Sie sind auf Seite 1von 10

Vaccine Reports 6 (2016) 1322

Contents lists available at ScienceDirect

Vaccine Reports
journal homepage: www.elsevier.com/locate/vacrep

Review Article

T-cell epitope mapping for the design of powerful vaccines


Tarek A. Ahmad a,b,, Amrou E. Eweida c, Laila H. El-Sayed b,d
a
Special Projects Department, Bibliotheca Alexandrina, Alexandria, Egypt
b
SeptivaK Research Group, Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
c
Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
d
Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt

a r t i c l e i n f o a b s t r a c t

Article history: Epitope mapping has emerged as a powerful tool to design vaccines. It proved itself a number of times to
Received 5 April 2016 reveal the building blocks of potent immuno-prophylactics. T-cell epitope mapping aims to identify the
Revised 7 July 2016 shortest amino acid sequence of an epitope of a specific antigen that is recognized by CD4+ and/or CD8+
Accepted 13 July 2016
T-cells. T-cell epitopes have the potential to stimulate both long lasting and exclusive cytotoxic immune
response. Therefore, they are crucial for vaccine development against emerging intracellular pathogens
such as Plasmodium malaria, Mycobacterium spp., genital Chlamydia, HIV, HCV as well as cancer cells.
Keywords:
This review documents and discusses the different methods used in T-cell epitope mapping, and the role
Epitope mapping
Vaccines
of each method to identify the potent epitopes in viruses, bacteria, parasites, as well as human diseases.
Cellular response The comment of the article guides the researchers to identify the most suitable mapping techniques for
Molecular immunology their antigens.
T-cells  2016 Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://
creativecommons.org/licenses/by-nc-nd/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2. Methods of T-cell epitope mapping . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.1. MHC Multimers (MM). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.2. Solid phase MHC-peptide complexes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.3. Lymphoproliferation assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.4. Intracytoplasmic cytokine staining (ICS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.5. Cell ELISA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.6. ELISPOT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.7. Activation markers induced on specific T-cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.8. Cell free system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.9. High throughput T-epitope mapping (HTP). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
2.10. Pepscan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
3. Comment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

1. Introduction usually initiate two major immune responses. The B-cells lympho-
cytes engulf the foreign body and differentiated into plasma cells,
When a foreign immunogen is exposed to the immune system, that secrete immunoglobulin antibodies. For this reason this acti-
it is first encountered by the antigen presenting cells (APCs) that vation is referred as B-cell humoral response. Antibodies recognize
foreign molecules called antigens or immunogens and bind to a
Corresponding author at: 34 Memphis St., 21525 Alexandria, Egypt. specific site on the antigen surface called the epitope or the
E-mail addresses: Tarekadnan@yahoo.com, tarek.adnan.ahmad@gmail.com antigenic determinant [1,2]. Unlike B-cells, T-cells do not directly
(T.A. Ahmad). recognize free floating antigens. However, antigens should be first

http://dx.doi.org/10.1016/j.vacrep.2016.07.002
2405-7843/ 2016 Published by Elsevier Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
14 T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322

