4-Peptide Vaccine of The Future
4-Peptide Vaccine of The Future
4-Peptide Vaccine of The Future
In Brief
Therapeutic peptide-based
vaccination approaches for the
treatment of cancer patients
have shown first glimmers of
success. However, to achieve
broad clinical efficacy and
implement peptide vaccinations
in the standard treatment of
cancer patients future peptide
vaccines need further
optimization in terms of target
antigen selection, adjuvant
choice, vaccination schedules,
delivery routes, biomarkers, and
combinatorial drugs.
Highlights
• Suitable combinations will enhance the efficacy of peptide vaccines in the future
The approach of peptide-based anticancer vaccination the surface of cells on human leukocyte antigen (HLA) mole-
has proven the ability to induce cancer-specific immune cules and are recognized by the T cell receptor of CD4+ and
responses in multiple studies for various cancer entities. CD8+ T cells.
However, clinical responses remain so far limited to single
patients and broad clinical applicability was not achieved.
Therefore, further efforts are required to improve peptide ANTIGEN SELECTION—THE DEVIL IS IN THE DETAILS
vaccination in order to integrate this low-side-effect The most essential aspect for a clinically effective peptide
therapy into the clinical routine of cancer therapy. To vaccination is indeed the selection of optimal antigen targets,
design clinically effective peptide vaccines in the future,
which should exhibit a highly frequent and tumor-exclusive
different issues have to be addressed and optimized
presentation on the surface of tumor cells and must be
comprising antigen target selection as well as choice of
optimal adjuvants and vaccination schedules. Further- capable of recognition by the patients’ T cells. In recent years
more, the combination of peptide-based vaccines with it has become apparent that neoepitopes derived from tumor-
other immuno- and molecular targeted therapies as well specific nonsynonymous somatic mutations are the central
as the development of predictive biomarkers could further specifications of immune checkpoint inhibitor-induced T cell
improve efficacy. In this review, current approaches in the responses (8). This is in line with the correlation of response to
development of peptide-based vaccines and critical im- checkpoint inhibitor therapy with tumor mutational burden
plications for optimal vaccine design are discussed. (9, 10). Owing to their true tumor specificity, neoepitopes are
not affected by central or peripheral tolerance, which renders
them ideal targets. The encouraging potential of neoantigen-
PEPTIDE-BASED VACCINATION
based vaccination approaches was already investigated in
The idea of activating the immune system to combat cancer preclinical and early-phase clinical trials assessing
cells was already documented in ancient Egypt (1) and was neoantigen-based peptide vaccines in several different cancer
then again pursued in the late 19th century by William Coley, entities (11–14). Yet, substantial challenges remain in the
who had been exploiting streptococcus-mediated tumor effective identification of immunogenic and naturally pre-
rejection (2, 3), probably after hearing from similar experiments sented neoepitopes derived from point mutations, insertion–
by Wilhelm Busch (4). Such antigen-independent strategies deletions or frame-shift mutations. Therefore, in recent
build the cornerstone for a broad and promising research area years, several different bioinformatic tools have been devel-
aiming to actively induce antitumor T cell responses using oped to improve the selection and identification of candidate
various different strategies. Today, multiple antitumor immu- neoepitopes for vaccine design. Based on whole-exome
notherapy approaches (5), including immune checkpoint sequencing data algorithms such as NeoPredPipe, MuPeXI,
blockade, CAR T cells, and antibody-based therapies, have pVAC-Seq, and CloudNeo identify patient-individual muta-
found their way into the clinic and even established in the tions, predict the affinity of neoantigen-derived peptides to
routine of anticancer treatment. Therapeutic cancer vaccines, HLA molecules regarding to the individual patient’s HLA al-
such as protein-, peptide-, DNA-, RNA-, and dendritic cell– or lotypes, integrate tumor mutation and expression data, predict
tumor cell–based vaccines (6, 7), are designed to generate the immunogenicity of these peptides, and evaluate their po-
new or amplify preexisting antigen-specific T cell responses tential as T cell epitopes (15–18). Still, this selection and pre-
against malignant cells. Peptide-based vaccines represent a diction process remains challenging especially for HLA class
low-side-effect vaccination approach using synthetic tumor- II–presented neoepitopes recognized by CD4+ T cells (19). In
associated or -specific peptides or peptide combinations addition, the use of mutation-derived vaccine peptides har-
that are designed to induce and activate peptide-specific tu- bors two main issues: the majority of tumor-specific mutations
mor-reactive T cells in vivo. These peptides are presented on are patient-specific and therefore not suitable for broadly
From the 1Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tübingen, Tübingen,
Germany; 2Department of Immunology, Institute for Cell Biology, 3Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany; 4German Cancer Consortium (DKTK) and German Cancer Research
Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
* For correspondence: Juliane S. Walz, juliane.walz@med.uni-tuebingen.de.
applicable immunotherapy, but rather can be used for reviewed journal. Furthermore, comparative transcriptome
personalized approaches. Furthermore, in cancer entities with and proteome expression profiling between tumor and normal
low mutational burden, the role and impact of neoepitope- tissues as provided, for example, by the Genotype-Tissue
directed T cell responses appear to be marginal, as several Expression Consortium (48) and the Human Protein Atlas (49),
studies have demonstrated that the frequency of naturally can help to reconsider vaccine candidates and filter out genes
presented HLA-presented neoepitopes is less than 1% of the and proteins highly expressed in normal tissues. However, the
number of nonsynonymous mutations based on mass above-described distorted correlation of gene expression,
spectrometric analysis (11, 20–22). Mass spectrometry also proteome, and immunopeptidome (21, 32–35) clearly limited
revealed that hotspot sequence regions within individual the informative value of these analysis. Furthermore, it is
proteins generate the majority of HLA-presented peptides in important to note that tumor antigens are not always deter-
contrast to other protein domains that are not efficiently pro- mined at the protein level. The individual peptides can be
cessed for HLA presentation (21, 23, 24), which highlight the clearly tumor-associated, even if the source protein itself is
need for immunopeptidomics to select truly processed and not. This can occur through differential processing, post-
naturally presented peptides and neoepitopes. Therefore, translational modifications, or antigen processing in the tumor
future large-scale immunopeptidomics studies may reveal cell and highlights the need for immunopeptidomic databases
such hotspots in the human proteome leading then to of benign tissues and cells.
improved bioinformatics tools that prioritize and select neo- In addition to the selection of optimal antigens, additional
epitopes based on their location within such hotspot regions. questions rise about the ideal peptide length, the choice of
Besides mutation-derived neoepitopes unmutated tumor- CD4+ versus CD8+ T cell epitopes, as well as the number of
associated self-peptides have gained renewed attention. different peptides within the vaccine cocktail. In recent years it
These unmutated peptides can arise from aberrantly has become evident that the selection of HLA class
expressed or processed antigens in the tumor cells owing to II–restricted antigen targets recognized by CD4+ T cells in
differential tumor-specific gene expression, posttranslational addition to CD8+ T cell–recognized HLA class I–presented
modification, antigen processing, or other tumor-specific peptides is of paramount importance for clinical outcome.
cellular processes (25, 26). For the selection of suitable CD4+ T cells harbor a crucial role for effective antitumor im-
tumor-associated unmutated peptides different strategies munity (50) and could further improve and sustain T cell
have been applied. For several years, the identification of reactivity (51) and tumor clearance (52). CD4+ T helper cells
tumor-associated antigens was based solely on gene induce and intensify more permanent tumor immune control
expression analyses identifying overexpressed proteins, fol- (53), epitope spreading (54), CD8+ T cell expansion and sur-
lowed by in silico predictions of HLA-presented peptides vival (55), as well as tumor immune cell infiltration (56). In
(27–29) with algorithms such as SYFPEITHI (30) and addition, CD4+ T cells also exhibit direct antitumor effector
NetMHCpan (31). Subsequently, the tremendous number of function (57). Therefore, the artificial elongation of tumor-
predicted candidate peptides had to be screened for immu- associated CD8+ Tcell epitopes or the application of multi-
nogenicity by extensive in vitro assays. A major drawback of valent long peptides comprising at the same time CD4+ and
such prediction-based approaches lies in the distorted cor- CD8+ T cell epitopes displays promising attempts to improve
relation of gene expression with HLA-restricted antigen pre- the efficacy and clinical outcome of peptide vaccination
sentation, since the immunopeptidome is an independent (58–63). The enhanced induction of T cell responses with long
complex layer formed by the antigen presentation machinery vaccine peptides could also be explained by the fact that
and therefore does not necessarily mirror the transcriptome or exact epitopes bind directly to HLA class I molecules of any
the proteome (21, 32–35). In contrast to epitope prediction somatic cell and thus also to nonprofessional antigen-
algorithms, the large-scale and in-depth direct identification of presenting cells (APCs) without further processing, which
naturally HLA-presented peptides, known as HLA ligandome could then lead to suboptimal T cell priming and induce T cell
or immunopeptidome, from primary patient samples by mass tolerance (51, 64, 65). Long peptides, on the other hand,
spectrometry (36–44) is the only unbiased approach to require processing and are therefore only presented by pro-
comprehensively identify tumor rejection antigen candidates fessional APCs (66, 67). We therefore believe that HLA class
and validate the natural processing and presentation of these II–restricted target antigens also in terms of elongated CD8+ T
antigens (45). For the identification of tumor-associated cell epitopes are an indispensable component for clinically
unmutated self-peptides not presented on benign cells and effective peptide vaccines. This is further supported by the
tissues, the extensive screening of immunopeptidomes higher promiscuity of HLA class II molecules leading to a not
derived from benign tissues is of paramount importance. The so tight HLA restriction of the presented peptides (68) in
HLA ligand atlas (www.hla-ligand-atlas.org/) is an open- contrast to CD8+ T cell epitopes. This enables the presenta-
source tool providing a comprehensive collection of mass tion of the same CD4+ T cell epitope on different HLA class II
spectrometry–identified HLA-presented peptides on benign molecules and allows the vaccination of all patients with the
tissues (46, 47), which has currently been submitted to a peer- same cocktail, regardless of their HLA allotype.
