-pulsed dendritic cells. Nature Medicine. 1998;4(3):328-332
  94. 94. Banchereau J et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Research. 2001;61(17):6451-6458
  95. 95. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nature Reviews Cancer. 2012;12:265
  96. 96. Fong L et al. Neoadjuvant sipuleucel-T in localized prostate cancer: Effects on immune cells within the prostate tumor microenvironment. Journal of Clinical Oncology. 2012;30(15_suppl):2564-2564
  97. 97. Wierecky J et al. Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Research. 2006;66(11):5910-5918
  98. 98. Van Tendeloo VF et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proceedings of the National Academy of Sciences. 2010;107(31):13824-13829
  99. 99. Rosenblatt J et al. Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions. Science Translational Medicine. 2016;8(368):368ra171-368ra171
  100. 100. Liau LM et al. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clinical Cancer Research. 2005;11(15):5515-5525
  101. 101. Prins RM et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clinical Cancer Research. 2011;17(6):1603-1615
  102. 102. Liau LM et al. First results on survival from a large phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. Journal of Translational Medicine. 2018;16(1):142
  103. 103. Bonehill A et al. Single-step antigen loading and activation of dendritic cells by mRNA electroporation for the purpose of therapeutic vaccination in melanoma patients. Clinical Cancer Research. 2009;15(10):3366-3375
  104. 104. Bonehill A et al. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Molecular Therapy. 2008;16(6):1170-1180
  105. 105. Minkis K et al. Type 2 bias of T cells expanded from the blood of melanoma patients switched to type 1 by IL-12p70 mRNA-transfected dendritic cells. Cancer Research. 2008;68(22):9441-9450
  106. 106. Breckpot K et al. Attenuated expression of A20 markedly increases the efficacy of double-stranded RNA-activated dendritic cells as an anti-cancer vaccine. The Journal of Immunology. 2009;182(2):860-870
  107. 107. Yi H et al. Targeting the immunoregulator SRA/CD204 potentiates specific dendritic cell vaccine-induced T-cell response and antitumor immunity. Cancer Research. 2011;71(21):6611-6620
  108. 108. Rüttinger D et al. Adjuvant therapeutic vaccination in patients with non-small cell lung cancer made lymphopenic and reconstituted with autologous PBMC: First clinical experience and evidence of an immune response. Journal of Translational Medicine. 2007;5(1):43
  109. 109. Baars A et al. Skin tests predict survival after autologous tumor cell vaccination in metastatic melanoma: Experience in 81 patients. Annals of Oncology. 2000;11(8):965-970
  110. 110. Berd D et al. Immunopharmacologic analysis of an autologous, Hapten-modified human melanoma vaccine. Journal of Clinical Oncology. 2004;22(3):403-415
  111. 111. Antonia SJ et al. Phase I trial of a B7-1 (CD80) gene modified autologous tumor cell vaccine in combination with systemic interleukin-2 in patients with metastatic renal cell carcinoma. The Journal of Urology. 2002;167(5):1995-2000
  112. 112. Berger M et al. Phase I study with an autologous tumor cell vaccine for locally advanced or metastatic prostate cancer. Journal of Pharmacy & Pharmaceutical Sciences. 2007;10(2):144-152
  113. 113. Harris JE et al. Adjuvant active specific immunotherapy for stage II and III colon cancer with an autologous tumor cell vaccine: Eastern cooperative oncology group study E5283. Journal of Clinical Oncology. 2000;18(1):148-148
  114. 114. Hanna JMG. Immunotherapy with autologous tumor cell vaccines for treatment of occult disease in early stage colon cancer. Human Vaccines & Immunotherapeutics. 2012;8(8):1156-1160
  115. 115. Lathuilière A, Mach N, Schneider BL. Encapsulated cellular implants for recombinant protein delivery and therapeutic modulation of the immune system. International Journal of Molecular Sciences. 2015;16(5):10578-10600
  116. 116. Migliorini D et al. 531 MVX-ONCO-1: First in man, phase I clinical trial combining encapsulation cell technology and irradiated autologous tumor cells for personalized cell-based immunotherapy. Safety, feasibility and clinical outcome results. European Journal of Cancer. 2015;51:S114