up-taken by an APC such as B-cells or dendritic cells or macro- and the prevention of re-infection by the same pathogen [23]. In
phages, and presented to the T-cells by the major histocompatibil- addition, it was shown that the integration of the action of cell-
ity complex (MHC). Then the T-cells receptor (TCR) recognizes the mediated immunity effectors such as IFN-c, Natural Killer (NK)
fragments exposed by the APCs and initiates the T-cell response cells, as well as Th1 helper T-cells are required in the control of
[3,4]. the pathogenesis of intracellular microbes such as Rickettsia [24],
The T-cell-mediated immune response depends on four differ- Salmonella [25], Listeria [26], Mycobacteria [27], and Chlamydia
ent types of T-cells lymphocytes. The cytotoxic T- lymphocytes [28,29], as well as allergens [16]. On the other hand, CD8+ T-cells
(CTL), or killer T-cells play a unique role to destroy cells displaying are highly involved in providing immune protection against
foreign motifs on their surfaces. Whereas helper T-cells (HTC), con- respiratory viruses [30], Vaccinia virus [31], Plasmodium spp.
tribute to both the humoral and the cellular immune responses by [3235], Toxoplasma gondii [36], and intracellular bacteria such as
stimulating the proliferation and differentiation of B-cells and Mycobacterium tuberculosis and Listeria monocytogenes [7,37,38].
killer T-cells. In addition, regulatory T-cells function in maintaining Moreover it was noted that, B-cell-deficient mice were able to
tolerance toward self-antigens, playing an important role in sustain complete recovery from genital tract infection with
autoimmune diseases. While, memory T-cells are responsible for Chlamydia trachomatis [39], which ensures the essential role of
maintaining a relatively long lasting immunological memory T-cells in the recovery from some diseases.
towards the same antigen [1,2,5]. Although the development of a T-cell protection is the corner-
The CTL expresses a cluster of differentiation (CD) receptor gly- stone for constructing potent vaccines [5,10,18], more recent
coprotein termed CD8+ on its surface, which recognizes the MHC studies showed that cellular response is not sufficient for the
class I-peptide complex displayed by the APCs. Association of complete irradiation of intracellular pathogens such as Chlamydia
MHC class I-peptide complex with the cytotoxic TCR, and other and M. tuberculosis. A comprehensive immunological study showed
accessory proteins, aids in activating a series of reactions that that long lasting persistence and increased susceptibility of
result in apoptosis of the infected cell by forcing the cell to suicide Chlamydia and Mycobacteria infections appeared in the B-cell
[4,6,7]. On the other hand, the HTC expresses a specific deficient models [38,39]. Therefore, it was found that antibodies,
glycoprotein on its surface termed CD4+ that has the ability to as well, play an indirect role in T-cell activation, thus resulting in
recognize class II MHC-peptide complex. Association of class II early and effective clearance of the pathogen in the process of
MHC-peptide complex with the helper TCR gives signals that a secondary infection. In addition in small pox and Ebola virus [40]
pathogen is invading the cell. The initiation, differentiation and it was found that although the elucidation of potent cellular
regulation of those responses trigger the secretion of small cell- immune response is required for the rapid recovery of infected
signal molecules referred to as cytokines [3,8]. Cytokines (CK) are individuals, the activation of the humoral response is essential
functionally classified into a T-cell helper 1 (Th1) group that for protection [41,42]. Therefore, the novel approach in the design
enhances the cellular immune response such as interferon-c of vaccines is to implement strategies capable of activating both
(IFN-c) and interleukins (IL-12 and IL-18), and a Th2 group that arms of the immune system, the cellular as well as the humoral
enhances the humoral immune response such IL-2, IL-4 and IL-10 immune responses [13]. The different techniques followed to
[9]. Those can be measured to monitor the cellular immune map the T-cell epitope are described hereafter, in order to integrate
response [10]. them to the mapping techniques of the B-cells [14].
The design of vaccines and diagnostics is a recent art that
attracted the attention of scientists worldwide [11]. Epitope
2. Methods of T-cell epitope mapping
mapping is a new branch of immunology which focuses on the
selection of the most potent epitopes that could serve as potential
Several techniques depending on in silico and binding methods
targets for the production of epitope-based immune-preparations.
developed to point the epitopes that activate the cellular response.
Several occasions highlighted the advantages of following the
The following paragraphs discuss those methods and document
route to design vaccines and diagnostics [1215]. T-cell epitope
the majority of the practical trials that involved their use. The
mapping is a term that refers to the process of identifying
T-cell epitope mapping techniques were the corner-stone to reveal
the shortest amino acid sequence of the epitope of a specific anti-
the potent antigenic determinants of many intracellular bacteria,
gen that is recognized by CD4+ and/or CD8+ T-cell receptors, and at
viruses, cancer, autoimmune, allergen and parasites antigens.
the same time has the potential to stimulate a long lasting and a
cytotoxic immune response [16,17]. Therefore, T-cell epitope
mapping is critical for constructing epitope-based vaccines as well 2.1. MHC Multimers (MM)
as potent therapeutics for intracellular pathogens and cancer cells
[9,1719]. Unlike the majority of B-cell epitopes, T-cell epitopes This method depends on synthetic MHC molecules that bind to
are linear protein molecules that consist of 12 to 20 amino acids. the tested peptide and propose it to the TCRs. Hence it isolates and
This fact facilitates their cloning and their synthesis as peptide identifies the bound T-lymphocytes, in case the tested peptide is an
vaccines [16]. Some methods may be used for both T- and B-cell appropriate epitope [43]. The most widely spread multimers, are
epitope mapping such as pepscan, in silico, ELISA and ELISPOT that MHC tetramers composed of four MHC molecules covalently
corresponds to the microarray in B-cell epitoping [14]. Simultane- attached to biotin [44]. The first approach that used this technique
ously, new techniques such as mRNA [20] and random phage allowed the peptide antigen to attach to the tetramer-complex.
display library [21] are under investigation. Further coupling was detected by a suitable fluorescent tag that
The selection of a target epitope that has the ability to induce a has a high affinity to biotin such as streptavidin, then the emitted
protective B-cell dependent immune response is crucial for vaccine fluorescence was measured [45]. Although this method is simple
development [5,7,18], however this will not always lead to the and recoverable as the cells can be reused for flow cytometry stud-
establishment of long lasting protective memory inside the host ies, it has some drawbacks. Since, the antigens had to be known in
[22]. Moreover, in the case of intracellular pathogens and tumors advance using some other methods of epitope mapping, and the
the cellular arm of the immune response managed by T-cell prior knowledge of MHC-peptide interactions is needed in order
activity, is highly involved in the process of fighting the foreign to design suitable tetramer components [46]. This method devel-
bodies as well as inducing a cytotoxic activity [4,23]. The genera- oped to apply a reversible binding between the peptide and the
tion of memory T-cells enables the rapid recovery of the host MM. In this later approach, a peptide that has a UV cleavage site
T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322 15