In contrast to other antigen-specific treatments, e.g., CAR T induction of peptide-specific T cell responses was demon-
cells, a remarkable advantage of peptide-based vaccines is strated against mutated and self-peptides equally (71) as well
the possibility of a simple and cost-effective combination of as short (77) and long peptides (11). However, patient and
several different tumor-associated antigens in a single injec- antigen selection, dosing, vaccine schedule, as well as usage
tion. Multipeptide vaccines could therefore overcome the of adjuvants and combinatorial treatments differ in all of these
issue of antigen loss and reduce the risk of immune escape, studies, which hampers a comprehensive comparison and
which often occurs under therapeutic pressure (69). The conclusion for the development of improved peptide vaccines.
approach of multipeptide vaccination is further supported by The chemical or sequence modification of the individual
the observation that patients with immune responses to mul- vaccine peptides represents an additional option to improve
tiple tumor-associated antigens showed higher levels of dis- the efficiency of the vaccine-induced antitumor responses
ease control and harbor a survival benefit (38, 70). (reviewed in 81, 82). Amino acid substitutions of self-antigens
Furthermore, validation of target antigen expression on could circumvent tolerance against unmutated self-peptides.
patients’ tumor cells prior to the administration of a peptide Altering the amino acid sequence could not only increase
vaccine to a patient could considerably improve the propor- the peptide binding affinity to its respective HLA molecule, but
tion of patients who can benefit from a specific vaccine. also improve its immunogenicity (83–85). Besides the impact
Validation of both unmutated and mutated target antigen on the strength of a T cell response, altered peptide se-
expression can be carried out at different levels such as quences could also modulate the character and polarization of
transcriptome, proteome, or immunopeptidome. For mutated the peptide-specific immune responses (86, 87). Especially in
antigens, the proof of the presence of the somatic mutation by chronic diseases such as cancer or viral infections heteroclitic
whole-exome sequencing or RNA-Seq is required, but the peptides can help to restore T cell function and proliferation
additional evidence of the natural presentation within the (88) and repeal frequently occurring T cell exhaustion caused
immunopeptidome is usually rather difficult for these antigens by antigen persistence (89). However, such changes also
(71). However, for unmutated target antigens the validation of harbor the potential risk of weakening the T cell response or
antigen expression using immunopeptidomic analysis is highly potentially inducing the expansion of T cells with irrelevant or
recommended, especially in contrast to transcriptomic or pro- even autoreactive specificities. Therefore, the careful evalua-
teomic detection of antigen expression, which often shows a tion of altered peptide ligands is of paramount importance.
distorted correlation to HLA antigen presentation (21, 32–35). In addition to the modified amino acid composition, direct
Besides the presentation of the target antigens on the pa- conjugation of ligands of pattern recognition receptors such
tients’ tumor cells, the immunogenicity of the individual vac- as CpG or Pam3CSK4 for Toll-like receptors (TLRs) (90, 91) or
cine peptides is of paramount importance and has to be C-type lectin receptor–specific mannosylation (92) on long
proven in T cell–based assays prior to inclusion of the specific vaccine peptides could enhance antigen uptake and guide
peptide in a vaccine cocktail (72, 73). Generally, two different intracellular trafficking, thereby favoring antigen presentation.
approaches can be utilized to assess T cell responses to a
ADJUVANTS AND DELIVERY MODE—IT IS ALL ABOUT ACTIVATION
specific antigen: 1) in vitro priming of naive T cells or 2) ex vivo
or in vitro stimulation of preexisting memory T cells. For the For the induction of effective T cell responses through
detection of preexisting memory T cell responses patient- peptide-based vaccines the choice of optimal, strong adju-
derived peripheral blood mononuclear cells or tumor- vants or immunostimulators and ideal delivery routes is of
infiltrating lymphocytes are needed since tumor antigen- paramount importance to ensure that the vaccine peptides are
specific T cells are normally not abundantly present in appropriately sensed by and activate the immune system. The
healthy individuals. Depending on the abundance of preex- main focus of peptide vaccination is here to induce a type
isting memory T cells, their detection can be performed 1–polarized, cell-mediated immunity rather than a type
directly ex vivo or after a short in vitro peptide-specific 2–polarized and humoral response (93). For achieving an
amplification with cytokines such as interleukin 2 (IL-2). As optimal T cell–mediated antitumor effect, the activation and
readout different methods can be applied such as enzyme- expansion of antigen-specific T cells is mandatory, which
linked immunospot assay (74), intracellular cytokine staining necessarily requires the three well-known signals of T cell
(75), or multimer staining (76). Such immunogenicity tests receptor stimulation, appropriate costimulation, and specific
should also be performed during a clinical trial to assess the cytokines (94, 95). Therefore, adjuvants need to deliver the
induction of vaccine-induced T cell responses in vaccinated peptide to dendritic cells and activate and mature these APCs
patients. to accomplish a solid T cell response. The purpose of such
In recent years, multiple peptide-based vaccination trials adjuvants thus comprises the protection of the peptide and
have tested different combinations of short (77) versus elon- prevention of immediate degradation, the efficient uptake by
gated peptides (11), of single peptide–based (78) versus APCs, as well as the appropriate and full activation of APCs.
multipeptide vaccines (79), or of mutated (11) versus unmu- Delivery vehicles, which ideally should have a depot effect,
tated (80) peptides in a variety of different cancer entities. The can be composed, for example, of oil depots such as
to vaccination to generate a water-in-oil emulsion. For efficient For the clinical application of immunotherapeutic ap-
uptake, the peptides can also be encapsulated in structures proaches three different study concepts and strategies
such as liposomes (98) and nanoparticles (99) or can be comprising distinct levels of personalization of drug products
covalently conjugated to adjuvants (90, 92). In multipeptide and anticancer therapies have been proposed: 1) stratifica-
vaccination approaches and especially in patient- tion, 2) warehouse-based personalization, and 3) individual-
individualized approaches the covalent linkage of adjuvants ization (114, 115). Each of these three concepts harbors
to each individual and single peptide is cumbersome for several advantages as well as disadvantages that need to be
clinical translation. considered. Stratification-based treatment decisions as the
So far the cytokine granulocyte-macrophage colony basic level of personalization are already standard in clinical
stimulating factor (GM-CSF) that initiates the recruitment, routine, for example in the biomarker-based application of
maturation, and activation of dendritic cells (100) has been targeted therapies for specific mutations (116) or antibody-
one of the most common adjuvants applied in anticancer based immunotherapies (117). Stratification selects suitable
peptide vaccination trials but its adjuvant effect remains patients who will benefit from a specific therapy based on the
weak (71, 101, 102). For activation and maturation of APCs, availability of predefined tumor-associated criteria and will
signaling through TLRs and their ligands is known to induce then all be treated with one invariant drug product. Only
optimal and strong activation. Therefore, potent adjuvants patients harboring the respective tumor feature will receive
often mimic such TLR ligands. Especially TLR4 ligands are the therapy. Thereby, stratification represents an approach
known to enable a potent activation of APCs (103). However, with an enhancement of treatment efficacy and in the same
the prototype among TLR4 ligands—lipopolysaccharide time minimization of side effects. However, for peptide-
(LPS)—is not sustainable for clinical application owing to based vaccination approaches stratification alone seems
substantial toxicity (104, 105). Its chemically detoxified form not suitable owing to the high intraindividual differences
MPL (3-O-desacyl-4’-monophosphoryl lipid A) (106) is an already starting with patient–individual HLA allotypes.
approved adjuvant, for example, in human papillomavirus Therefore, stratification-based peptide vaccine approaches
vaccines and furthermore investigated in different vaccina- focus on very common HLA allotypes such as HLA-A*02 or
tion approaches (107, 108). The most commonly used TLR HLA-A*24 thereby excluding a substantial proportion of pa-
agonist (71) poly-ICLC (Hiltonol) is a polyinosinic- tients (70, 71). Consequently, current vaccine designs are
polycytidylic acid (poly-IC) stabilized by lysine and more and more focusing on warehouse approaches (77, 80,
carboxymethylcellulose (109), which enhances vaccine- 118), patient-individualized concepts (119), or also a com-
induced T cell responses (110) by TLR3 signaling. bination of both (71). The so-called warehouse concept en-
Recently, the novel, water-soluble adjuvant XS15, a synthetic ables the composition of patient-specific drug products
TLR1/2-binding Pam3-Cys-derivate covalently linked to a assembled from a collection of predefined and pre-
single synthetic—nonvaccine—peptide (GDPKHPKSF), was manufactures high-frequent tumor-associated peptides.
