Putting into Perspective the Future of Cancer : Targeted Immunotherapy

Authors: Issam Makhoul,1,2 *Thomas Kieber-Emmons2,3

1. Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA 2. Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA 3. Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA *Correspondence to [email protected]

Disclosure: The authors have declared no conflicts of interest.

Received: 11.11.19

Accepted: 12.02.20

Keywords: Cancer, cancer , checkpoint inhibitors.

Citation: EMJ. 2020;5[3]:102-113.

Abstract Pre-clinical models and human clinical trials have confirmed the ability of cancer vaccines to induce immune responses that are tumour-specific and, in some cases, associated with clinical response. However, cancer vaccines as a targeted immunotherapy strategy have not yet come of age. So, why the discordance after so much research has been invested in cancer vaccines? There are several reasons for this that include: limited tumour immunogenicity (limited targeted antigen expression, antigen tolerance); antigenic heterogeneity in tumours; heterogeneity of individual immune responses; multiple mechanisms associated with suppressed functional activity of immune effector cells, the underlying rationale for the use of immune checkpoint inhibitors; and immune system exhaustion. The success of checkpoint therapy has refocussed investigations into defining relationships between tumours and host immune systems, appreciating the mechanisms by which tumour cells escape immune surveillance and reinforcing recognition of the potential of vaccines in the treatment and prevention of cancer. Recent developments in cancer immunotherapies, together with associated technologies, for instance, the unparalleled achievements by immune checkpoint inhibitors and neo- antigen identification tools, may foster potential improvements in cancer vaccines for the treatment of malignancies.

INTRODUCTION various genetic alterations occurs later in the transformation process when genetic instability 2 Cancer is a genetic and epigenetic disease of has reached a critical level. Understanding these multicellularity, driving transformed cells to molecular changes, along with their protein uncontrolled growth, invasion, and metastasis. expression correlates, adds more precision to In cancer, intracellular mechanisms controlling the anatomical classification of cancers and has cellular proliferation are damaged first, which ushered in the era of targeted therapy. This effort allows certain cells to progress to malignant has led to the discovery of new targets and drugs, transformation.1 Activation of oncogenes by and the definition of new biomarkers.

102 EMJ • September 2020 EMJ The recent, dramatic success of immunotherapy STRATEGIES AND APPROACHES IN for treatment of some of the most highly DEVELOPMENT standard chemotherapy-resistant cancers (e.g., melanoma, lung cancer) has refocussed Interest in targeted by research on the role of the immune system as has been reinvigorated by the U.S. the main extracellular mechanism for cancer Food and Drug Administration (FDA) approval control. Using established databases, this new of ICI, which has had an impact on vaccine direction of research allows for the definition strategies for use in cancer therapy.9,10 Cancer of different immune profiles across cancer vaccines, as a targeted immunotherapy strategy types.3,4 Novel biomarkers, identified using used for prevention (primary or secondary) or simple immunohistochemistry (Immunoscore®) for treatment, have shown promise in preclinical or multi-omic methods, have been defined and animal studies, with the future aim of translation validated, as well as led to better prognostication that may surpass the traditional tumour, node, to the clinic. Some success is evident in the ® metastasis (TNM) staging system.3,5 A major FDA approval of PROVENGE (sipuleucel-T), a focus of ongoing research is to determine why herpes simplex virus Type 1-derived oncolytic immunotherapies work or fail, and how they can (T-VEC) immunotherapy that is injected directly ® be improved to reach their hoped-for potential into melanoma lesions; and in , which as a broadly transformative treatment for cancer. targets human papillomavirus known to cause Based on the presence of lymphocyte infiltrates cervical cancer. General research consensus is and their types, three major immune phenotypes that a vaccine for cancer as a single entity is not have emerged that correlate with response to practical because cancer reflects a myriad of immunotherapy: hot or inflamed, which respond different conditions. well to immune checkpoint inhibitors (ICI); cold The role of immunity in eradicating cancer is or ‘immune desert,’ which do not respond to now considered in terms of stepwise events or ICI; and two subtypes within the altered (both the ‘immunity cycle’, a framework proposed excluded and immunosuppressed) immune by Chen and Mellman.6,11 This cycle identifies phenotype. The immunosuppressed phenotype six steps preceding the killing of cancer cells is also expected to respond to ICI as it has pre- by the immune system (Figure 1). Once the 6,7 existing activated immune cells in the tumour. immune system is activated it is expected, It is these phenotypes that cancer-focussed based on clinical and preclinical studies, that vaccines aim to address. ‘Hot’ tumours often have immunosuppression would ensue to stop the a high mutational load and therefore are expected immune attack against the tumour.7 Therefore, to express neo-antigens to provoke a strong testing is underway of multiple strategies and immune response. Cold tumours, by contrast, approaches to activate the immune system for are cancers that, for various reasons, have not both hot and cold tumours (Table 1). been recognised or provoked a strong response Peptide-based vaccines rely on the development by the immune system. Herein lies one of the of molecular tools for improving, as well as limitations of immunotherapy. Characteristically studying, peptide-based vaccines.12 Whole-cell hot tumours are limited and include bladder lysate vaccines are applicable to all patients, cancer, head and neck cancers, kidney regardless of human leukocyte antigen (HLA) cancer, liver cancer, melanoma, and non-small type.13 Recombinant DNA or viral vector- cell lung cancer, as well as tumours of different based vaccines focus on design, delivery, and types with high microsatellite instability. The combination strategies that break tolerance and challenge is in the application of immunotherapy generate a strong immune response.14 Dendritic to cancers that are immunologically cold, such cells are the most effective antigen-presenting as glioblastomas, ovarian, prostate, and pancreatic cells for inducing T-cell proliferation, activation, cancer.8 One of the most important questions and cross priming.15 Tumour-associated for the future of immunotherapy is to determine carbohydrate mimetics are peptides that mimic how to make cold tumours immunoresponsive. the carbohydrate three-dimensional configuration on certain cancer-related proteins.16,17

Creative Commons Attribution-Non Commercial 4.0 September 2020 • EMJ 103 INSIGHT REVIEW