is left to bind to the MM. Once exposed to the UV light, it dissoci- in identifying the epitopes of the flagella filament protein FliC of
ates from the MM allowing other peptides with the same binding Salmonella typhimurium [60], and the Smith antigen commonly
capacity, and with different binding specificity to bind the MM. detected in Systemic Lupus Erythematosus (SLE) patients [61]
This allows the construction of many MM for T-cell epitope map- when used alone. It proved to be successful as well when combined
ping [47]. It was first thought that this technique is only limited with pepscan and in silico methods in mapping the epitopes of
to few MHC class I alleles due to the limited number of MHC class Babesia bovis parasite [62], and the epitopes of CEA in mice [63].
II molecules created so far. Therefore, it was used for CTL epitopes Together with pepscan it highlighted several epitopes of
detection and not the HTCs [46]. However, more recent studies M. tuberculosis [64,65], two MHC class II epitopes of prostate cancer
addressed the use of this approach for MHC class II tetramers as [66], several epitopes of Coxsackie virus B4 structural proteins [67],
an important method of T-cell epitope mapping [45]. several CD4+ T-cell epitopes of ORF2 and ORF3 proteins of human
MM proved several times to be a successful tool for epitope Hepatitis E virus [68], and to investigate the potency of a peptide
mapping when combined with other mapping techniques. The cancer vaccine [69]. Moreover, with IFN-c ELISPOT assay it was
approach was combined with pepscan and the lymphoproliferation able to identify a novel epitope of the flagella filament protein FliC
assay to map the epitopes of M. tuberculosis [48], or with a in silico- of S. typhimurium [70] or with flow cytometry to highlight a
based method and flow cytometry to identify the potent epitopes potential cancer antigen [71]. In combination with cell-ELISA it
of Chlamydia trachomatis [49]. It was simultaneously combined identified a promising renal cell carcinoma-associated antigen
with pepscan, the flow cytometry, and cell ELISA to highlight the [72], while with pepscan, flow cytometry, and TEPITOPE software
epitopes of Mycobacterium leprae [50] or with pepscan and an it highlighted several CD4+ T-cell epitopes of MAGE-3, a potential
algorithm-based method to screen the epitopes of HIV-1 [51]. It tumor antigen [73]. The lymphoproliferation assay data were
was applied with pepscan and flow cytometry to highlight the consolidated with pepscan and flow cytometry to identify eleven
epitopes of Herpes Simplex Virus (HSV-2) [52], or with ELISPOT potent T-cell epitopes of HIV-1 [74] or with in silico approach to
to identify the epitopes of small pox virus [53]. Moreover, it was identify novel epitopes of peanut allergy [75]. Moreover, as
used with pepscan and IFN-c ELISPOT to investigate the epitopes discussed before the lymphoproliferation assay could be combined
of hexon, an adenoviral capsid protein antigen [54], or with the with the MM approach [48], or the MM approach and the flow
lymphoproliferation approach and the flow cytometry to investi- cytometry method [55].
gate the ability of carcino-embryonic antigen (CEA) to elucidate
potent T-cell response in cancer patients [55]. 2.4. Intracytoplasmic cytokine staining (ICS)