described as an effective vaccine adjuvant inducing unpre- Therefore, each peptide within the warehouse is separately
ceded strong and long-lasting CD8+ and CD4+ T cell re- manufactured and depending on the patient’s individual
sponses in first-in-man proof-of-concept experiments (111). characteristics such as HLA allotypes or tumor-presented
The first clinical trials using XS15, including antitumor peptides the peptide vaccine cocktail is then individually
(EudraCT 2020-002367-65) as well as highly relevant anti- assembled using these off-the-shelf peptides. Thus, the
SARS-CoV-2 peptide vaccinations (EudraCT 2020-002502- warehouse concept enables the individualization of vaccine
75; EudraCT 2020-002519-23), will start recruitment within cocktails in a time- and cost-saving manner. For example, for
2020. Since the induction of the antitumor T cell responses a hypothetical warehouse peptide vaccination study covering
and the clinical outcome has so far been unconvincing, it is eight different HLA allotypes with five peptides each and
mandatory to investigate novel adjuvants like XS15 in clinical including 25 patients, who should be vaccinated with a
trials to further increase the strength and unlock the power of multipeptide cocktail comprising, for example, as commonly
peptide-based vaccines. practiced and approved by regulatory authorities (71, 80), 10
Further improvement of imaging technologies to track either peptides, only 40 different peptides have to be produced.
the peptide vaccine at the molecular and (sub)cellular level The number of 40 peptides is calculated by multiplying the
(112) or the peptide-specific T cells in the organism (113) will number of unique peptides per allotype by the number of
further expand our knowledge of antigen delivery, uptake, and different HLA allotypes covered within the respective ware-
processing as well as of antigen-specific T cell routes. This house. A warehouse covering, for example, eight of the
knowledge can then be utilized to further improve peptide- world’s most common HLA class I allotypes could be used to
based vaccination regarding delivery vehicles, adjuvants, treat approximately 90% of a patient cohort, which could be
and administration route.
calculated for individual allotype combinations using the comparison of different vaccination regimens and each clinical
population coverage tool of IEDB (http://tools.iedb.org/ trial uses different vaccination schedules, thus making evalu-
population/) (120, 121). ation extremely difficult.
In contrast, completely individualized peptide-vaccine
concepts are based on the selection and on-demand de COMBINATIONS—IT IS EASIER TOGETHER
novo production of single patient-specific drug products. The
Important considerations for clinically effective peptide
advantage of such approaches is the patient-specific design
vaccination encompass not only the actual design and
of such vaccines individually tailored on the patient’s HLA
formulation of the peptide vaccine itself, but also furthermore
allotypes, single rare mutations, and peptides presented on
require the awareness of effective combinatorial treatment
the individual tumor reaching the maximal drug benefit for
options. Cancer vaccines alone may be effective in cases of
each cancer patient. Personalized peptide vaccines can
early cancer diagnosis or in the setting of minimal residual
therefore comprise patient-tailored neoepitopes based on
disease to prevent relapse or recurrence. However, combi-
individual sequencing data and/or nonmutated tumor-
natorial treatments might enable to successfully treat even
associated peptides based on the patient-individual mass
established cancers as well as to overcome tumor-mediated
spectrometric analysis of tumor-presented peptides. There-
immunosuppression, immune escape mechanisms, impaired
fore, sequencing or mass spectrometry-based identification of
T cell infiltration to the tumor side, or profound immune de-
patient-individual neoepitopes and nonmutated tumor-
fects (126–129). The range of possible combinations is
associated peptides becomes indispensable. The translation
endless, e.g., combinatorial approaches of cancer vaccines
of sequencing data, mass spectrometry–based immuno-
with immune checkpoint inhibitors, chemotherapies, radio-
peptidomics and their combination is already investigated in
therapy, neutralizing antibodies to inhibitory cytokines, small
different clinical trials (71, 80, 122). However, the low-
molecule inhibition of regulatory T cells, or immunomodulatory
throughput sample capacity is still a major issue. Further
drugs have already been investigated in several studies (80,
limitations of completely individualized approaches are the
130–133) with the main goal to optimize the T cell compart-
huge peptide production costs and the limited drug produc-
ment. However, combinatorial cancer therapies might also
tion capacity. The enormous differences in the estimated
negatively impact T cell responses, as shown in a phase III
costs between peptide warehouse approaches and patient-
peptide vaccination study combined with the tyrosine kinase
individualized vaccines arise mainly from the increased
inhibitor sunitinib (134), which failed to confirm the vaccine-
number of different peptides required for patient-individual
induced immune responses and clinical outcome of the cor-
approaches. Compared with the warehouse approach, for
responding phase II trial testing the peptide vaccine without
the hypothetical study with 25 patients and 10 peptides per
sunitinib (70). This calls for the extensive preclinical and clin-
cocktail a fully individual approach would require the pro-
ical analysis of the effect of combinatorial drugs on T cell
duction of in total 250 peptides resulting in more than 5-fold
response.
higher peptide production costs.
In addition, anticancer drugs can also have marked effects
on the immunopeptidome of tumor cells, including HLA sur-
VACCINATION SCHEDULE—THE RIGHT TIME POINT ALSO MATTERS face expression (135), HLA allotype distribution (136), the
presentation of vaccine target peptides, as well as the in-
Besides the selection of the best fitting treatment concept
duction of novel, cryptic, treatment-associated ligands
also the schedule and timing of vaccination represent an
(136, 137). Therefore, it is of paramount importance to char-
essential pillar for clinically effective vaccination. The selection
acterize also the effects of combinatorial drugs not only on the
of the optimal time point for vaccination in the course of
effector cells but also on the antigenic landscape of the target
cancer treatment is extremely important. Peptide-based
cells (138). Furthermore, the induction of novel, treatment-
vaccination approaches should ideally be administered in
associated HLA ligands on cancer cells under specific treat-
the setting of an intact T cell compartment with an optimal
ments could be utilized for the identification of novel suitable
effector to target cell ratio (123). This could be optimally
antigen targets that could be eligible as novel treatment-
achieved in an adjuvant setting after surgery or in first-line
associated targets for combinatorial peptide vaccinations
remission, e.g., minimal residual disease induced by stan-
(137–139).
dard chemotherapy or radiation (124, 125). Furthermore, for
selection of the optimal vaccination time point, potential
concomitant therapies must be taken into account as these BIOMARKERS—IMPORTANT TO KNOW
drugs can influence the outcome and efficacy of peptide The identification of prognostic factors and predictive bio-
vaccination, as discussed below. Furthermore, the vaccination markers is very important to assist the selection of those
schedule, including primary and boost vaccinations, may have patients who are expected to respond well to peptide
an impact on the effectiveness and duration of the antitumor T vaccination, in the best case even before the start of therapy
cell response. However, there is hardly any systematic or at least early after treatment initiation. However, the criteria
for patients selected for peptide vaccination and the selection Thus, multiple epitope vaccine approaches are already
of individual vaccine peptides vary greatly between different applied in the majority of clinical trials. Furthermore, the HLA
clinical trials, including preexisting T cell responses (77), allotype restriction of HLA class I–presented peptides might
expression of target antigens on the patient’s tumor (71, 80) or exclude patients with unmatching HLA class I allotypes, which
on soluble HLA molecules in the plasma (140–142), peptide- could be overcome by using HLA class II–presented promis-
specific preexisting immunoglobulin G responses (77), or in- cuous CD4+ T cell epitopes embedding multiple HLA class I
dividual transcriptome analysis (71). Several studies have re- peptides in the vaccine.
ported that the immunoglobulin G response against the Despite these rather peptide-specific limitations, there is in
vaccine peptides is a potential biomarker for the prediction of our view a major obstacle for the clinical success of all T cell–
overall survival (143, 144). However, the biological role of based immunotherapies including vaccines, which is the
peptide-specific antibodies remains still unknown. Increased limited accessibility of the tumor for cancer-specific T cells.
peptide-specific immunoglobulin G levels might reflect the Especially in solid tumors, T cell recruitment is often impaired
activation of CD4+ T cells (144). Further plasma biomarkers, by an immunosuppressive tumor environment (149). To
which were found to be useful predictive markers for clinical overcome the limited T cell recruitment combinatorial ap-
outcome after peptide vaccination, are the level and integrity proaches with direct or indirect microenvironment modifiers
of circulating cell-free DNA (145), or specific microRNAs (146). such as BRAF, MEK, or PARP inhibitors or VEGF-targeting
Recently, a study also demonstrated that the prevaccination, antibody- or inhibitor-based therapies have already been
but not postvaccination, blood cell composition (high evaluated (150).
lymphocyte cell counts and percentage) and the inflammatory Peptide-based cancer vaccines are in generally well toler-
signature (low levels of CRP, IL-6) are associated with overall ated with only a few side effects including local reaction on the
survival after personalized peptide vaccination (147). The vaccination site (70, 134). In a meta-analysis including 500
expression level of the immune checkpoints PD-1 and Tim-3 patients, only 1.2% of the vaccinated patients suffer from
on T cells has been demonstrated to correlate with overall vaccine-related serious adverse events (151). Augmented
survival in a peptide vaccination trial (148) further highlighting immune responses seem to be involved as both cellular and
the importance of combinatorial approaches with immune humoral responses to the vaccine peptides are boosted in
checkpoint inhibitors. affected patients. Beside allergic reaction against the vaccine,
The identification of biomarkers that can be utilized for the a major potential risk that can be anticipated for patients
selection of patients and predict the response to peptide- treated with peptide-based vaccines is autoimmunity induced
based immunotherapy is both important and attractive. by immune reactions against normal tissues expressing the
Since multiple factors may influence the induction of clinically vaccinated targets at considerable levels. Therefore, a precise
effective anticancer T cell responses after peptide vaccination, characterization of the target expression on normal tissues is
their predictive and prognostic capabilities have to be vali- of paramount importance. However, such autoimmunity
dated by further large-scale, prospective, and randomized events are very rare. In the phase I to III clinical trials of the off-
clinical trials. the-shelf multipeptide vaccine IMA901 for the treatment of
renal cell carcinoma with a total of 302 vaccinated patients
LIMITATIONS AND SIDE EFFECTS OF PEPTIDE-BASED VACCINATIONS (70, 134) as well as in the phase I/II trial of an antiprostate
AND THE WAY OUT—LITTLE BY LITTLE ONE GOES FAR
cancer vaccine treating 19 patients (152) no evidence of
Although the expectations placed in peptide-based vacci- autoimmunity was observed.