Trafficking of T cells to tumours 4 Immune-excluded tumour

RANKL Priming and Glucocorticoids Cancer activation of T cells Therapeutic 3 1,25(OH) Antigen agents immunogenicity Fas ligand Infiltration of T cells NSAID Vitamin A Environmental 2 HMGCR D3 5 into tumours inhibitors factors TGF-β Age-related loss of LOX Microbiome TCR diversity Anti-VEGF Host VEGF Traditional genetics Chinese TCR Endotoxin CAF/collagen medicine HLA type response Exercise- TNF-α Chemokines NF-κB induced IL-6 FcγRIII Inflammasome pathway Antigen expression tumour TLR D3 NOD2 1,25(OH)2 ATG16L MHC lass I expression Immune-desert TNF-α ↓ C HMGCR ↓ TAP-1 2 inhibitors JAK–STAT MEKi Cancer-antigen Photoimmune ROS presentation IL-6 ARNTL ↓ B2M Recognition of BRAF 6 Neoantigens cancer cells by KRAS T cells Viral Exercise- antigens induced IL-10 Cancer- RANKL associated PD-L1 antigens IDO-1 Inflamed Hypoxia tumour Negative effect 1 Positive effect Release of 7 Negative or positive effect cancer-cell antigens Killing of cancer cells

Figure 5 | Factors that influence the cancer–immune set point. A map nuclear translocator–like protein 1; ATG16L, autophagy-related ofcancerimmunityshowingthefactorsthataffectthecancer–immune protein 16; FcγRIII, Fc γ receptor III; HMGCR, HMG-CoA setpoint.Thefactorsareplacedonringsthatdenotetheirtype,and reductase; JAK/STAT, Janus kinase–signal transducers and activators eachfactorisalsoplacedinthestepofthecancer-immunitycycleinFigure 1: Multiple factors (tumour, host, and environment) affectof transcription; each step LOX, of the lysyl immunity oxidase; NOD,cycle. nucleotide-binding whichtheymainlyact.Forexample,variationsinHLAtypereflecthost oligomerization domain-containing protein; NSAIDs, non-steroidal geneticsandareofgreatestimportanceforTThe immune cycle can be correlatedcell activation. with the Additionaldifferent immuneanti-inflammatory phenotypes, drugs;and molecular RANKL; receptorand cellular activator abnormalities. of NF-κB ligand; factors areARNTL: being discovered aryl hydrocarbon rapidly. ARNTL, receptor aryl nuclear hydrocarbon translocator-like; receptor ATG16L:ROS, reactive autophagy oxygen species.related 16-like; B2M: β2 microglobulin; CAF: cancer-associated fibroblasts; FcγRIII: Fcγ receptor III; HLA: human leukocyte antigen; HMGCR: 3-hydroxy-3-methylglutaryl-CoA reductase; JAK–STAT: Janus kinase-signal transducer and activator of transcription; Inflamed versus non-inflamed tumours formaintainingimmunehomeostasis,eveninthepresenceofanactive MEKi: mitogen-activated extracellular signal-regulated kinase inhibitor; MHC: major histocompatibility complex; What is the basis for the three immune profiles observed in tumours? antitumour immune response80,81. Toafirstapproximation,differencesbetweentheprofilescanbeNOD2: nucleotide binding oligomerisation domain-containing 2; NSAID: non-steroidal anti-inflammatory drugs; PD- ascribedL1: to programmed whether tumours death harbour ligand-1; an RANKL: inflammatory receptor microen activator- ofPredicting nuclear factor response κ-Β ligand; ROS: reactive oxygen species; vironment,whichcanreflectvariationsinanumberofcellularandTCR: T-cell receptor; TLR: toll-like receptors; VEGF: vascular endothelialThe immune-inflamed growth factor. phenotype correlates generally with higher other factorsAdapted (Fig. from4).Thedegreeofinflammationcanbegaugedby Chen and Mellman,11 used with permission. response rates to anti-PD-L1/PD-1 therapy51,62,67,69–71, which suggests the cellular content of the tumour —forexample,thepresenceof thatbiomarkerscouldbeusedaspredictivetools.Mostattentionhas immunecells,eitherintheparenchymaorattheinvasivemarginofthe beenpaidtoPD-L1,whichisthoughttoreflecttheactivityofeffector tumourThe78,79 . Inflamedanti-idiotype tumours also containtherapeutic proinflammatory vaccine cytokines harmfulTcellsbecauseitcanbeadaptivelyexpressedbymostcelltypesfollow- ingredients, many antigens are in 6,82 that shouldracotumomab provide a more has favourable been conditionallyenvironment for T-cellapproved activa- developmenting exposure to IFN- as γplant-based. In an increasingly vaccines, large clinical with dataonly set, it is tion and expansion, including type I and type II IFNs, IL-12, IL-23, becomingclearthattheexpressionofPD-L1inpretreatmentbiopsies 20,21 IL-1βin, tumour-necrosis Latin America factor (TNF)-as maintenanceα andIL-2.However,itisunclear therapy for afacilitatesenrichmentwithpeoplewhoaremostlikelytorespondto few undertaking clinical trials in humans. whetheradvanced the presence non-small of these cytokines cell islung the cause cancer. or consequence18 Recent of antibodies against PD-L1 or PD-1 (refs 62, 69, 70, 73, 75, 83 and 84). thecellularinflux.Theproductionoftropicchemokinesbylymphobreakthroughs in cancer immunotherapy- ThePD-L1expressionalsocorrelatesstronglywithvariousmarkersof discovery that cancer cells may evade the cytes demonstrateand myeloid cells isthat therefore clinical likely toresponses be an important correlate feature of responseactive cellular of immunity, tumour-reactive including IFN- Tγ ,cells granzymes has andignited CXCL9 and inflamed tumours. effortsCXCL10.ThepresenceofthesebiomarkersorotherssuchasTcellsthat to improve the efficacy of antitumour Non-inflamedwith activation tumours and generally expansion express cytokinesof tumour-specific that are associ- carrytheCD3antigenortumourmutationalburdenmayalsoenrich atedwithimmunesuppressionortolerance.TheycanalsocontaincellT lymphocytes that mostly target mutation- immunefor responders responses,1,2,67,85.WhenusedincombinationwithPD-L1expres- with the hope of removing typesassociatedwithimmunesuppressionortissuehomeostasis.Asbased neo-antigens.19 Due to their economically limitssion, these on biomarkersthe activation may enhance and maintenance predictive power of86 .T-cell Clinically, it well aseffective, regulatory T cells,cold these chain cells includetransport the lesser and characterized lack of effectorwillbeimportanttoselectindividualswhoaremostlikelytorespond function. populations of myeloid-derived suppressor cells (for example, immature to anti-PD-L1/PD-1 therapies given as single agents rather than those granulocytes) and tumour-associated macrophages, which are unacti- who might require combination therapy14,51, which could add consid- vated and often called M2 macrophages. However, regulatory T cells are erable toxicity54. This therapeutic approach, which combines a set of not associated uniquely with non-inflamed tumours as they typically specific biomarkers with a selection of potential therapeutic options, is EMJ • September 2020 EMJ accompany104 effector T cellsintoinflammatorysitesandareimportant referred to as personalized cancer immunotherapy. Scientifically, the

326 | NATURE | 541 | 19 JANUARY 2017 ǟ ƐƎƏƗ !,(++ - 4 +(2'#12 (,(3#"Ʀ / 13 .$ /1(-%#1  341#ƥ ++ 1(%'32 1#2#15#"ƥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ Table 1: Main types of cancer vaccines and adaptive immunotherapy.