2.2. Solid phase MHC-peptide complexes In the late 1990, ICS or the so-called flow cytometry technique
developed [76] on the principle that stimulated T-lymphocytes
The solid phase MHC-peptides complexes can be created by could be re-activated in the presence of a suitable antigen that is
spotting the MHC class I molecules on a suitable microarray slide, bound to MHC molecule in the presence of large number of APCs.
then allowing it to bind to the peptides that can be recognized by Once T-lymphocytes are re-activated, specific cytokines should
the T-lymphocytes [56]. However, a novel approach developed by be released. A specific protein inhibitor is added to allow retaining
applying an anti-cytokine antibody to the microarray so that the of cytokines within their releasing T-lymphocytes. Later on cells
cytokine secreted by the target T-cell can be captured as it appears are permeabilized using a suitable detergent, stained with fluores-
as a spot and this later can be identified using immune-fluorescent cent antibodies, and the emitted fluorescence is detected [58,77].
techniques [57]. The technique allows the screening of large num- The technique has the ability to identify and to quantify different
ber of T-cell epitopes. Although the technique was used to reveal cytokines using monoclonal antibodies labeled with different col-
the activation of human cluster of differentiation types CD8+ and ors in a single cell, and the ability to test different phenotypic
CD4+ T-cells corresponding to Vaccinia, Epstein-Barr viruses markers on a specific T-cell. However, it has some drawbacks such
(EBV), Influenza, and HIV-1 [57], it is not widely practiced due to as the professional expertise and high cost to use the device and its
the complexity of the preparations and the instrumentation [16]. reagents [18,58].
The ICS proved to be successful in the mapping of the epitopes
2.3. Lymphoproliferation assay of HER-2/neu, a potential tumor antigen [78] when used
exclusively. However, the technique was implemented in many
Lymphocyte-proliferation assay or the so-called 3H-thymidine occasions in combination with other mapping methods. When
uptake assay is used to determine the lymphocytes activation, combined with pepscan, the approach managed to identify the
and hence gives an overall idea on the cell-mediated immune epitopes of human cytomegalovirus (HCMV) [79], Nef protein of
response. In this approach, the proliferation of the T-lymphocytes HIV [80], human herpes virus 8 that cause Kaposis sarcoma [81],
can be determined by the fact that cells undergoing activation P53 cancer antigen [82], parathyroid hormone-related protein
increase their rate of DNA and protein synthesis [16]. The specimen (PTH-rP), a potent cancer antigen [83], and M. tuberculosis
of activated purified peripheral blood mononuclear cells (PBMCs) [8486]. Furthermore, the ICS was implemented with the IFN-c
or T-cells is mixed with the antigen gradient. The 3H-thymidine ELISPOT to identify the epitopes of Nef protein of HIV [87], the
is added after 72 h to 120 h and DNA synthesis is quantified using matrix protein of respiratory syncytial virus (RSV) [88], Salmonella
a gamma counter to calculate the amount of radio-labeled thymi- enterica serovars [89], and dengue virus-2 E domain III [90]. Also
dine integrated into the DNA [16,58]. Although the technique was the technique was combined with cell-ELISA to detect the epitopes
widely practiced due to its sensitivity, it has some drawbacks such of Influenza A/California/7/2009 (pH1N1) [91], and Ebola viruses
as the limited number of well plates (only 96), the use of a radioac- (EBOVs) glycoproteins [40].
tive label, the lack of data on the frequency and secreted cytokines It was combined with pepscan and the IFN-c ELISPOT assay to
obtained, and long culture days (35 days). In addition, unlike the highlight the epitopes of P53 [92], or with IFN-c ELISPOT assay
MM technique, cells cannot be reused for future work. However, and in silico-based method to investigate the epitopes of mice hep-
new procedures appeared to reduce the time of the experiment aranase, a universal tumor associated antigen [93] and of HIV-1
and to depend on safer cheap colorimetric detections [59]. [94], or with ELISA and in silico-based method to screen the
Some mapping protocols followed the lymphoproliferation epitopes of the avian Influenza virus [95]. In addition to that, the
assays to map T-cell epitopes. The approach proved to be successful approach was implemented with pepscan, and in silico-based
16 T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322