nation approaches have not been fulfilled so far and clinical To provide a maximum of safety for vaccinated patients and
success is still limited, more and more ideas and ways to to ensure early notice of side effects, especially allergic re-
overcome these disadvantages are emerging. In this section actions or autoimmunity, risk mitigation measures must be
we will discuss the drawbacks of peptide-based vaccine ap- implemented in each clinical trial. First, patients should be
proaches, take a look at the issue of adverse effects, and point monitored and kept under medical supervision for at least 2 h
out solutions to overcome these disadvantages, which have following each vaccination, including close monitoring of vital
already been partly applied and evaluated in different clinical parameters (pulse, blood pressure, temperature, oxygen
trials. A first limitation is the low immunogenicity of vaccine saturation) and subjective well-being, as allergic reactions
peptides reported in some clinical trials (11), which can be were reported to occur mostly in the first hour after vaccina-
overcome in different ways: optimized preclinical selection of tion (71, 134). In case of severe anaphylactic reactions, stan-
suitable antigens and the characterization of their immuno- dardized medical antiallergic treatment such as antihistamine
genicity, the modification and affinity maturation (81, 82), as should be applied. Furthermore, special emphasis should be
well as the use of improved strong but nontoxic adjuvants as put on any sign of the development of autoimmune disease.
discussed above are the most important points to address this Any unclear inflammation should be followed up closely until
problem. A further disadvantage is a potential immune escape it resolves, and pausing or stopping of vaccinations and
if vaccination is performed only with single target peptides. immunosuppressive treatment should be considered.
23. Müller, M., Gfeller, D., Coukos, G., and Bassani-Sternberg, M. (2017) M., et al. (2018) The natural HLA ligandome of glioblastoma stem-like
“Hotspots” of antigen presentation revealed by human leukocyte anti- cells: antigen discovery for T cell-based immunotherapy. Acta Neuro-
gen ligandomics for neoantigen prioritization. Front. Immunol. 8, 1367 pathol. 135, 923–938
24. Pearson, H., Daouda, T., Granados, D. P., Durette, C., Bonneil, E., Cour- 40. Reustle, A., Di Marco, M., Meyerhoff, C., Nelde, A., Walz, J. S., Winter, S.,
celles, M., Rodenbrock, A., Laverdure, J. P., Côté, C., Mader, S., Kandabarau, S., Büttner, F., Haag, M., Backert, L., Kowalewski, D. J.,
Lemieux, S., Thibault, P., and Perreault, C. (2016) MHC class I-associ- Rausch, S., Hennenlotter, J., Stühler, V., Scharpf, M., et al. (2020)
ated peptides derive from selective regions of the human genome. J. Integrative -omics and HLA-ligandomics analysis to identify novel drug
Clin. Invest. 126, 4690–4701 targets for ccRCC immunotherapy. Genome Med. 12, 32
25. Laumont, C. M., Daouda, T., Laverdure, J. P., Bonneil, É., Caron-Lizotte, 41. Teck, A. T., Urban, S., Quass, P., Nelde, A., Schuster, H., Letsch, A.,
O., Hardy, M. P., Granados, D. P., Durette, C., Lemieux, S., Thibault, P., Busse, A., Walz, J. S., Keilholz, U., and Ochsenreither, S. (2020) Cancer
and Perreault, C. (2016) Global proteogenomic analysis of human MHC testis antigen Cyclin A1 harbors several HLA-A*02:01-restricted T cell
class I-associated peptides derived from non-canonical reading frames. epitopes, which are presented and recognized in vivo. Cancer Immunol.
Nat. Commun. 7, 10238 Immunother. 69, 1217–1227
26. Erhard, F., Halenius, A., Zimmermann, C., L’Hernault, A., Kowalewski, D. 42. Bilich, T., Nelde, A., Bauer, J., Walz, S., Roerden, M., Salih, H. R., Weisel,
J., Weekes, M. P., Stevanović, S., Zimmer, R., and Dölken, L. (2018) K., Besemer, B., Marcu, A., Lübke, M., Schuhmacher, J., Neidert, M. C.,
Improved Ribo-seq enables identification of cryptic translation events. Rammensee, H. G., Stevanović, S., and Walz, J. S. (2020) Mass
Nat. Methods 15, 363–366 spectrometry-based identification of a B-cell maturation antigen-derived
27. Parvizpour, S., Razmara, J., Pourseif, M. M., and Omidi, Y. (2019) In silico T-cell epitope for antigen-specific immunotherapy of multiple myeloma.
design of a triple-negative breast cancer vaccine by targeting cancer Blood Cancer J. 10, 24
testis antigens. Bioimpacts 9, 45–56 43. Heidenreich, F., Rücker-Braun, E., Walz, J. S., Eugster, A., Kühn, D., Dietz,
28. Safavi, A., Kefayat, A., Abiri, A., Mahdevar, E., Behnia, A. H., and Ghah- S., Nelde, A., Tunger, A., Wehner, R., Link, C. S., Middeke, J. M., Stölze,
remani, F. (2019) In silico analysis of transmembrane protein 31 F., Tonn, T., Stevanovic, S., Rammensee, H. G., et al. (2017) Mass
(TMEM31) antigen to design novel multiepitope peptide and DNA cancer spectrometry-based identification of a naturally presented receptor
vaccines against melanoma. Mol. Immunol. 112, 93–102 tyrosine kinase-like orphan receptor 1-derived epitope recognized by
29. Qazilbash, M. H., Wieder, E., Thall, P. F., Wang, X., Rios, R., Lu, S., CD8+ cytotoxic T cells. Haematologica 102, e460–e464
Kanodia, S., Ruisaard, K. E., Giralt, S. A., Estey, E. H., Cortes, J., 44. Walz, S., Stickel, J. S., Kowalewski, D. J., Schuster, H., Weisel, K.,
Komanduri, K. V., Clise-Dwyer, K., Alatrash, G., Ma, Q., et al. (2017) PR1 Backert, L., Kahn, S., Nelde, A., Stroh, T., Handel, M., Kohlbacher, O.,
peptide vaccine induces specific immunity with clinical responses in Kanz, L., Salih, H. R., Rammensee, H.-G., and Stevanović, S. (2015) The
myeloid malignancies. Leukemia 31, 697–704 antigenic landscape of multiple myeloma: mass spectrometry (re)defines
30. Rammensee, H.-G., Bachmann, J., Emmerich, N. P., Bachor, O. A., and targets for T-cell-based immunotherapy. Blood 126, 1203–1213
Stevanovic, S. (1999) SYFPEITHI: database for MHC ligands and pep- 45. Freudenmann, L. K., Marcu, A., and Stevanović, S. (2018) Mapping the
tide motifs. Immunogenetics 50, 213–219 tumour human leukocyte antigen (HLA) ligandome by mass spectrom-
31. Reynisson, B., Alvarez, B., Paul, S., Peters, B., and Nielsen, M. (2020) etry. Immunology 154, 331–345
NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC 46. Marcu, A., Bichmann, L., Kuchenbecker, L., Backert, L., Kowalewski, D. J.,
antigen presentation by concurrent motif deconvolution and integration Freudenmann, L. K., Löffler, M. W., Lübke, M., Walz, J. S., Velz, J.,
of MS MHC eluted ligand data. Nucleic Acids Res. 48, W449–W454 Moch, H., Regli, L., Silginer, M., Weller, M., Schlosser, A., et al. (2019)
32. Weinzierl, A. O., Lemmel, C., Schoor, O., Müller, M., Kruger, T., Wernet, D., The HLA ligand atlas. A resource of natural HLA ligands presented on
Hennenlotter, J., Stenzl, A., Klingel, K., Rammensee, H.-G., and benign tissues. bioRxiv https://doi.org/10.1101/778944
Stevanović, S. (2007) Distorted relation between mRNA copy number 47. Marcu, A., Bichmann, L., Kuchenbecker, L., Backert, L., Kowalewski, D. J.,
and corresponding major histocompatibility complex ligand density on Freudenmann, L.-K., Löffler, M. W., Lübke, M., Walz, J., Velz, J., Moch,
the cell surface. Mol. Cell. Proteomics 6, 102–113 H., Regli, L., Silginer, M., Weller, M., Schlosser, A., et al. (2020) The HLA
33. Fortier, M. H., Caron, E., Hardy, M. P., Voisin, G., Lemieux, S., Perreault, Ligand Atlas: a novel immuno-oncology resource for T-cell antigen
C., and Thibault, P. (2008) The MHC class I peptide repertoire is molded discovery. J. Clin. Oncol. https://doi.org/10.1200/jco.2020.38.15_suppl.
by the transcriptome. J. Exp. Med. 205, 595–610 3128
34. Bassani-Sternberg, M., Pletscher-Frankild, S., Jensen, L. J., and Mann, M. 48. GTEx Consortium. (2013) The genotype-tissue expression (GTEx) project.