Technology Important consideration

Peptide-based vaccines11,12 Identification of peptide: natural, designed

Tumour cell vaccines (autologous or allogeneic)13 All relevant candidate antigens should be contained within cell

Recombinant viruses or bacteria with tumour antigens14 Delivery efficacy of antigen-encoding genes

Dendritic cell vaccines15 Choice of antigen in loading

DNA or RNA vaccines16 Easy delivery of multiple antigens with one immunisation

Anti-idiotype vaccines17,18 Choosing the right anti-idiotype

TACA mimetics19 Fidelity of mimicry

Neo-antigen vaccines20-22 Personalised

TACA: tumour-associated carbohydrate mimetics.

Reprogramming a tumour microenvironment existing immunity may allow it to be utilised and to trigger T-cell activation and enhance tumour amplified in vaccine strategies. ICI development immunity, in effect, making a tumour hotter, demonstrated the concept of two nascent provides insight into enhancing immune responses: first, the innate immune surveillance response. A strategy in support of this involves of cancer cells, and second, the adaptive immune targeting the checkpoint’s programmed cell- response generated by the emerging tumour. death protein-1 (PD-1) and programmed death ligand-1 (PD-L1) along with cytotoxic THE NASCENT IMMUNE SYSTEM T-lymphocyte-associated antigen-4 (CTLA-4). Developed ICI are negative-regulators of T-cell The immune system has evolved to distinguish immune function but with different mechanisms self from non-self as a means to protect the host. 22 of action. A general feature of immune system mechanisms PD-1 is expressed on activated T and B cells, is that they detect structural features of non- natural killer T cells, Type 2 innate lymphoid cells self that mark them as distinct from host cells, (ILC-2), and myeloid cells.23 CTLA-4 is expressed reflecting a danger signal to the immune 24 25 on Treg, activated T, and B cells. Expression of system. The emergence of cancer co-opts CTLA-4 on human natural killer T cells is unknown. tissue-specific immune development to escape As a result, CTLA-4 blockade disrupts the T-cell detection, augmented by failure of the immune interaction with other antigen-presenting cells, system to perform its primary task of surveillance 26 such as dendritic cells, macrophages, or B cells, and elimination. However, there is evidence while anti-PD-1 blockade primarily blocks the that innate and adaptive surveillance does occur. tumour cell and cytotoxic CD8+ T-cell interaction. Natural anti-carbohydrate antibodies are known to mediate cancer cell death.27 Such natural Biomarker studies with anti-PD-1 and PD-L1, antibodies, as part of innate immune surveillance, along with CTLA-4 clinical trials, support the could promote tumour immunity by inducing hypothesis that these agents are most effective immunogenic cell death, leading to immune in patients who have pre-existing anticancer priming and epitope spreading. Vaccine-induced immunity. Determination of the basis of this pre- anti-carbohydrate antibodies have displayed

Creative Commons Attribution-Non Commercial 4.0 September 2020 • EMJ 105 the same antitumour characteristics as natural antigen-presenting cells. Two antigen types anti-glycan antibodies.17 Thus, it may be possible are mainly represented in clinical trials in to emulate innate antitumour responses in Table 2, with some considered as neo-antigens cancer therapies. and others as tumour-associated antigens (TAA), incorporated into various platforms. Burnet28-30 and Thomas31,32 hypothesised that Immune activity in cancer supports combining the immune system can recognise nascent ICI with trials involving personalised tumour- transformed cells, leading to elimination of the specific neo-antigens and adaptive responses primary tumour formation.33 Tumours that are in general. However, currently there are 45 not eliminated undergo a process of immune open vaccine trials without ICI, compared to 37 editing,34 a reflection of the dynamic nature of with ICI. immune surveillance, suggesting that at some point the antitumour immune surveillance was It is not clear whether cancer vaccines are working, whether innate or adaptive. When part of a rational approach aimed at defined the immune system is successful in eradicating mechanisms. Systems vaccinology is an incipient cancer cells, based on innate or emerging field that applies omics technologies, adaptive immune response, no traces remain in combination with bioinformatics tools such as of its action. Mouse studies suggest that the transcriptional network analysis and predictive immune system could initially see tumours as modelling, to study immune responses to 39 immunogenic, lending to a primed immune vaccination. This integration to vaccine design response.35-37 After this initial phase, and if the requires the understanding of the molecular cancer is not eliminated, the immune system network mobilised by vaccination. What are shuts down, which leads to cancer escape from the global correlates of successful vaccination, beyond the specific immune response to the immune surveillance. As a result, the failure antigens administered, for understanding to eradicate cancer is not a failure of immune the mechanisms that underlie successful priming. These observations have led to the immunogenicity? Functional genomics are hypothesis that immune sculpting may result being used to analyse specific molecular in the emergence of a less-immunogenic clone signatures and antigens, for use as predictors of that is undetected by the immune system or vaccination efficiency. The immune response to downstream suppressive mechanisms. These vaccination involves the coordinated induction downstream suppressive mechanisms include of master transcription factors that leads to immune checkpoints that may allow malignant the development of a broad, polyfunctional, cells to evade an effectively-primed immune and persistent immune response, integrating all response, hence the rationale for the emergence effector cells of the immune systems. of ICI.22 It was 54 years following the research of Burnett and Thomas that anti-CTLA-4 therapies were approved.38 TUMOUR MUTATIONAL BURDEN, NEO-ANTIGENS, This leads to the question, where are we with enhancing the nascent response? Among AND THE PERSONALISATION clinical trials on clinicaltrials.gov, the search term OF IMMUNOTHERAPY “cancer vaccine” returned 58 early Phase I and combined Phase I/II, and 25 Phase II cancer Significant past research has focussed on vaccine trials that are actively recruiting. The genetic abnormalities affecting cancer-related distribution of these trials is listed in Table 2. The genes (oncogenes and tumour suppressor focus on T cells as the major immune effector genes), to define oncogenic drivers, and select mechanism for the action of cancer vaccines the most important ones for therapeutic relies on processed tumour-directed peptides targeting. The remainder of the mutations that activate T cells. This simplified view discovered through genomic analyses were necessitates that, for acquired nascent considered irrelevant. With the advent of immunity, the cancer-immunity cycle is initiated next generation sequencing, a multitude of by the release of cancer cell antigens either mutations were detected, leading the field to shed by living cancer cells or released from consider tumour mutation burden and discover dying tumour cells (see Figure 1). In either neo-antigens. Research highlighting the role case, antigens are taken up and presented by of the immune system led to the discovery