approach to identify the epitopes of prostate specific antigen (PSA) allergen [124], and a human inhibitor of apoptosis [125]. However,
[96], and M. tuberculosis [97,98]. Additionally, it was combined the technique is usually combined with pepscan to highlight the
with pepscan, the IFN-c ELISPOT and in silico-based approach to epitopes HIV-1 [126129], the IE1 protein of the HCMV [130],
identify the epitopes of the HIV Nef protein [99], the Human H5N1 vaccination [131], the pneumonia virus of mice glycoprotein
Papilloma virus type 16 (HPV-16) E5 protein [100], and the cancer G [132], the outer surface protein A of Borrelia garinii [133],
testis antigen HOM-TES-14/SCP1 [101]. DNA-binding protein 1 (MDP1) of M. tuberculosis [134], bladder
As mentioned before, the flow cytometry approach was com- cancer [135], prostate stem cell antigen [136], over-expressed
bined with the MM technique [52], the MM approach and a tumor antigen calcium-activated chloride channel 2 [137], The
bioinformatics-based method [49], the MM approach and cell NY-ESO-1 tumor antigen [138], and Plasmodium falciparum
ELISA [50], the lymphoproliferation assay [71,73,74]. The flow circum-sporozoite protein [139]. Moreover, it was used with in
cytometry was integrated with binding methods of B-cell epitope silico-based method to identify the potent epitopes of Ebola virus
mapping to map EBV [102]. [140], Influenza A virus [141], Hepatitis C virus (HCV) [142],
Vaccinia virus [17], M. tuberculosis [143,144], mammaglobin-A, a
2.5. Cell ELISA highly expressed breast cancer tumor antigen [145], cervical
cancer [146], and human heparanase that is normally over-
In ELISA the 96 or 384 or 1536 [58,103,104] wells are coated expressed in the majority of tumors [147]. As highlighted before,
with the first primary anti-cytokine antibody (captured on the the ELISPOT approach could be combined with the MM approach
solid wells), and then the tested antigens and the cell medium [53,54], the lymphoproliferation assay [70], the flow cytometry
are introduced into the wells, followed by the addition of the approach, and the flow cytometry and in silico-based method.
stimulated T-lymphocytes. After an incubation period that ranges Simultaneously, ELISPOT was used in combination with B-cell
between 18 h and 36 h, specific T-cells recognizing a certain anti- epitope mapping to reveal the potent antigenic determinants of
gen will release a specific cytokine that could be captured by the ovarian cancer [148], bovine milk allergen [149], and tau protein
solid phase antibody [58]. The technique could be used for quanti- of the Alzheimers disease [150].
tative as well as qualitative detection of cytokines. It has the
advantage of using frozen PBMC without losing effectiveness 2.7. Activation markers induced on specific T-cells
[18,58]. A major limitation is the inability to define the phenotype
of the T-cells that secrete the cytokine without testing purified CD4 In the previously demonstrated methods, the T-helper (Th)-cell
+ and CD8+ cells [105]. Therefore, the assay cannot estimate the response was assessed by measuring cytokines or MHC-II
frequency of an antigen recognized by a specific T-cell [106]. multimers. Although these techniques provide quantitative and
The ELISA method was practiced to quantify the cellular phenotypic information, they do not give complete results for
response of several pathogens such as HIV [107], Filaria [108], non-Th1 CD4+ T-cell recall antigens such as tetanus, Mycobacteria,
and M. avium [109]. It was combined with pepscan to highlight CMV, HSV-1, Influenza, Candida, and HIV-1. Since tetanus for
the epitopes of the Lck protein kinase, a potential tumor antigen example, yields a vigorous proliferation response that does not
[110], the glycoprotein B homolog of human herpes virus 8 correspond to the cytokines assay measurement [151,152].
[111], and the kallikrein 4, a prostate specific antigen [112]. As Therefore, the activation markers induced-method was applied to
discussed before, the ELISA technique could be combined with allow the identification, counting, and selection of specific CD4+
other mapping protocols, such as the flow cytometry, MM, and and CD8+ for T-lymphocytes recall antigens. The technique is
lymphoproliferation. based on culturing the whole blood cells stimulated with an
antigen or mitogen for 4048 h only. Thereafter, the desired CD
2.6. ELISPOT was measured by surface staining with specific antibodies [151].
The cells retained their viability since the methodology is not
T-cells secrete different cytokines, especially IFN-c, which can dependent on cells permeabilization, and they frequently undergo
be detected as a result of antigenic recognition on antigen present- phenotypic and functional characterization by flow cytometry
ing cells (APCs). The technique is very similar to ELISA, but with [151,153]. It was noticed that the detected CD expression was
slight variations. A ninety-six well plates with polyvinylidene sometimes associated with specific cytokines measurement [153].
fluoride or nitrocellulose membrane lined bottom portion is coated The activation marker technique is simple and time saving
with a primary anti-cytokine antibody [58,77]. The plate is [151,152]. This technique may be also applied for quantitative
incubated as in ELISA and the anti-cytokine antibody captures and qualitative detection of specific CDs [152]. Although, some
the target T-cell cytokine. The primary antibody is conjugated with findings claimed that some CDs are only transiently expressed
a secondary biotinylated antibody that is linked to streptavidin- during the course of Th-lymphocytes activation, intracellular
enzyme conjugate and the suitable substrate [113]. Therefore, stabilizers for CD were applied to overcome this defect [152].
antigen specific T-lymphocytes can be seen as a spot that can be Several markers were proposed for use by this technique such as
counted using a low magnification instrument. The technique is CD25 (IL-2Ra)/CD134 (OX40) assay [151] and CD154 assay [152]
widely practiced since it requires simple devices and it allows for the detection of CD4+ cells, while CD137 (TNF4-1BB) [153]
the use of delicate scanners to detect and count spots [58,114]. was applied for the detection of both CD8+ and CD4+ cells. To
Moreover, it enables the detection of two cytokine that are the best of the authors knowledge no epitope was practically
secreted by the same cell simultaneously using two different described by this method.
fluorochrome-labeled reagents [114]. However, the assay is not
able to determine the T-cell phenotype and cells cannot be reused 2.8. Cell free system
neither studied using flow cytometry [115]. In addition, the spot
may contain many different cells if more than one type of T-cells Hartman et al. used the cell free system (CFS) approach to iden-
binds to the same APC [16]. tify Influenza epitopes. This approach could be briefly summarized
The ELISPOT technique was recognized in several occasions in three main steps: (1) incubating the whole antigens with HLA to
such as the mapping of the epitopes of HPV16 [116], H5N1 allow binding, (2) application of proteases to the antigens/HLA
[53,117], Influenza A [118,119], HIV-1 [120,121], tumor hep- mixture, (3) then using mass spectrometry to sequence HLA bound
aranase [122], humans tuberculosis [123], house dust mite peptides [154]. Although this methods did not find its way for
T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322 17