(2015) Mass spectrometry of human leukocyte antigen class I pepti- Nat. Genet. 45, 580–585
domes reveals strong effects of protein abundance and turnover on 49. Uhlén, M., Fagerberg, L., Hallström, B. M., Lindskog, C., Oksvold, P.,
antigen presentation. Mol. Cell. Proteomics 14, 658–673 Mardinoglu, A., Sivertsson, Å., Kampf, C., Sjöstedt, E., Asplund, A.,
35. Shraibman, B., Barnea, E., Kadosh, D. M., Haimovich, Y., Slobodin, G., Olsson, I. M., Edlund, K., Lundberg, E., Navani, S., Szigyarto, C. A. K.,
Rosner, I., López-Larrea, C., Hilf, N., Kuttruff, S., Song, C., Britten, C., et al. (2015) Proteomics. Tissue-based map of the human proteome.
Castle, J., Kreiter, S., Frenzel, K., Tatagiba, M., et al. (2019) Identification Science 347, 1260419
of tumor antigens among the HLA peptidomes of glioblastoma tumors 50. Melssen, M., and Slingluff, C. L. (2017) Vaccines targeting helper T cells for
and plasma. Mol. Cell. Proteomics 18, 1255–1268 cancer immunotherapy. Curr. Opin. Immunol. 47, 85–92
36. Bilich, T., Nelde, A., Bichmann, L., Roerden, M., Salih, H. R., Kowalewski, 51. Bijker, M. S., van den Eeden, S. J. F., Franken, K. L., Melief, C. J. M.,
D. J., Schuster, H., Tsou, C. C., Marcu, A., Neidert, M. C., Lübke, M., Offringa, R., and van der Burg, S. H. (2007) CD8+ CTL priming by exact
Rieth, J., Schemionek, M., Brümmendorf, T. H., Vucinic, V., et al. (2019) peptide epitopes in incomplete Freund’s adjuvant induces a vanishing
The HLA ligandome landscape of chronic myeloid leukemia delineates CTL response, whereas long peptides induce sustained CTL reactivity.
novel T-cell epitopes for immunotherapy. Blood 133, 550–565 J. Immunol. 179, 5033–5040
37. Schuster, H., Peper, J. K., Bösmüller, H. C., Röhle, K., Backert, L., Bilich, 52. Zwaveling, S., Mota, S. C. F., Nouta, J., Johnson, M., Lipford, G. B., Off-
T., Ney, B., Löffler, M. W., Kowalewski, D. J., Trautwein, N., Rabsteyn, ringa, R., van der Burg, S. H., and Melief, C. J. M. (2002) Established
A., Engler, T., Braun, S., Haen, S. P., Walz, J. S., et al. (2017) The human papillomavirus type 16-expressing tumors are effectively eradi-
immunopeptidomic landscape of ovarian carcinomas. Proc. Natl. Acad. cated following vaccination with long peptides. J. Immunol. 169, 350–
Sci. U. S. A. 114, E9942–E9951 358
38. Kowalewski, D. J., Schuster, H., Backert, L., Berlin, C., Kahn, S., Kanz, L., 53. Spitzer, M. H., Carmi, Y., Reticker-Flynn, N. E., Kwek, S. S., Madhireddy,
Salih, H. R., Rammensee, H.-G., Stevanović, S., and Stickel, J. S. (2015) D., Martins, M. M., Gherardini, P. F., Prestwood, T. R., Chabon, J.,
HLA ligandome analysis identifies the underlying specificities of spon- Bendall, S. C., Fong, L., Nolan, G. P., and Engleman, E. G. (2017)
taneous antileukemia immune responses in chronic lymphocytic leu- Systemic immunity is required for effective cancer immunotherapy. Cell
kemia (CLL). Proc. Natl. Acad. Sci. U. S. A. 112, E166–E175 168, 487–502.e15
39. Neidert, M. C., Kowalewski, D. J., Silginer, M., Kapolou, K., Backert, L., 54. Hu, Y., Petroni, G. R., Olson, W. C., Czarkowski, A., Smolkin, M. E., Grosh,
Freudenmann, L. K., Peper, J. K., Marcu, A., Wang, S. S., Walz, J. S., W. W., Chianese-Bullock, K. A., and Slingluff, C. L. (2014) Immunologic
Wolpert, F., Rammensee, H.-G., Henschler, R., Lamszus, K., Westphal, hierarchy, class II MHC promiscuity, and epitope spreading of a
melanoma helper peptide vaccine. Cancer Immunol. Immunother. 63, 71. Hilf, N., Kuttruff-Coqui, S., Frenzel, K., Bukur, V., Stevanović, S., Goutte-
779–786 fangeas, C., Platten, M., Tabatabai, G., Dutoit, V., van der Burg, S. H.,
55. Giuntoli, R. L., Lu, J., Kobayashi, H., Kennedy, R., and Celis, E. (2002) Thor Straten, P., Martinez-Ricarte, F., Ponsati, B., Okada, H., Lassen, U.,
Direct costimulation of tumor-reactive CTL by helper T cells potentiate et al. (2019) Actively personalized vaccination trial for newly diagnosed
their proliferation, survival, and effector function. Clin. Cancer Res. 8, glioblastoma. Nature 565, 240–245
922–931 72. Di Marco, M., Peper, J. K., and Rammensee, H.-G. (2017) Identification of
56. Bos, R., and Sherman, L. A. (2010) CD4+ T-cell help in the tumor milieu is immunogenic epitopes by MS/MS. Cancer J. 23, 102–107
required for recruitment and cytolytic function of CD8+ T lymphocytes. 73. Peper, J. K., and Stevanović, S. (2015) A combined approach of human
Cancer Res. 70, 8368–8377 leukocyte antigen ligandomics and immunogenicity analysis to improve
57. Quezada, S. A., Simpson, T. R., Peggs, K. S., Merghoub, T., Vider, J., Fan, peptide-based cancer immunotherapy. Cancer Immunol. Immunother.
X., Blasberg, R., Yagita, H., Muranski, P., Antony, P. A., Restifo, N. P., 64, 1295–1303
and Allison, J. P. (2010) Tumor-reactive CD4+ T cells develop cytotoxic 74. Mashishi, T., and Gray, C. M. (2002) The ELISPOT assay: an easily
activity and eradicate large established melanoma after transfer into transferable methods for measuring cellular responses and identifying T
lymphopenic hosts. J. Exp. Med. 207, 637–650 cell epitopes. Clin. Chem. Lab. Med. 40, 903–910
58. Maslak, P. G., Dao, T., Bernal, Y., Chanel, S. M., Zhang, R., Frattini, M., 75. Freer, G., and Rindi, L. (2013) Intracellular cytokine detection by
Rosenblat, T., Jurcic, J. G., Brentjens, R. J., Arcila, M. E., Rampal, R., fluorescence-activated flow cytometry: basic principles and recent ad-
Park, J. H., Douer, D., Katz, L., Sarlis, N., et al. (2018) Phase 2 trial of a vances. Methods 61, 30–38
multivalent WT1 peptide vaccine (galinpepimut-S) in acute myeloid 76. Klenerman, P., Cerundolo, V., and Dunbar, P. R. (2002) Tracking T cells
leukemia. Blood Adv. 2, 224–234 with tetramers: new tales from new tools. Nat. Rev. Immunol. 2, 263–272
59. Rabu, C., Rangan, L., Florenceau, L., Fortun, A., Charpentier, M., Dupré, 77. Yoshimura, K., Minami, T., Nozawa, M., Kimura, T., Egawa, S., Fujimoto, H.,
E., Paolini, L., Beauvillain, C., Dupel, E., Latouche, J. B., Adotevi, O., Yamada, A., Itoh, K., and Uemura, H. (2016) A phase 2 randomized
Labarrière, N., and Lang, F. (2019) Cancer vaccines: designing artificial controlled trial of personalized peptide vaccine immunotherapy with low-
synthetic long peptides to improve presentation of class I and class II T dose dexamethasone versus dexamethasone alone in chemotherapy-
cell epitopes by dendritic cells. Oncoimmunology 8, e1560919 naive castration-resistant prostate cancer. Eur. Urol. 70, 35–41
60. Sayem, M. A., Tomita, Y., Yuno, A., Hirayama, M., Irie, A., Tsukamoto, H., 78. Shima, H., Tsurita, G., Wada, S., Hirohashi, Y., Yasui, H., Hayashi, H.,
Senju, S., Yuba, E., Yoshikawa, T., Kono, K., Nakatsura, T., and Nishi- Miyakoshi, T., Watanabe, K., Murai, A., Asanuma, H., Tokita, S., Kubo,
mura, Y. (2015) Identification of glypican-3-derived long peptides acti- T., Nakatsugawa, M., Kanaseki, T., Tsukahara, T., et al. (2019) Ran-
vating both CD8+ and CD4+ T cells; prolonged overall survival in cancer domized phase II trial of survivin 2B peptide vaccination for patients with
patients with Th cell response. Oncoimmunology 5, e1062209 HLA-A24-positive pancreatic adenocarcinoma. Cancer Sci. 110, 2378–
61. Pedersen, S. R., Christensen, J. P., Buus, S., Rasmussen, M., Korsholm, 2385
K. S., Nielsen, M., and Claesson, M. H. (2016) Immunogenicity of HLA 79. Ravandi, F., Walter, R. B., and Freeman, S. D. (2018) Evaluating measur-
class I and II double restricted influenza a-derived peptides. PLoS One able residual disease in acute myeloid leukemia. Blood Adv. 2, 1356–
11, e0145629 1366
62. Ohtake, J., Ohkuri, T., Togashi, Y., Kitamura, H., Okuno, K., and Nishi- 80. Bilich, T., Nelde, A., Kowalewski, D. J., Kanz, L., Rammensee, H.-G.,
mura, T. (2014) Identification of novel helper epitope peptides of survivin Stevanović, S., Salih, H. R., and Walz, J. S. (2017) Definition and char-
cancer-associated antigen applicable to developing helper/killer-hybrid acterization of a peptide warehouse for the patient-individualized pep-
epitope long peptide cancer vaccine. Immunol. Lett. 161, 20–30 tide vaccination study (iVAC-L-CLL01) after first line therapy of CLL.