106 EMJ • September 2020 EMJ Table 2: Summary of open vaccine trials with and without checkpoint inhibitors.

Vaccine type Early Phase I Phase I Phase I/II Phase II

Neoantigen peptide N/A 6 (1) 3 N/A

Cell-based 1 (3) N/A 1 (1) 5 (4)

TAA-peptide N/A 2 (8) 5 (2) 1 (5)

Vector-based (2) 4 (11) 2 (1) 7 (7)

Number in parentheses reflects the number of trials without immune checkpoint inhibitors combination. TAA: tumour-associated antigen.

that efficacy of ICI was correlated with tumour consideration.48 The fourth step is to select the mutation burden and the presence of these clinical setting for therapeutic application. Current neo-antigens.40-44 practice suggests that these vaccines would work best in the adjuvant or minimal residual Neo-antigens are specific to hot tumours and disease settings. may be unique to each patient. As a result, the anticancer vaccine effort has sought to develop Some of these tumour-specific neo-antigens are specific reactive T cells to these neo-antigens. known to be, or expected to be, common across Their diversity, however, makes adaptation a subset of patients and are called shared neo- for immediate clinical use more difficult,antigens. Clinical trials of neo-antigens are shown requiring complicated platforms capable of in Table 3,54-66 with some trials making use of rapid sequencing of the patient’s genome to typical peptide formulations while others involve determine the most likely neo-antigens to be a DNA or plasmid format. Neo-antigens such as produced and given to the patient.45-47 tumour-specific antigens (TAA) are considered more immunogenic compared to self-antigens. The first step in the development of neo-antigen TAA are now considered less favourable as vaccines is the definition of the cancer mutanome vaccine candidates for several reasons: 1) being for a specific patient.48 The availability of high shared with normal tissues; 2) immune tolerance; performance platforms for next generation and 3) heterogeneity within the same tissue, sequencing allows the rapid identification of tumour mutations in comparison to matched and among patients. However, heterogeneity healthy tissue samples. Alterations that are may also occur with neo-antigens due to likely to result in immunologically meaningful intratumour heterogeneity. Vaccines encoding mutations are single nucleotide variations, xenoantigens, ‘non-self’ proteins that are highly gene fusions, frame shifts by small insertions or homologous to their autologous counterparts, deletions, and cancer-associated epigenetic have been investigated as a means to increase aberrations49 (at the transcriptional, translational, immunogenicity and overcome tolerance to ‘self’ or post-translational levels). The second step antigens.67 Likewise, mimotopes or vaccines is selection of the best neo-epitopes for that incorporate peptide mimics of tumour vaccine design. Computational models have antigens can function by eliciting increased been developed to achieve this goal.50-53 The numbers of T cells that cross react with the third step is to select the format of delivery of native tumour antigen.68 Mimotopes, which are the vaccine. Commonly used formats include xenoantigens, can function like neo-antigens in long peptides and RNA. Others, such as DNA inducing immune responses because they are plasmids, engineered bacteria or viruses, different from self-antigens. Mimotopes have and antigen-loaded dendritic cells, are under broad applications.

Creative Commons Attribution-Non Commercial 4.0 September 2020 • EMJ 107 Table 3: Open clinical trials with neoantigen formulations.

Mode Intervention Phase Cancer Identification

Peptide NeoVax I Kidney NCT0295076654 Ipilimumab Peptide GRT-C903 I/II Non-small cell lung, colorectal, pancreatic, NCT0395323555 GRT-R904 shared neoantigen-positive solid tumours Nivolumab Ipilimumab

Peptide GRT-C901 I/II Non-small cell lung cancer, colorectal cancer, NCT0363971456 GRT-R902 gastroesophageal adenocarcinoma, urothelial Nivolumab carcinoma Ipilimumab

Peptide Atezolizumab I Urothelial/bladder cancer NCT0335923957 PGV-001 Poly ICLC Peptide Personalised vaccine I Advanced cancer NCT0356805858 Pembrolizumab

Peptide NEO-PV-01 I Advanced melanoma NCT0359728259 Nivolumab Adjuvant APX005M Ipilimumab Peptide ASV™ AGEN2017 I Solid tumour (adult) NCT0367302060

Peptide RO7198457 I Melanoma, non-small cell lung cancer, NCT0328996261 Atezolizumab bladder cancer, colorectal cancer, triple- negative breast cancer, renal cancer, head and neck cancer, other solid cancers

Peptide GEN-009 adjuvanted I/II Cutaneous melanoma, non-small cell NCT0363311062 vaccine lung cancer, squamous cell carcinoma of the Nivolumab head and neck/urothelial carcinoma, renal cell Pembrolizumab carcinoma

Neo-antigen Personalised neo- I Pancreatic cancer NCT0312210663 vector antigen DNA vaccine

Vector Durvalumab I Triple-negative breast cancer NCT0319904064 neo-antigen DNA vaccine Vector PROSTVAC-V I Metastatic hormone-sensitive prostate cancer NCT0353221765 PROSTVAC-F Nivolumab Ipilimumab Neo-antigen DNA vaccine Vector YE-NEO-001 I Colorectal cancer, breast cancer, head and NCT0355271866 Yeast neck squamous cell carcinoma, melanoma, non-small cell lung cancer, pancreatic cancer, liver cancer

Poly ICLC: polyinosinic-polycytidylic acid-poly-l-lysine carboxymethylcellulose; YE-NEO-001: neoepitope yeast vaccine.