  117. 117. Migliorini D et al. First report of clinical responses to immunotherapy in 3 relapsing cases of chordoma after failure of standard therapies. OncoImmunology. 2017;6(8):e1338235
  118. 118. Morton DL et al. Prolongation of survival in metastatic melanoma after active specific immunotherapy with a new polyvalent melanoma vaccine. Annals of Surgery. 1992;216(4):463-482
  119. 119. Morton DL et al. Prolonged survival of patients receiving active immunotherapy with Canvaxin therapeutic polyvalent vaccine after complete resection of melanoma metastatic to regional lymph nodes. Annals of Surgery. 2002;236(4):438-448 discussion 448-9
  120. 120. Hsueh EC et al. Prolonged survival after complete resection of disseminated melanoma and active immunotherapy with a therapeutic cancer vaccine. Journal of Clinical Oncology. 2002;20(23):4549-4554
  121. 121. Sondak VK, Sabel MS, Mulé JJ. Allogeneic and autologous melanoma vaccines: Where have we been and where are we going? Clinical Cancer Research. 2006;12(7):2337s-2341s
  122. 122. Ward S et al. Immunotherapeutic potential of whole tumour cells. Cancer Immunology, Immunotherapy. 2002;51(7):351-357
  123. 123. Simons JW et al. Phase I/II trial of an allogeneic cellular immunotherapy in hormone-Naïve prostate cancer. Clinical Cancer Research. 2006;12(11):3394-3401
  124. 124. Small EJ et al. Granulocyte macrophage colony-stimulating factor–secreting allogeneic cellular immunotherapy for hormone-refractory prostate cancer. Clinical Cancer Research. 2007;13(13):3883-3891
  125. 125. Emens LA et al. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: A chemotherapy dose-ranging factorial study of safety and immune activation. Journal of Clinical Oncology. 2009;27(35):5911-5918
  126. 126. Lutz E et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A Phase II trial of safety, efficacy, and immune activation. Annals of Surgery. 2011;253(2):328-335
  127. 127. Le DT et al. Results from a phase IIb, randomized, multicenter study of GVAX pancreas and CRS-207 compared with chemotherapy in adults with previously treated metastatic pancreatic adenocarcinoma (ECLIPSE study). Clinical Cancer Research; 24 June 2019. DOI: 10.1158/1078-0432.CCR-18-2992
  128. 128. Nemunaitis J et al. Phase II trial of belagenpumatucel-L, a TGF-β2 antisense gene modified allogeneic tumor vaccine in advanced non small cell lung cancer (NSCLC) patients. Cancer Gene Therapy. 2009;16:620
  129. 129. Nemunaitis J et al. Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer. Journal of Clinical Oncology. 2006;24(29):4721-4730
  130. 130. Giaccone G et al. A phase III study of belagenpumatucel-L, an allogeneic tumour cell vaccine, as maintenance therapy for non-small cell lung cancer. European Journal of Cancer. 2015;51(16):2321-2329
  131. 131. Bendandi M et al. Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma. Nature Medicine. 1999;5(10):1171-1177
  132. 132. Schuster SJ et al. Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma. Journal of Clinical Oncology. 2011;29(20):2787-2794
  133. 133. Levy R et al. Active idiotypic vaccination versus control immunotherapy for follicular lymphoma. Journal of Clinical Oncology. 2014;32(17):1797-1803
  134. 134. Levy R et al. Results of a Phase 3 trial evaluating safety and efficacy of specific immunotherapy, recombinant idiotype (Id) conjugated to KLH (Id-KLH) with GM-CSF, compared to non-specific immunotherapy, KLH with GM-CSF, in patients with follicular non-Hodgkin's lymphoma (fNHL). Cancer Research. 2008;68(9 Supplement):LB-204
  135. 135. Link CJ Jr et al. Eliciting hyperacute xenograft response to treat human cancer: Alpha(1,3) galactosyltransferase gene therapy. Anticancer Research. 1998;18(4A):2301-2308
  136. 136. Rossi GR et al. Effective treatment of preexisting melanoma with whole cell vaccines expressing alpha(1,3)-galactosyl epitopes. Cancer Research. 2005;65(22):10555-10561
  137. 137. Hardacre JM et al. Addition of algenpantucel-L immunotherapy to standard adjuvant therapy for pancreatic cancer: A phase 2 study. Journal of Gastrointestinal Surgery. 2013;17(1):94-101