63. Daftarian, P., Mansour, M., Benoit, A. C., Pohajdak, B., Hoskin, D. W., Blood 130(Suppl 1), 5346
Brown, R. G., and Kast, W. M. (2006) Eradication of established HPV 16- 81. Slansky, J. E., and Nakayama, M. (2020) Peptide mimotopes alter T cell
expressing tumors by a single administration of a vaccine composed of function in cancer and autoimmunity. Semin. Immunol. 47, 101395
a liposome-encapsulated CTL-T helper fusion peptide in a water-in-oil 82. Candia, M., Kratzer, B., and Pickl, W. F. (2016) On peptides and altered
emulsion. Vaccine 24, 5235–5244 peptide ligands: from origin, mode of action and design to clinical
64. Toes, R. E. M., Offringa, R., Blom, R. J. J., Melief, C. J. M., and Kast, W. M. application (immunotherapy). Int. Arch. Allergy Immunol. 170, 211–233
(1996) Peptide vaccination can lead to enhanced tumor growth through 83. Li, Y., Huang, Y., Liang, J., Xu, Z., Shen, Y., Zhang, N., Liu, Z., and Zhao, Y.
specific T-cell tolerance induction. Proc. Natl. Acad. Sci. U. S. A. 93, (2013) Immune responses induced in HHD mice by multiepitope HIV
7855–7860 vaccine based on cryptic epitope modification. Mol. Biol. Rep. 40, 2781–
65. Aichele, P., Brduscha-Riem, K., Zinkernagel, R. M., Hengartner, H., and 2787
Pircher, H. (1995) T cell priming versus T cell tolerance induced by 84. Sun, W., Shi, J., Wu, J., Zhang, J., Chen, H., Li, Y., Liu, S., Wu, Y., Tian, Z.,
synthetic peptides. J. Exp. Med. 182, 261–266 Cao, X., and Li, N. (2018) A modified HLA-A*0201-restricted CTL epitope
66. Kast, W. M., Brandt, R. M. P., and Melief, C. J. M. (1993) Strict peptide from human oncoprotein (hPEBP4) induces more efficient antitumor
length is not required for the induction of cytotoxic T lymphocyte- responses. Cell Mol. Immunol. 15, 768–781
mediated antiviral protection by peptide vaccination. Eur. J. Immunol. 85. Zaremba, S., Barzaga, E., Zhu, M., Soares, N., Tsang, K.-Y., and Schlom,
23, 1189–1192 J. (1997) Identification of an enhancer agonist cytotoxic T lymphocyte
67. Bijker, M. S., van den Eeden, S. J. F., Franken, K. L., Melief, C. J. M., van peptide from human carcinoembryonic antigen. Cancer Res. 57, 4570–
der Burg, S. H., and Offringa, R. (2008) Superior induction of anti-tumor 4577
CTL immunity by extended peptide vaccines involves prolonged, DC- 86. Gladney, K. H., Pohling, J., Hollett, N. A., Zipperlen, K., Gallant, M. E., and
focused antigen presentation. Eur. J. Immunol. 38, 1033–1042 Grant, M. D. (2012) Heteroclitic peptides enhance human immunodefi-
68. Southwood, S., Sidney, J., Kondo, A., delGuercio, M. F., Appella, E., ciency virus-specific CD8+ T cell responses. Vaccine 30, 6997–7004
Hoffman, S., Kubo, R. T., Chesnut, R. W., Grey, H. M., and Sette, A. 87. Salazar, E., Zaremba, S., Arlen, P. M., Tsang, K. Y., and Schlom, J. (2000)
(1998) Several common HLA-DR types share largely overlapping pep- Agonist peptide from a cytotoxic T-lymphocyte epitope of human car-
tide binding repertoires. J. Immunol. 160, 3363–3373 cinoembryonic antigen stimulates production of TC1-type cytokines and
69. Grupp, S. A., Kalos, M., Barrett, D., Aplenc, R., Porter, D. L., Rheingold, S. increases tyrosine phosphorylation more efficiently than cognate pep-
R., Teachey, D. T., Chew, A., Hauck, B., Wright, J. F., Milone, M. C., tide. Int. J. Cancer 85, 829–838
Levine, B. L., and June, C. H. (2013) Chimeric antigen receptor-modified 88. Adegoke, A., Gladney, K., Gallant, M., and Grant, M. (2015) Heteroclitic pep-
T cells for acute lymphoid leukemia. N. Engl. J. Med. 368, 1509–1518 tides increase proliferation and reduce evidence of human immunodefi-
70. Walter, S., Weinschenk, T., Stenzl, A., Zdrojowy, R., Pluzanska, A., ciency virus-specific CD8+ T cell dysfunction. Viral Immunol. 28, 455–463
Szczylik, C., Staehler, M., Brugger, W., Dietrich, P. Y., Mendrzyk, R., 89. Wherry, E. J. (2011) T cell exhaustion. Nat. Immunol. 12, 492–499
Hilf, N., Schoor, O., Fritsche, J., Mahr, A., Maurer, D., et al. (2012) 90. Zom, G. G., Welters, M. J. P., Loof, N. M., Goedemans, R., Lougheed, S.,
Multipeptide immune response to cancer vaccine IMA901 after single- Valentijn, R. R. P. M., Zandvliet, M. L., Meeuwenoord, N. J., Melief, C. J.
dose cyclophosphamide associates with longer patient survival. Nat. M., de Gruijl, T. D., Van der Marel, G. A., Filippov, D. V., Ossendorp, F.,
Med. 18, 1254–1261 and Van der Burg S, H. (2016) TLR2 ligand-synthetic long peptide
conjugates effectively stimulate tumor-draining lymph node T cells of 110. Sabbatini, P., Tsuji, T., Ferran, L., Ritter, E., Sedrak, C., Tuballes, K.,
cervical cancer patients. Oncotarget 7, 67087–67100 Jungbluth, A. A., Ritter, G., Aghajanian, C., Bell-McGuinn, K., Hensley,
91. Deres, K., Schild, H., Wiesmüller, K. H., Jung, G., and Rammensee, H.-G. M. L., Konner, J., Tew, W., Spriggs, D. R., Hoffman, E. W., et al. (2012)
(1989) In vivo priming of virus-specific cytotoxic T lymphocytes with Phase I trial of overlapping long peptides from a tumor self-antigen and
synthetic lipopeptide vaccine. Nature 342, 561–564 poly-ICLC shows rapid induction of integrated immune response in
92. Rauen, J., Kreer, C., Paillard, A., van Duikeren, S., Benckhuijsen, W. E., ovarian cancer patients. Clin. Cancer Res. 18, 6497–6508
Camps, M. G., Valentijn, A. R. P. M., Ossendorp, F., Drijfhout, J. W., 111. Rammensee, H.-G., Wiesmüller, K.-H., Chandran, P. A., Zelba, H., Rusch,
Arens, R., and Burgdorf, S. (2014) Enhanced cross-presentation and E., Gouttefangeas, C., Kowalewski, D. J., Di Marco, M., Haen, S. P., Walz,
improved CD8+ T cell responses after mannosylation of synthetic long J. S., Gloria, Y. C., Bödder, J., Schertel, J.-M., Tunger, A., Müller, L., et al.