108 EMJ • September 2020 EMJ For example, one mimotope of tumour- of the effector CD8 T cells. Macrophage and associated carbohydrate antigens is currently myeloid cell differentiation is shifted to the undergoing clinical testing.16,17 Mimotopes immunosuppression type. Multiple therapeutic could be utilised to facilitate responses to cold strategies have been proposed to overcome tumours by recruiting TAA cross-reactive T cells these obstacles.73,75 and antibodies.

Presence of neo-antigens alone does not DEFINING PATIENT COHORTS: completely trigger an effective immune WHO BENEFITS? response; how the new antigens are presented also plays a role. For example, the presentation The clinical experience with ICI has revealed of a neo-antigen in low quantities, and with that these drugs do not work for everyone; progressive minor modifications, may lead to there are responders and non-responders, and immune tolerance rather than immune rejection.69 only a minority of patients benefit.76 ICI work in defined cohorts of patients, relating to Conversely, high immunogenicity can still curtail levels of expression of the PD-1/PD-L1 axis, a ‘one size fits all’ vaccine because of inherent expression of mutated genes that lend to heterogeneity in mutational rates. Neo-antigen nascent responses, and those that have tumour- vaccines are considered a means to enhance the infiltrating lymphocytes and other immune nascent adaptive response, but to do so requires cells (i.e., hot tumours). This contributes to a vaccine to be developed from neo-antigens complexity in determining patient cohorts for for every patient. This challenge may suggest vaccine trials, which requires consideration an alternative strategy of using a whole-cell of the vaccine therapeutic mechanism to approach from a patient’s own tumour, to enhance the immune response in hot or customise or tailor a personalised vaccine. inflamed tumours (characterised by tumour- infiltrating lymphocytes), or alter the immune CANCER METABOLIC STRESS AND response in cold tumours to make them ‘hot’. In RESISTANCE TO IMMUNOTHERAPY addition, the PD-1/PD-L1 axis has the potential to be upregulated by transcriptional regulators that are yet to be defined but potentially Immune escape in cancer may occur in early associated with a cancer vaccine response. stages of the immune response, as the cancer Some vaccines under consideration might have undergoes immune editing and becomes invisible been dismissed because they upregulated to the immune system. Activating mutations transcriptional regulators known to shut down of certain oncogenes (KRAS, BRAF, or MAPK) the immune response. may result in decreased expression of major histocompatibility complex class-1 (MHC-I).70,71 In the current era of immunotherapy, with Alternatively, cancer may escape eradication at the lack of definitive biomarkers, evaluation the later stages of the immune cycle, even after of tumours based on both their immune a vigorous effector T-cell response, by losing the phenotype and genomic mutation profile may ability to be destroyed (for example, by mutation help determine which patients have a higher of CASP8).72 The cancer microenvironment likelihood of responding to immunotherapies. is characterised by hypoxia and decreased Clinically, tumour burden reveals patient cohorts availability of nutrients required for energy and associated with therapeutic efficacy for cancer cell structure maintenance, including glucose, vaccines. Passively administered antibodies lipids, and amino acids.73,74 Anaerobic metabolism have been found to eliminate circulating tumour in the presence of oxygen (Warburg effect), a cells and systemic or intraperitoneal hallmark of cancer, leads to the production of micrometastases in a variety of preclinical large quantities of lactic acid that impair the models; antibody-inducing vaccines may be function of immune cells. These metabolic beneficial in the adjuvant setting. Minimal changes lead to reprogramming of both the residual disease is an indication for effective use cancer cells and the immune cells in their of both monoclonal antibodies77 and for cancer microenvironment, resulting in a blunted vaccines.78 The results of the Keynote 522 Phase immune response to the cancer and suppression III ,79 comparing chemotherapy with

Creative Commons Attribution-Non Commercial 4.0 September 2020 • EMJ 109 pembrolizumab or placebo, revealed greater variables are included in this model: tumour benefit in advanced stage of breast cancer than foreignness, the patient’s general immune status, in early stage disease. immune cell infiltration, checkpoints, soluble inhibitors, inhibitory tumour metabolism, and Insights and strategies from the immune tumour sensitivity to immune effectors. foundation of ICI can be applied to the design and application of cancer vaccines, particularly to overcome the low antigenicity and heterogeneity CONCLUSION of tumour-specific antigens. These include: 1) targeting multiple immunogenic antigens through The ultimate goal of immunotherapy is to polyvalent formulations;80-82 2) targeting a high establish a durable population of highly active, fraction of tumour cells bearing each antigen, tumour-specific responses that can lyse tumour by considering the clonal nature of an antigen;83 cells and eradicate cancers. Evidence from and 3) deriving cancer vaccines from the most various clinical trials that reflect the biology of immunogenic clonal antigen-loaded patients.84 immune response and cancer targeting lends to The majority of patients are not responsive our understanding that cancer immunotherapy to ICI because of the lack of tumour-specific is a multifaceted strategy and that a single effector cells. Consequently, cancer vaccines treatment modality will not suffice. The discovery may be a means to elicit diverse antigen-specific of immune checkpoints and the success of their effector cells.85 inhibitors has led to detailed investigation of the complicated interactions between different Different measures of antigen-specific tolerance components of the immune system and or regulation may help predict immunological microenvironment involved in the anticancer outcome from vaccination.86 Santegoets et response.91 A plethora of co-stimulatory pathways al.87 demonstrated prolonged overall survival have been identified, with some now the subject following treatment with a cancer vaccine of intense investigation to assess the benefit of (GVAX) in combination with ipilimumab in their activation for augmenting the anticancer patients with advanced prostate cancer who immune response. Other inhibitory pathways had either: high pre-treatment frequencies of were identified and are being explored to assess CD4+ CTLA-4+, CD4+ PD-1+, or differentiated their role in different cancers. This line of research (non-naïve) CD8+ T cells; or low pre-treatment has revealed the complexity of the immune frequencies of regulatory T cells or differentiated landscape. CD4+ T cells. These parameters suggest a highly immunocompetent patient. Such findings CTLA-4 and PD-1/PD-L1 appear to be the suggest that the identification of predictive predominant immune checkpoints, but they biomarkers associated with long-term immune are not the only ones. Different tumours outcome could be beneficial for identifying may preferentially utilise particular inhibitory patients most likely to benefit from antitumour pathways. Eliciting an immune response through vaccines. One measure of immunocompetency, a tumour vaccine may also trigger these specific for consideration as an inclusion criterion inhibitory pathways. Research understanding at for cohort recruitment, is delayed-type this point assumes that vaccines that lead to the hypersensitivity to recall antigens.17 However, it release of high concentrations of INF-γ are likely has been suggested that this does not accurately to induce the overexpression of PD-L1 on tumour reflect immune competence in patients with cells, and may benefit from the combination advanced- stage breast cancer, as research has of the vaccine with PD-1/PD-L1 inhibitors. If demonstrated that patients who failed responses a vaccine were to increase the expression of 92 93 to recall antigens could still mount tumour- GAL9/Tim3 or GITRL/GITR, in addition to or specific T-cell responses to a tumour antigen instead of the PD-1/PD-L1 pathway, PD-1/PD- upon vaccination.88 L1 inhibitors alone would be of limited use, as targeting the specific pathways triggered by Blank et al.89 suggested the integration of the vaccine would be required. ICI rely on a all the parameters involved in the immune primed nascent response. Cancer vaccines response into one dynamic framework; they can provide priming and boosting of nascent called it the ‘cancer immunogram.’90 Seven responses but require ICI both to enhance a