  138. 138. Hemstreet GP 3rd et al. Cellular immunotherapy study of prostate cancer patients and resulting IgG responses to peptide epitopes predicted from prostate tumor-associated autoantigens. Journal of Immunotherapy. 2013;36(1):57-65
  139. 139. Graner MW, Bigner DD. Chaperone proteins and brain tumors: Potential targets and possible therapeutics. Neuro-Oncology. 2005;7(3):260-278
  140. 140. Bloch O et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: A phase II, single-arm trial. Neuro-Oncology. 2014;16(2):274-279
  141. 141. Bloch O et al. Autologous heat shock protein peptide vaccination for newly diagnosed glioblastoma: Impact of peripheral PD-L1 expression on response to therapy. Clinical Cancer Research. 2017;23(14):3575-3584
  142. 142. Bloch O et al. ATIM-14. Alliance A071101: A phase II randomized trial comparing the efficacy of heat shock protein peptide complex-96 (HSPPC-96) vaccine given with bevacizumab versus bevacizumab alone in the treatment of surgically resectable recurrent glioblastoma. Neuro-Oncology. 2017;19(suppl_6):vi29-vi29
  143. 143. Shevtsov M, Multhoff G. Heat shock protein–peptide and HSP-based immunotherapies for the treatment of cancer. Frontiers in Immunology. 2016;7:171-177
  144. 144. Larocca C, Schlom J. Viral vector-based therapeutic cancer vaccines. Cancer Journal. 2011;17(5):359-371
  145. 145. Harrop R, John J, Carroll MW. Recombinant viral vectors: Cancer vaccines. Advanced Drug Delivery Reviews. 2006;58(8):931-947
  146. 146. Gulley JL et al. Results of PROSPECT: A randomized phase 3 trial of PROSTVAC-V/F (PRO) in men with asymptomatic or minimally symptomatic metastatic, castration-resistant prostate cancer. Journal of Clinical Oncology. 2018;36(15_suppl):5006-5006
  147. 147. Kantoff PW et al. Overall survival analysis of a phase II randomized controlled trial of a poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. Journal of Clinical Oncology. 2010;28(7):1099-1105
  148. 148. Gulley JL et al. Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunology, Immunotherapy. 2010;59(5):663-674
  149. 149. Gulley JL et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunology Research. 2014;2(2):133-141
  150. 150. Mohebtash M et al. A pilot study of MUC-1/CEA/TRICOM poxviral-based vaccine in patients with metastatic breast and ovarian cancer. Clinical Cancer Research. 2011;17(22):7164-7173
  151. 151. Gulley JL et al. Pilot study of vaccination with recombinant CEA-MUC-1-TRICOM Poxviral-based vaccines in patients with metastatic carcinoma. Clinical Cancer Research. 2008;14(10):3060-3069
  152. 152. Duggan MC et al. A phase I study of recombinant (r) vaccinia-CEA(6D)-TRICOM and rFowlpox-CEA(6D)-TRICOM vaccines with GM-CSF and IFN-α-2b in patients with CEA-expressing carcinomas. Cancer Immunology, Immunotherapy. 2016;65(11):1353-1364
  153. 153. Lou E et al. A phase II study of active immunotherapy with PANVAC™ or autologous, cultured dendritic cells infected with PANVAC™ after complete resection of hepatic metastases of colorectal carcinoma. Clinical Colorectal Cancer. 2006;5(5):368-371
  154. 154. Heery CR et al. Phase I study of a poxviral TRICOM-based vaccine directed against the transcription factor brachyury. Clinical Cancer Research. 2017;23(22):6833-6845
  155. 155. Gatti-Mays ME et al. A phase 1 dose escalation trial of BN-CV301, a recombinant poxviral vaccine targeting MUC1 and CEA with costimulatory molecules. Clinical Cancer Research. 25(16):4933-4944
  156. 156. Russell L et al. Oncolytic viruses: Priming time for cancer immunotherapy. BioDrugs. 2019. https://doi.org/10.1007/s40259-019-00367-0
  157. 157. Andtbacka RHI et al. Final analyses of OPTiM: A randomized phase III trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in unresectable stage III–IV melanoma. Journal for Immunotherapy of Cancer. 2019;7(1):145
  158. 158. Franke V et al. High response rates for T-VEC in early metastatic melanoma (stage IIIB/C-IVM1a). International Journal of Cancer. 2019;145(4):974-978
  159. 159. Harrington KJ et al. Phase I/II study of oncolytic HSV GM-CSF in combination with radiotherapy and cisplatin in untreated stage III/IV squamous cell cancer of the head and neck. Clinical Cancer Research. 2010;16(15):4005-4015