peptides in mice. PLoS One 9, e103755 (2019) A new synthetic toll-like receptor 1/2 ligand is an efficient adjuvant
93. Vermaelen, K. (2019) Vaccine strategies to improve anticancer cellular for peptide vaccination in a human volunteer. J. Immunother. Cancer 7,
immune responses. Front. Immunol. 10, 8 307
94. Delves, P. J., and Roitt, I. M. (2000) The immune system. First of two parts. 112. van Montfoort, N., Camps, M. G., Khan, S., Filippov, D. V., Weterings, J. J.,
N. Engl. J. Med. 343, 37–49 Griffith, J. M., Geuze, H. J., van Hall, T., Verbeek, J. S., Melief, C. J., and
95. Delves, P. J., and Roitt, I. M. (2000) The immune system. Second of two Ossendorp, F. (2009) Antigen storage compartments in mature dendritic
parts. N. Engl. J. Med. 343, 108–117 cells facilitate prolonged cytotoxic T lymphocyte cross-priming capac-
96. He, X., Abrams, S. I., and Lovell, J. F. (2018) Peptide delivery systems for ity. Proc. Natl. Acad. Sci. U. S. A. 106, 6730–6735
cancer vaccines. Adv. Ther. 1, 1800060 113. Krekorian, M., Fruhwirth, G. O., Srinivas, M., Figdor, C. G., Heskamp, S.,
97. Aucouturier, J., Dupuis, L., Deville, S., Ascarateil, S., and Ganne, V. (2002) Witney, T. H., and Aarntzen, E. H. J. G. (2019) Imaging of T-cells and
Montanide ISA 720 and 51: a new generation of water in oil emulsions as their responses during anti-cancer immunotherapy. Theranostics 9,
adjuvants for human vaccines. Expert Rev. Vaccines 1, 111–118 7924–7947
98. Varypataki, E. M., Benne, N., Bouwstra, J., Jiskoot, W., and Ossendorp, F. 114. Haen, S. P., Löffler, M. W., Rammensee, H. G., and Brossart, P. (2020)
(2017) Efficient eradication of established tumors in mice with cationic Towards new horizons: characterization, classification and implications
liposome-based synthetic long-peptide vaccines. Cancer Immunol. Res. of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 17, 595–610
5, 222–233 115. Britten, C. M., Singh-Jasuja, H., Flamion, B., Hoos, A., Huber, C., Kallen,
99. Zhou, S., Huang, Y., Chen, Y., Liu, S., Xu, M., Jiang, T., Song, Q., Jiang, G., K. J., Khleif, S. N., Kreiter, S., Nielsen, M., Rammensee, H. G., Sahin, U.,
Gu, X., Gao, X., and Chen, J. (2020) Engineering ApoE3-incorporated Hinz, T., and Kalinke, U. (2013) The regulatory landscape for
biomimetic nanoparticle for efficient vaccine delivery to dendritic cells actively personalized cancer immunotherapies. Nat. Biotechnol. 31,
via macropinocytosis to enhance cancer immunotherapy. Biomaterials 880–882
235, 119795 116. Seol, Y. M., Kwon, C. H., Lee, S. J., et al. (2019) A pilot prospective study
100. Mach, N., Gillessen, S., Wilson, S. B., Sheehan, C., Mihm, M., and Dranoff, of refractory solid tumor patients for NGS-based targeted anticancer
G. (2000) Differences in dendritic cells stimulated in vivo by tumors therapy. Transl Oncol. 12, 301–307
engineered to secrete granulocyte-macrophage colony-stimulating 117. Parakh, S., Gan, H. K., Parslow, A. C., Burvenich, I. J. G., Burgess, A. W.,
factor or Flt3-ligand. Cancer Res. 60, 3239–3246 and Scott, A. M. (2017) Evolution of anti-HER2 therapies for cancer
101. Rausch, S., Gouttefangeas, C., Hennenlotter, J., Laske, K., Walter, K., treatment. Cancer Treat. Rev. 59, 1–21
Feyerabend, S., Chandran, P. A., Kruck, S., Singh-Jasuja, H., Frick, A., 118. Narita, Y., Arakawa, Y., Yamasaki, F., Nishikawa, R., Aoki, T., Kanamori,
Kröger, N., Stevanović, S., Stenzl, A., Rammensee, H.-G., and Bedke, J. M., Nagane, M., Kumabe, T., Hirose, Y., Ichikawa, T., Kobayashi, H.,
(2019) Results of a phase 1/2 study in metastatic renal cell carcinoma Fujimaki, T., Goto, H., Takeshima, H., Ueba, T., et al. (2019)
patients treated with a patient-specific adjuvant multi-peptide vaccine A randomized, double-blind, phase III trial of personalized peptide
after resection of metastases. Eur. Urol. Focus 5, 604–607 vaccination for recurrent glioblastoma. Neuro Oncol. 21, 348–359
102. Hoeller, C., Michielin, O., Ascierto, P. A., Szabo, Z., and Blank, C. U. (2016) 119. Rabsteyn, A., Kyzirakos, C., Schroeder, C., Sturm, M., Mohr, C., Matthes,
Systematic review of the use of granulocyte–macrophage colony- J., Feldhahn, M., Casadei, N., Ebinger, M., Stevanovic, S., Bauer, P.,
stimulating factor in patients with advanced melanoma. Cancer Immu- Kohlbacher, O., Gouttefangeas, C., Schaefer, J., Rammensee, H.-G.,
nol. Immunother. 65, 1015–1034 et al. (2019) Abstract B124: personalized peptide vaccination based on
103. Krutzik, S. R., Tan, B., Li, H., Ochoa, M. T., Liu, P. T., Sharfstein, S. E., patient-individual tumor-specific variants induces T-cell responses in
Graeber, T. G., Sieling, P. A., Liu, Y. J., Rea, T. H., Bloom, B. R., and pediatric patients. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR
Modlin, R. L. (2005) TLR activation triggers the rapid differentiation of International Cancer Immunotherapy Conference: Translating Science
monocytes into macrophages and dendritic cells. Nat. Med. 11, 653– into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA):
660 AACR; Cancer Immunol Res
104. Ribi, E., Parker, R., Strain, S. M., Mizuno, Y., Nowotny, A., Von Eschen, K. 120. Bui, H. H., Sidney, J., Dinh, K., Southwood, S., Newman, M. J., and Sette,
B., Cantrell, J. L., McLaughlin, C. A., Hwang, K. M., and Goren, M. B. A. (2006) Predicting population coverage of T-cell epitope-based di-
(1979) Peptides as requirement for immunotherapy of the Guinea-pig agnostics and vaccines. BMC Bioinformatics 7, 153
line-10 tumor with endotoxins. Cancer Immunol. Immunother. 7, 43–58 121. Vita, R., Overton, J. A., Greenbaum, J. A., Ponomarenko, J., Clark, J. D.,
105. Beutler, B., and Rietschel, E. T. (2003) Innate immune sensing and its Cantrell, J. R., Wheeler, D. K., Gabbard, J. L., Hix, D., Sette, A., and
roots: the story of endotoxin. Nat. Rev. Immunol. 3, 169–176 Peters, B. (2015) The immune epitope database (IEDB) 3.0. Nucleic
106. Qureshi, N., Takayama, K., and Ribi, E. (1982) Purification and structural Acids Res. 43, D405–D412
determination of nontoxic lipid A obtained from the lipopolysaccharide 122. Wick, W., Dietrich, P. Y., Kuttruff, S., Hilf, N., Frenzel, K., Admon, A.,
of Salmonella typhimurium. J. Biol. Chem. 257, 11808–11815 van der Burg, S. H., von Deimling, A., Gouttefangeas, C., Kroep, J. R.,
107. Rastakhiz, S., Yazdani, M., Shariat, S., Arab, A., Momtazi-Borojeni, A. A., Martinez-Ricarte, F., Okada, H., Ottensmeier, C., Ponsati, B., Poulsen,
Barati, N., Mansourian, M., Amin, M., Abbasi, A., Saberi, Z., Jalali, S. A., H. S., et al. (2018) GAPVAC-101: first-in-human trial of a highly
Badiee, A., and Jaafari, M. R. (2019) Preparation of nanoliposomes personalized peptide vaccination approach for patients with newly
linked to HER2/neu-derived (P5) peptide containing MPL adjuvant as diagnosed glioblastoma. J. Clin. Oncol. 36, 2000
vaccine against breast cancer. J. Cell Biochem. https://doi.org/10.1002/ 123. Le Dieu, R., Taussig, D. C., Ramsay, A. G., Mitter, R., Miraki-Moud, F.,
jcb.27090 Fatah, R., Lee, A. M., Andrew Lister, T., and Gribben, J. G. (2009) Pe-
108. Smith, M., García-Martínez, E., Pitter, M. R., Fucikova, J., Spisek, R., Zit- ripheral blood T cells in acute myeloid leukemia (AML) patients at
vogel, L., Kroemer, G., and Galluzzi, L. (2018) Trial Watch: toll-like re- diagnosis have abnormal phenotype and genotype and form defective
ceptor agonists in cancer immunotherapy. Oncoimmunology 7, immune synapses with AML blasts. Blood 114, 3909–3916
e1526250 124. Nakata, J., Nakae, Y., Kawakami, M., Morimoto, S., Motooka, D., Hosen,
109. Gouttefangeas, C., and Rammensee, H.-G. (2018) Personalized cancer N., Fujiki, F., Nakajima, H., Hasegawa, K., Nishida, S., Tsuboi, A., Oji, Y.,
vaccines: adjuvants are important, too. Cancer Immunol. Immunother. Oka, Y., Kumanogoh, A., and Sugiyama, H. (2018) Wilms tumour 1
67, 1911–1918 peptide vaccine as a cure-oriented post-chemotherapy strategy for
patients with acute myeloid leukaemia at high risk of relapse. Br. J. 139. Chong, C., Müller, M., Pak, H. S., Harnett, D., Huber, F., Grun, D., Leleu,
Haematol. 182, 287–290 M., Auger, A., Arnaud, M., Stevenson, B. J., Michaux, J., Bilic, I., Hir-
125. Brown, T. A., Mittendorf, E. A., Hale, D. F., Myers, J. W., Peace, K. M., sekorn, A., Calviello, L., Simó-Riudalbas, L., et al. (2020) Integrated
Jackson, D. O., Greene, J. M., Vreeland, T. J., Clifton, G. T., Ardavanis, proteogenomic deep sequencing and analytics accurately identify non-
A., Litton, J. K., Shumway, N. M., Symanowski, J., Murray, J. L., Pon- canonical peptides in tumor immunopeptidomes. Nat. Commun. 11,
niah, S., et al. (2020) Prospective, randomized, single-blinded, multi- 1293
center phase II trial of two HER2 peptide vaccines, GP2 and AE37, in 140. Bassani-Sternberg, M., Barnea, E., Beer, I., Avivi, I., Katz, T., and Admon,
breast cancer patients to prevent recurrence. Breast Cancer Res. Treat. A. (2010) Soluble plasma HLA peptidome as a potential source for
181, 391–401 cancer biomarkers. Proc. Natl. Acad. Sci. U. S. A. 107, 18769–18776
126. Ahmad, M., Rees, R. C., and Ali, S. A. (2004) Escape from immunotherapy: 141. Shraibman, B., Barnea, E., Kadosh, D. M., Haimovich, Y., Slobodin, G.,
possible mechanisms that influence tumor regression/progression. Rosner, I., López-Larrea, C., Hilf, N., Kuttruff, S., Song, C., Britten, C.,
Cancer Immunol. Immunother. 53, 844–854 Castle, J., Kreiter, S., Frenzel, K., Tatagiba, M., et al. (2018) Identification
127. Gao, J., Shi, L. Z., Zhao, H., Chen, J., Xiong, L., He, Q., Chen, T., Roszik, of tumor antigens among the HLA peptidomes of glioblastoma tumors
J., Bernatchez, C., Woodman, S. E., Chen, P. L., Hwu, P., Allison, J. P., and plasma. Mol. Cell. Proteomics 17, 2132–2145
Futreal, A., Wargo, J. A., et al. (2016) Loss of IFN-gamma pathway genes 142. Ritz, D., Gloger, A., Neri, D., and Fugmann, T. (2017) Purification of
in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell soluble HLA class I complexes from human serum or plasma deliver
167, 397–404.e9 high quality immuno peptidomes required for biomarker discovery.