110 EMJ • September 2020 EMJ response, in the case of CTL-4 therapy, and to allows for consideration of new combinations. block tumour suppression, as in the case of the Current understanding has determined that PD-1/PD-L1 axis. certain vaccines increase the release of IFN-γ, which in turn increases the expression of PD-L1 Tumours are heterogeneous in their antigenic on the tumour, leading to immune suppression make-up, and different antigens of the same that can be overcome with the use of ICI. tumour may have different immunogenicity. Some Questions remain concerning the timing of cancers succeed in evading the immune system treatments, adjuvants, immunisation routes, by decreasing their foreignness; others lose their optimal immunogenic vaccines, tumour expression of MHC-I and become invisible to the remodelling, and the cohort these combinations immune system. The metabolic microenvironment should be tested in. of the tumour favours the reprogramming of immune cells to Type 2 responses. Tumours differ A rational approach to the development in their ways of shutting off the immune system of vaccine-ICI combinations would require using different immune checkpoints. Hosts are detailed definition of the tumour antigenic also heterogeneous in their ability to mount an immunogenicity, immunogenic heterogeneity, immune response to tumour cells. Vaccines that and the inhibitory mechanisms that the tumour utilise tumour antigens could potentially induce uses to suppress the immune system; in effect, similar responses to their respective tumours and a systems-based immunology/vaccinology may trigger different inhibitory mechanisms. approach is needed. With various vaccine modalities and combinations to alter the cancer There is insufficient clinical data to reveal a microenvironment and the immune response breakthrough in cancer vaccines, but a better under research, personalised immunotherapy understanding of the tumour microenvironment could be a reality in the near future.

References

1. Baylin SB, Jones PA. Epigenetic 2019;10:168. doi: 10.3389/ the clinic. Hum Vaccin Immunother. determinants of cancer. Cold Spring fimmu.2019.00168. 2015;11(1):37-44. Harb Perspect Biol. 2016;8(9). doi: 019510.011101/cshperspect.a019505. 9. Cebon J. Perspective: cancer vaccines 17. Hutchins LF et al. Targeting tumor- in the era of immune checkpoint associated carbohydrate antigens: 2. Pancione M et al. Genetic and blockade. Mamm Genome. a phase I study of a carbohydrate epigenetic events generate 2018;29(11):703-13. mimetic- in stage IV multiple pathways in colorectal breast cancer subjects. Oncotarget. cancer progression. Patholog 10. Ye Z et al. Cancer vaccine: learning 2017;8(58):99161-78. Res Int. 2012;2012:509348. doi: lessons from immune checkpoint 10.1155/2012/509348. inhibitors. J Cancer. 2018;9(2):263-8. 18. Gabri MR et al. Racotumomab for 3. Thorsson V et al. The immune 11. Chen DS, Mellman I. Oncology meets treating lung cancer and pediatric landscape of cancer. Immunity. immunology: the cancer-immunity refractory malignancies. Expert Opin 2018;48(4):812-30.e814. cycle. Immunity. 2013;39(1):1-10. Biol Ther. 2016;16(4):573-8. 4. Thomas A et al. Tumor mutational 12. Hos BJ et al. Approaches to improve 19. Aurisicchio L et al. Poly-specific burden is a determinant of immune- chemically defined synthetic peptide neoantigen-targeted cancer vaccines mediated survival in breast cancer. vaccines. Front Immunol. 2018;9:884. delay patient derived tumor growth. Oncoimmunology. 2018;7:e1490854. doi: 10.3389/fimmu.2018.00884. J Exp Clin Cancer Res. 2019;38(1):78. doi: 10.1186/s13046-019-1084-4. 5. Pages F et al. International validation 13. Chiang CL et al. Whole tumor antigen of the consensus Immunoscore for vaccines: where are we? Vaccines 20. Takeyama N et al. Plant-based the classification of colon cancer: (Basel). 2015;3(2):344-72. vaccines for animals and humans: a prognostic and accuracy study. recent advances in technology and 14. Duperret EK et al. Designing Lancet. 2018;391(10135):2128-39. clinical trials. Ther Adv Vaccines. consensus immunogens to break 2015;3(5-6):139-54. 6. Chen DS, Mellman I. Elements tolerance to self-antigens for of cancer immunity and the cancer therapy. Oncotarget. 21. Wong-Arce A et al. Plant-made cancer-immune set point. Nature. 2018;9(85):35513-4. vaccines in the fight against cancer. 2017;541:321-30. Trends Biotechnol. 2017;35(3):241-56. 15. Mookerjee A et al. A cancer vaccine 7. Galon J, Bruni D. Approaches to with dendritic cells differentiated with 22. Wei SC et al. Distinct cellular treat immune hot, altered and GM-CSF and IFNalpha and pulsed mechanisms underlie anti-CTLA-4 cold tumours with combination with a squaric acid treated cell lysate and anti-PD-1 checkpoint blockade. immunotherapies. Nat Rev Drug improves T cell priming and tumor Cell. 2017;170(6):1120-33. Discov. 2019;18(3):197-218. growth control in a mouse model. Bioimpacts. 2018;8(3):211-21. 23. Beldi-Ferchiou A, Caillat-Zucman 8. Bonaventura P et al. Cold tumors: S. Control of NK cell activation by a therapeutic challenge for 16. Makhoul I et al. Moving a immune checkpoint molecules. Int J immunotherapy. Front Immunol. carbohydrate mimetic peptide into Mol Sci. 2017;18(10):2129.