frequent application since it only detects antigens of known library of peptides from the protein of interest (antigen) to test
structure and it is expensive [155], few researchers used it to their ability to bind to the desired antibody [191]. Pepscan
map the epitope of HIV/HSV as being previously well studied [156]. technique was able to identify B-cell epitopes [9,14], as well as
T-cell epitopes [16].
2.9. High throughput T-epitope mapping (HTP) The pepscan technique proved its success to screen the epitopes
of the hemagglutinin molecule of Influenza A virus [192], JC
These are a group of in silico prediction based on algorithmic polyoma virus [193], and the tumor-associated antigen, a potent
immuno-informatics approaches. They offer the preliminary step antigen that is highly expressed in breast cancer patients [194].
aiming to screen and reduce the number of antigenic peptides The technique was usually used as a preliminary step for T-cell
for further mapping by any of the binding methods previously epitope mapping as mentioned before.
described [157]. Moreover, in silico approaches overcome the
shortage in the number of functional T-lymphocytes available for 3. Comment
laboratory work [19], saves time and effort by focusing only on
targeted antigenic peptides which in turn will reduce the cost of Epitope mapping has emerged as a powerful tool for the design
the overall process of epitope mapping [158], and increases the and production of vaccines. Although the selection of target B-cell
specificity by only screening short panels of antigenic peptides as epitopes is crucial for vaccine development, it does not necessarily
well as using small number of cells [106,159]. The in silico methods lead to the design of potent long lasting or intracellular vaccines.
offer accurate results since the majority of T-cell epitopes are linear T-cell response demonstrated its involvement in eliminating intra-
[160]. Several software showed to perform in silico T-cell epitope cellular pathogens, destroying abnormal cells, and inducing long
prediction [161] such as iVax [162], Tepitope [163], EpiMatrix, term protection. Although T-cell epitope mapping proved its
[164167], SYPPEITHI [168], EpiMer [144], Conservatrix [167], importance for the development of vaccines, its integration with
Rankpep [169], Epi-Assembler and Vaccine-CAD [170], iTopia assay B-cell mapping demonstrated its necessity to control several
platform [171], EpiMatrixs extension JanusMatrix [172], DNAStar pathogens. Unlike B-cell epitopes, the T-cells are linear. This
[173], Proped [174], and PEPFR [175]. However, T-cell epitopes offered the privilege to the design and production of T-cell epitopes
predicted by HTP needs further confirmation by binding ones based vaccines, especially the new trend DNA and recombinant
[158]. preparations. A special concern in research was dedicated to
The approach demonstrated success especially when combined emerging influenza viruses, HIV, HCV, M. tuberculosis, M. avium,
with other binding methods. The success was very obvious in the malarias parasite and cancers.
identification of several potent HIV epitopes as being an obvious Although the majority of T-cell epitope mapping are based on
challenge for vaccinologists due to HLA polymorphism and high binding methods, some are bioinformatics based or integrated
mutation rate [160]. In 1997, De Groot et al. [164] identified 134 with physical techniques. Pepscan was the first method to be intro-
T-cell epitopes of HIV using EpiMatrix/HIV algorithm, then duced for epitope mapping since the mid 1980s, and found several
discovered several other epitopes of HIV-1 using the ELISPOT applications in B-cell epitoping. It was simultaneously used in the
approach and the EpiMatrix algorithm [144], then revealed 31 1990s as a preliminary step to reveal the T-cell target epitopes in