  160. 160. Harrington KJ et al. Safety and preliminary efficacy of talimogene laherparepvec (T-VEC) in combination (combo) with pembrobrolizumab (Pembro) in patients (pts) with recurrent or metastatic squamous cell carcinoma of the head and neck (R/M HNSCC): A multicenter, phase 1b study (MASTERKEY-232). Journal of Clinical Oncology. 2018;36(15_suppl):6036-6036
  161. 161. Temizoz B, Kuroda E, Ishii KJ. Vaccine adjuvants as potential cancer immunotherapeutics. International Immunology. 2016;28(7):329-338
  162. 162. Okada H. Brain tumor immunotherapy with type-1 polarizing strategies. Annals of the New York Academy of Sciences. 2009;1174:18-23
  163. 163. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews Cancer. 2012;12(4):252-264
  164. 164. Madan RA et al. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: A phase 1 dose-escalation trial. The Lancet Oncology. 2012;13(5):501-508
  165. 165. van den Eertwegh AJM et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: A phase 1 dose-escalation trial. The Lancet Oncology. 2012;13(5):509-517
  166. 166. Le DT et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. Journal of Immunotherapy. 2013;36(7):382-389
  167. 167. Alspach E, Lussier DM, Schreiber RD. Interferon γ and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity. Cold Spring Harbor Perspectives in Biology. 2019;11(3):a028480-a028500
  168. 168. Gibney GT et al. Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clinical Cancer Research. 2015;21(4):712-720
  169. 169. McNeel DG et al. Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer. Oncotarget. 2018;9(39):25586-25596
  170. 170. Ott PA, Adams S. Small-molecule protein kinase inhibitors and their effects on the immune system: Implications for cancer treatment. Immunotherapy. 2011;3(2):213-227
  171. 171. Amato RJ et al. Vaccination of metastatic renal cancer patients with MVA-5T4: A randomized, double-blind, placebo-controlled phase III study. Clinical Cancer Research. 2010;16(22):5539-5547
  172. 172. Generali D et al. Immunomodulation of FOXP3+ regulatory T cells by the aromatase inhibitor letrozole in breast cancer patients. Clinical Cancer Research. 2009;15(3):1046-1051
  173. 173. Mercader M et al. T cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancer. Proceedings of the National Academy of Sciences. 2001;98(25):14565-14570
  174. 174. Drake CG et al. Androgen ablation mitigates tolerance to a prostate/prostate cancer-restricted antigen. Cancer Cell. 2005;7(3):239-249
  175. 175. Ibrahim NK et al. Survival advantage in patients with metastatic breast cancer receiving endocrine therapy plus Sialyl Tn-KLH vaccine: Post hoc analysis of a large randomized trial. Journal of Cancer. 2013;4(7):577-584
  176. 176. Madan RA et al. Analysis of overall survival in patients with nonmetastatic castration-resistant prostate cancer treated with vaccine, nilutamide, and combination therapy. Clinical Cancer Research. 2008;14(14):4526-4531
  177. 177. Galluzzi L et al. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell. 2015;28(6):690-714
  178. 178. Zitvogel L, Kroemer G. Anticancer immunochemotherapy using adjuvants with direct cytotoxic effects. The Journal of Clinical Investigation. 2009;119(8):2127-2130
  179. 179. Dutoit V et al. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy. Brain. 2012;135(Pt 4):1042-1054
  180. 180. Rampling R et al. A cancer research UK first time in human phase I trial of IMA950 (novel multipeptide therapeutic vaccine) in patients with newly diagnosed glioblastoma. Clinical Cancer Research. 2016;22(19):4776-4785
  181. 181. Migliorini D et al. Phase I/II trial testing safety and immunogenicity of the multipeptide IMA950/poly-ICLC vaccine in newly diagnosed adult malignant astrocytoma patients. Neuro-oncology. 2019;21(7):923-933
  182. 182. Demaria S, Formenti S. Radiation as an immunological adjuvant: Current evidence on dose and fractionation. Frontiers in Oncology. 2012;2:153-159

Written By

Carmen Murias Henriquez, Hendrik-Tobias Arkenau, Valérie Dutoit and Anna Patrikidou

Submitted: 12 March 2019 Reviewed: 08 August 2019 Published: 13 November 2019