Creative Commons Attribution-Non Commercial 4.0 September 2020 • EMJ 111 24. Ramadan A et al. Editorial: Immunol. 2016;39:14-22. https://clinicaltrials.gov/ct2/show/ danger signals triggering immune NCT03953235. response and inflammation. Front 42. Joshi K et al. The "Achilles' heel" of cancer and its implications Immunol. 2017;8:979. doi: 10.3389/ 56. Gritstone Oncology, Inc. A study of a for the development of novel fimmu.2017.00979. personalized cancer vaccine targeting immunotherapeutic strategies. Cold shared neoantigens. NCT03639714. 25. Pashov A et al. Thinking cancer. Spring Harb Perspect Med. 2018;8(1). https://clinicaltrials.gov/ct2/show/ Monoclon Antib Immunodiagn doi: 027010.021101/cshperspect. NCT03639714. Immunother. 2018;37(3):117-25. a027086. 57. Matthew Galsky. Atezolizumab given 26. Nirschl CJ et al. IFNγ-dependent 43. Yu H et al. Correlation of PD-L1 in combination with a personalized tissue-immune homeostasis expression with tumor mutation vaccine in patients with urothelial is co-opted in the tumor burden and gene signatures for cancer. NCT03359239. https:// microenvironment. Cell. prognosis in early-stage squamous www.clinicaltrials.gov/ct2/show/ 2017;170(1):127-141.e15. cell lung carcinoma. J Thorac Oncol. NCT03359239. 2019;14(1):25-36. 27. Vollmers HP, Brandlein S. Natural 58. Ezra Cohen. Personalized antibodies and cancer. N Biotechnol. 44. Hartmaier RJ et al. Genomic analysis immunotherapy in adults with 2009;25(5):294-8. of 63,220 tumors reveals insights advanced cancers immunotherapy into tumor uniqueness and targeted in adults with advanced cancers. 28. Burnet M. Cancer; a biological cancer immunotherapy strategies. NCT03568058. https://clinicaltrials. approach. I. The processes of control. Genome Med. 2017;9:16. doi: 10.1186/ gov/ct2/show/NCT03568058. Br Med J. 1957;1(5022):779-86. s13073-13017-10408-13072. 59. Neon Therapeutics, Inc. A personal 29. Burnet FM. Immunological 45. Parvizpour S et al. Breast cancer cancer vaccine (NEO-PV-01) recognition of self. Science. vaccination comes to age: impacts and APX005M or ipilimumab 1961;133(3449):307-11. of bioinformatics. Bioimpacts. with nivolumab in patients with 2018;8(3):223-35. 30. Burnet FM. The concept of advanced melanoma. NCT03597282. immunological surveillance. Prog Exp 46. Parvizpour S et al. In silico design of a https://clinicaltrials.gov/ct2/show/ Tumor Res. 1970;13:1-27. triple-negative breast cancer vaccine NCT03597282. by targeting cancer testis antigens. 31. Thomas L., “Discussion,” Lawrence 60. Agenus Inc. Phase 1a study to Bioimpacts. 2019;9(1):45-56. HS et al. (eds.), Cellular and Humoral evaluate immunogenicity of ASV. Aspects of Hypersensitive States 47. Hollingsworth RE, Jansen K. Turning NCT03673020. https://clinicaltrials. (1959), New York: Hoeber–Harper, the corner on therapeutic cancer gov/ct2/show/NCT03673020. pp.529-32. vaccines. NPJ Vaccines. 2019;4:7. doi: 61. Genentech, Inc. A study of 10.1038/s41541-41019-40103-y. 32. Thomas L. On immunosurveillance RO7198457 as a single agent and in in human cancer. Yale J Biol Med. 48. Sahin U, Tureci O. Personalized combination with atezolizumab in 1982;55(3-4):329-33. vaccines for cancer immunotherapy. participants with locally advanced or Science. 2018;359(6382):1355-60. metastatic tumors. NCT03289962. 33. Ribatti D. The concept of immune https://clinicaltrials.gov/ct2/show/ surveillance against tumors. The first 49. Laumont CM, Perreault C. Exploiting NCT03289962. theories. Oncotarget. 2017;8(4):7175- non-canonical translation to identify 80. new targets for T cell-based cancer 62. Genocea Biosciences, Inc. Safety, immunotherapy. Cell Mol Life Sci. tolerability, immunogenicity, and 34. Dunn GP et al. Cancer immunoediting: 2018;75(4):607-21. antitumor activity of GEN-009 from immunosurveillance to tumor adjuvanted vaccine. NCT03633110. escape. Nat Immunol. 2002;3(11):991-8. 50. Gfeller D et al. Current tools for https://clinicaltrials.gov/ct2/show/ predicting cancer-specific T cell 35. Foley EJ. Antigenic properties of NCT03633110. immunity. Oncoimmunology. methylcholanthrene-induced tumors 2016;5(7):e1177691. doi: 1177610.117108 63. Washington University School of in mice of the strain of origin. Cancer 0/2162402X.1172016.1177691. Medicine. Neoantigen DNA vaccine in Res. 1953;13(12):835-7. pancreatic cancer patients following 51. Shao XM et al. High-throughput 36. Klein G et al. Demonstration surgical resection and adjuvant prediction of MHC class I and class chemotherapy. NCT03122106. of resistance against II neoantigens with MHCnuggets. https://clinicaltrials.gov/ct2/show/ methylcholanthrene-induced Cancer Immunol Res. 2019. doi: NCT03122106. sarcomas in the primary 10.1158/2326-6066.CIR-19-0464. autochthonous host. Cancer Res. 64. Washington University School 1960;20:1561-72. 52. Bonsack M et al. Performance of Medicine. Neoantigen DNA evaluation of MHC class-I binding vaccine alone vs. neoantigen DNA 37. Prehn RT, Main JM. Immunity to prediction tools based on an vaccine plus durvalumab in triple methylcholanthrene-induced experimentally validated MHC- negative breast cancer patients sarcomas. J Natl Cancer Inst. peptide binding data set. Cancer following standard of care therapy. 1957;18(6):769-78. Immunol Res. 2019;7:719-36. NCT03199040. https://clinicaltrials. 38. Lipson EJ, Drake CG. Ipilimumab: an 53. Mei S et al. A comprehensive review gov/ct2/show/NCT03199040. anti-CTLA-4 antibody for metastatic and performance evaluation of 65. Washington University School of melanoma. Clin Cancer Res. bioinformatics tools for HLA class Medicine. Neoantigen DNA vaccine 2011;17(22):6958-62. I peptide-binding prediction. Brief in combination with nivolumab/ Bioinform. 2019. [Epub ahead 39. Hagan T et al. Systems vaccinology: ipilimumab and PROSTVAC in of print]. enabling rational vaccine design metastatic hormone-sensitive with systems biological approaches. 54. Patrick Ott, MD. A study combining prostate cancer. NCT03532217. Vaccine. 2015;33(40):5294-301. NeoVax, a personalized NeoAntigen https://clinicaltrials.gov/ct2/show/ 40. Alexandrov LB, Stratton MR. cancer vaccine, with ipilimumab to NCT03532217. Mutational signatures: the patterns of treat high-risk renal cell carcinoma. 66. NantBioScience, Inc. QUILT-2.025 somatic mutations hidden in cancer NCT02950766. https://clinicaltrials. NANT neoepitope yeast vaccine (YE- genomes. Curr Opin Genet Dev. gov/ct2/show/NCT02950766. NEO-001): adjuvant immunotherapy 2014;24(100):52-60. 55. Gritstone Oncology, Inc. A study of a using a personalized neoepitope 41. Vormehr M et al. Mutanome directed personalized cancer vaccine targeting yeast-based vaccine to induce T-cell cancer immunotherapy. Curr Opin shared neoantigens. NCT03953235. responses in subjects w/ previously