novel epitopes by the same method [165], thereafter identified 5 combination with other binding and bioinformatics methods. In
epitopes that were selected to serve as building blocks for the mid-1990s T-cell epitoping flourished by using the MHC mul-
epitope-driven cross-clade HIV-1 vaccine (the GAIA vaccine) timers. This technique was applied to reveal the T-cell epitopes for
against HIV [166]. In 2005, the results were enriched by combining Mycobacteria, Chlamydia, HIV, HSV, small pox and cancer.
the EpiMatrix with the Epi-Assembler and Vaccine-CAD methods Although it is simple and recoverable, the prior knowledge of the
[170]. Furthermore in 2006, Koita et al. [176] evaluated 21 antigenic structure and its first exclusive use for MHC class I
immunogenic consensus sequence. In 2012, Levitz et al. [177] used epitopes limited its application as a sole ideal method for T-cell
the same technique to identify 11 HLA-A2 epitopes of HIV, epitope mapping. The limited number of tests allowed by MM,
followed by De Groot et al. [178] that marked 27 HLA-A3 epitopes introduced the solid phase MHC-complexes method that depends
of HIV, while Mori et al. [179] used a novel in silico approach to on microarray. However, the limitation of the method to MHC class
identify HLA-restricted epitopes of HIV. I epitopes, and expensive instrumentation restricted its use as well.
Simultaneously, the technique was used alone in mapping the Later on the lympho-proliferation assay appeared. It was simple,
epitopes of HCV and Influenza A virus [180], swine-origin Influenza cheap, and sensitive. However, it was time consuming, lacked the
A (H3N2) variant virus [181,182], 2009-H1N1 and 2004-H5N1 ability to measure CK types and quantity, as well as using risky
Influenza virus [183], Leptospira borgpetersenii [184] and Eimeria radio-labeling for sometimes and cells were not recoverable.
[173]. As discussed before, the EpiMatrix approach was combined However, it developed to safer, cheaper, but less sensitive
with the MM technique [51]. In addition, bioinformatics-based colorimetric-based methods. Despite those primary limitations it
approaches were combined with the lymphoproliferation assay was widely practiced to unravel the epitopes of intracellular bacte-
[62,63,75,185], the flow cytometry approach [9698], the ELISPOT ria, viruses, tumor markers, autoimmune disease, and the peanut
technique, the MM approach and the flow cytometry analysis allergy epitopes. In the late 1990s the flow cytometry method
[49], the flow cytometry and the ELISPOT approaches appeared. It overcame the defects of time consumption, CK quali-
[90,93,94,100,101], the cell-ELISA assay and the flow cytometry tative and quantitative measurement, number of tested samples,
approach [95], as previously mentioned. New approaches based but lacked the advantage of being cost effective. It had unique
on the detection of both B-cell binding techniques, and T-cell advantages of testing different CKs in one sample, and measuring
epitope by in silico methods appeared for Eimeria [173,186], human different phenotypic markers. The ICS advantages allowed the
[187,188] and avian [174] tuberculosis, H1N1[189], and foot- method to map several new viruses like Ebola, avian flu, HPV and
mouth disease virus [190]. CMV. Moreover it was applied to map the epitopes of other
intracellular bacteria, viruses, and tumor markers.
2.10. Pepscan In the beginnings of the new millennium, cell-ELISA adaptation
in T-cell epitope mapping evolved. It was simple, relatively cheap,
In 1984 Mario Geysen, introduced pepscan as a new method for tested large number of samples, saved times, and had the
epitope mapping, where the target antibody is examined against a advantage of measuring CK qualitatively and quantitatively.
18 T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322