128. Ramsay, A. G., Johnson, A. J., Lee, A. M., Gorgün, G., Le Dieu, R., Blum, Proteomics 17, 10.1002
W., Byrd, J. C., and Gribben, J. G. (2008) Chronic lymphocytic leukemia 143. Noguchi, M., Mine, T., Komatsu, N., Suekane, S., Moriya, F., Matsuoka, K.,
T cells show impaired immunological synapse formation that can be Yutani, S., Shichijo, S., Yamada, A., Toh, U., Kawano, K., Azuma, K.,
reversed with an immunomodulating drug. J. Clin. Invest. 118, 2427– Uemura, H., Okuno, K., Matsumoto, K., et al. (2010) Assessment of
2437 immunological biomarkers in patients with advanced cancer treated by
129. Duan, Q., Zhang, H., Zheng, J., and Zhang, L. (2020) Turning cold into hot: personalized peptide vaccination. Cancer Biol. Ther. 10, 1266–1279
firing up the tumor microenvironment. Trends Cancer 6, 605–618 144. Kanekiyo, S., Hazama, S., Takenouchi, H., Nakajima, M., Shindo, Y.,
130. Khleif, S. (2015) Strategies and designs for combination immune therapy. Matsui, H., Tokumitsu, Y., Tomochika, S., Tsunedomi, R., and Tokuhisa,
J. Transl. Med. 13(Suppl 1), K7 Y. (2018) IgG response to MHC class I epitope peptides is a quantitative
131. MacKay, A., Weir, G., Koblish, H., Leahey, A. V., Kaliaperumal, V., Tram, predictive biomarker in the early course of treatment of colorectal cancer
C., Scherle, P., and Stanford, M. (2018) Combination of a T cell acti- using therapeutic peptides. Oncol. Rep. 39, 2385–2392
vating immunotherapy with immune modulators alters the tumor 145. Kitahara, M., Hazama, S., Tsunedomi, R., Takenouchi, H., Kanekiyo, S.,
microenvironment and promotes more effective tumor control in pre- Inoue, Y., Nakajima, M., Tomochika, S., Tokuhisa, Y., Iida, M., Saka-
clinical models. Cancer Res. 78(13 Supplement), 1761 moto, K., Suzuki, N., Takeda, S., Ueno, T., Yamamoto, S., et al. (2016)
132. Sharabi, A. B., Nirschl, C. J., Kochel, C. M., Nirschl, T. R., Francica, B. J., Prediction of the efficacy of immunotherapy by measuring the integrity
Velarde, E., Deweese, T. L., and Drake, G. C. (2015) Stereotactic radi- of cell-free DNA in plasma in colorectal cancer. Cancer Sci. 107, 1825–
ation therapy augments antigen-specific PD-1–mediated antitumor im- 1829
mune responses via cross-presentation of tumor antigen. Cancer 146. Kijima, T., Hazama, S., Tsunedomi, R., Tanaka, H., Takenouchi, H.,
Immunol. Res. 3, 345–355 Kanekiyo, S., Inoue, Y., Nakashima, M., Iida, M., and Sakamoto, K.
133. Ali, O. A., Lewin, S. A., Dranoff, G., and Mooney, D. J. (2016) Vaccines (2017) MicroRNA-6826 and-6875 in plasma are valuable non-invasive
combined with immune checkpoint antibodies promote cytotoxic T-cell biomarkers that predict the efficacy of vaccine treatment against met-
activity and tumor eradication. Cancer Immunol. Res. 4, 95–100 astatic colorectal cancer. Oncol. Rep. 37, 23–30
134. Rini, B. I., Stenzl, A., Zdrojowy, R., Kogan, M., Shkolnik, M., Oudard, S., 147. Suekane, S., Yutani, S., Yamada, A., Sasada, T., Matsueda, S., Takamori,
Weikert, S., Bracarda, S., Crabb, S. J., Bedke, J., Ludwig, J., Maurer, D., S., Toh, U., Kawano, K., Yoshiyama, K., Sakamoto, S., Sugawara, S.,
Mendrzyk, R., Wagner, C., Mahr, A., et al. (2016) IMA901, a multipeptide Komatsu, N., Yamada, T., Naito, M., Terasaki, M., et al. (2020) Identifi-
cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy cation of biomarkers for personalized peptide vaccination in 2,588
for advanced or metastatic renal cell carcinoma (IMPRINT): a multi- cancer patients. Int. J. Oncol. 56, 1479–1489
centre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 148. Shindo, Y., Hazama, S., Suzuki, N., Iguchi, H., Uesugi, K., Tanaka, H.,
17, 1599–1611 Aruga, A., Hatori, T., Ishizaki, H., Umeda, Y., Fujiwara, T., Ikemoto, T.,
135. Shi, J., Tricot, G. J., Garg, T. K., Malaviarachchi, P. A., Szmania, S. M., Shimada, M., Yoshimatsu, K., Takenouchi, H., et al. (2017) Predictive
Kellum, R. E., Storrie, B., Mulder, A., Shaughnessy, J. D., Jr, Barlogie, B., biomarkers for the efficacy of peptide vaccine treatment: based on the
and van Rhee, F. (2008) Bortezomib down-regulates the cell-surface results of a phase II study on advanced pancreatic cancer. J. Exp. Clin.
expression of HLA class I and enhances natural killer cell-mediated Cancer Res. 36, 36
lysis of myeloma. Blood 111, 1309–1317 149. Murciano-Goroff, Y. R., Warner, A. B., and Wolchok, J. D. (2020) The future
136. Kowalewski, D. J., Walz, S., Backert, L., Schuster, H., Kohlbacher, O., of cancer immunotherapy: microenvironment-targeting combinations.
Weisel, K., Rittig, S. M., Kanz, L., Salih, H. R., Rammensee, H.-G., Cell Res. 30, 507–519
Stevanović, S., and Stickel J, S. (2016) Carfilzomib alters the HLA- 150. Pelster, M. S., and Amaria, R. N. (2019) Combined targeted therapy and
presented peptidome of myeloma cells and impairs presentation of immunotherapy in melanoma: a review of the impact on the tumor
peptides with aromatic C-termini. Blood Cancer J. 6, e411 microenvironment and outcomes of early clinical trials. Ther. Adv. Med.
137. Shraibman, B., Melamed Kadosh, D., Barnea, E., and Admon, A. (2016) Oncol. https://doi.org/10.1177/1758835919830826
HLA peptides derived from tumor antigens induced by inhibition of DNA 151. Yoshida, K., Noguchi, M., Mine, T., Komatsu, N., Yutani, S., Ueno, T.,
methylation for development of drug-facilitated immunotherapy. Mol. Yanagimoto, H., Kawano, K., Itoh, K., and Yamada, A. (2011) Charac-
Cell. Proteomics 15, 3058–3070 teristics of severe adverse events after peptide vaccination for advanced
138. Nelde, A., Kowalewski, D. J., Backert, L., Schuster, H., Werner, J. O., Klein, cancer patients: analysis of 500 cases. Oncol. Rep. 25, 57–62
R., Kohlbacher, O., Kanz, L., Salih, H. R., Rammensee, H. G., Stevanovic, 152. Feyerabend, S., Stevanovic, S., Gouttefangeas, C., Wernet, D., Hennen-
S., and Walz J, S. (2018) HLA ligandome analysis of primary chronic lotter, J., Bedke, J., Dietz, K., Pascolo, S., Kuczyk, M., Rammensee, H.
lymphocytic leukemia (CLL) cells under lenalidomide treatment confirms G., and Stenzl, A. (2009) Novel multi-peptide vaccination in Hla-A2+
the suitability of lenalidomide for combination with T-cell-based immu- hormone sensitive patients with biochemical relapse of prostate can-
notherapy. Oncoimmunology 128, 3234 cer. Prostate 69, 917–927