112 EMJ • September 2020 EMJ treated cancers. NCT03552718. metabolism in cancer immunotherapy. 85. Carreno BM et al. Cancer https://clinicaltrials.gov/ct2/show/ Front Immunol. 2018;9:2029. doi: immunotherapy. A dendritic cell NCT03552718. 10.3389/fimmu.2018.02029. vaccine increases the breadth and diversity of melanoma 67. Johnson LE et al. 76. Cogdill AP et al. Hallmarks of neoantigen-specific T cells. Science. with a prostate cancer xenoantigen response to immune checkpoint 2015;348(6236):803-8. elicits a xenoantigen epitope-specific blockade. Br J Cancer. 2017;117(1):1-7. T-cell response. Oncoimmunology. 86. Johnson LE et al. Pretreatment 2012;1(9):1546-56. 77. Riethmuller G et al. Monoclonal antigen-specific immunity and antibody therapy for resected Dukes' regulation - association with 68. Buhrman JD et al. Improving C colorectal cancer: seven-year subsequent immune response to antigenic peptide vaccines for cancer outcome of a multicenter randomized anti-tumor DNA vaccination. J immunotherapy using a dominant trial. J Clin Oncol. 1998;16(5):1788-94. Immunother Cancer. 2017;5(1):56. tumor-specific T cell receptor. J Biol Chem. 2013;288(46):33213-25. 78. Kim SK et al. Impact of minimal 87. Santegoets SJ et al. T cell profiling tumor burden on antibody response reveals high CD4+CTLA-4 + 69. Blankenstein T et al. The determinants to vaccination. Cancer Immunol T cell frequency as dominant of tumour immunogenicity. Nat Rev Immunother. 2011;60(5):621-7. predictor for survival after prostate Cancer. 2012;12(4):307-13. GVAX/ipilimumab treatment. 79. Schmid P et al. Keynote-522: phase 70. Seliger B et al. Down-regulation of Cancer Immunol Immunother. 3 study of pembrolizumab (pembro) 2013;62(2):245-56. the MHC class I antigen-processing + chemotherapy (chemo) vs placebo machinery after oncogenic (pbo) + chemo as neoadjuvant 88. Schiffman K et al. Delayed type transformation of murine fibroblasts. treatment, followed by pembro vs hypersensitivity response to recall Eur J Immunol. 1998;28(1):122-33. pbo as adjuvant treatment for early antigens does not accurately reflect immune competence in advanced 71. Atkins D et al. MHC class I antigen triple-negative breast cancer (TNBC). stage breast cancer patients. Breast processing pathway defects, ras Presidential Symposium II. ESMO Cancer Res Treat. 2002;74(1):17-23. mutations and disease stage in Congress, 27 September-1 colorectal carcinoma. Int J Cancer. October, 2019. 89. Blank CU et al. CANCER 2004;109(2):265-73. IMMUNOLOGY. The "cancer 80. Starr SP. Immunology update: new immunogram". Science. 72. Rooney MS et al. Molecular and vaccines. FP Essent. 2016;450:28-34. 2016;352(6286):658-60. genetic properties of tumors 81. Petricciani J et al. Analysis of the in associated with local immune 90. van Dijk N et al. The cancer vivo proliferative capacity of a whole cytolytic activity. Cell. 2015; immunogram as a framework cell cancer vaccine. Biologicals. 160(1-2):48-61. for personalized immunotherapy 2016;44(2):60-3. in urothelial cancer. Eur Urol. 73. Le Bourgeois T et al. Targeting T 2019;75(3):435-44. cell metabolism for improvement 82. Ragupathi G et al. Antibody inducing of cancer immunotherapy. Front polyvalent cancer vaccines. Cancer 91. Binnewies M et al. Understanding the Oncol. 2018;8:237. doi: 10.3389/ Treta Res. 2005;123:157-80. tumor immune microenvironment fonc.2018.00237. 83. Gejman RS et al. Rejection of (TIME) for effective therapy. Nat Med. 2018;24(5):541-50. 74. Marijt KA et al. Metabolic stress immunogenic tumor clones is in cancer cells induces immune limited by clonal fraction. eLife. 92. Anderson AC. Tim-3: an emerging escape through a PI3K-dependent 2018;7:e41090. doi: 10.7554/ target in the cancer immunotherapy blockade of IFNγ receptor signaling. eLife.41090. landscape. Cancer Immunol Res. J Immunother Cancer. 2019;7(1):152. 2014;2(5):393-8. 84. Guo Y et al. Neoantigen vaccine doi: 110.1186/s40425-40019- delivery for personalized anticancer 40627-40428. 93. Knee DA et al. Rationale for anti-GITR immunotherapy. Front Immunol. cancer immunotherapy. Eur J Cancer. 75. Roszik J et al. Editorial: targeting 2018;9:1499. 2016;67:1-10.

FOR REPRINT QUERIES PLEASE CONTACT: [email protected]

Creative Commons Attribution-Non Commercial 4.0 September 2020 • EMJ 113