Table 1
Comparison between the different methods of T-cell epitope mapping.

Pepscan MM Solid Lympho- ICS (Flow Cell-ELISA ELISPOT Activation CFS In silico (HTP)
phase proliferation cytometry) markers
MHC
Method Idea Screen Signal for Signal for Detect Signal for Signal for a Signal for Marker Identify Algorithms
against a coupling of CK on a PMNCs specific CK specific CK specific detects bound based
protein MHC with microarray activation CKs specific CD epitopes
library TCR by mass-
spec.
Role/Use Preliminary General General General Screen Quantify a CK Screen up Detects Rough Preliminary
several CKs to 2-CKs Th-1 or -2
activity
Markers No No No No Yes No No Yes No No
phenotype
Identify CK No No No No Yes Yes Yes No No No
Quantify CK No No No No Yes Yes Semi No No No
Antigen Structure Known Known No No No No No No No Known
Genetically Yes No No No No No No No No Yes
mapped
Application Number of Large Limited Large Limited Large Large Large Limited Limited Large
samples
Recoverable Yes No No No No No Yes Yes
cells
Combined EL; ICS; EL; EP; EL; EP; ICS; EL; EP; LY; EP; ICS; LY; ICS; LY; ICS EL; EP; ICS;
with other LY; MM. ICS; LY; MM; PC; PP. MM, PC; PP. MM, PC; PP. MM, PC; LY; MM.
T-methods PC; PP. PP.
Previous Bt; Cs; Pa; Bt; Cs; vs. vs. Al; Bt; Cs; Bt; Cs; vs. Bt; Cs; Pa; vs. Al; Bt; Cs; vs. Al; Bt; Cs;
uses vs. Ds; Pa; vs. Pa; vs. Pa; vs.
Combined vs. Al; Cs; Ds Bt; Pa; vs.
with B-
methods
Pros./Cons. Detect B Easy, Complex & Easy & very Fast, test Simple Simple, Simple, Costs Save time
and T- simple, but costs cheap several CK at device, cheap, cheap & commercial and efforts,
epitopes each once, need save time & can kit that but
peptide expertise &is can use measure 2 costs confirmatory
needs a expensive. freezed cells CKs at test needed
MHC once

Key: Al = Allergens; Bt = Bacteria; CD = Cluster of differentiation; CK = Cytokine; Cs = Cancer; Ds = Disease; EL = Cell ELISA; EP = ELISPOT; PC = in silico; LP = Lymphoproliferation;
MHC = Major Histocompatibility Complex; Pa = Parasite; PMNCs = Poly-morpho-nuclear cells; PP = Pepscan; Vs = Virus.

However, it lacked the ability to measure different CKs at the same were performed by in silico and pepscan methods. The general
time. ELISPOT appeared to ameliorate the defects of ELISA and the detection of T-cell response against a specific epitope was per-
expensive ICS. ELISPOT technique has lower price, and was able to formed by the low cost lymphoproliferation assay, while the
measure different CK types in one sample, without losing the screening of cytokine responses was fulfilled by ICS or ELISPOT,
major privileges already owned by ELISA. In 2005, a commercial depending on the available facilities and expertise. The cell-ELISA
method called activation markers was introduced to measure was usually applied when an accurate evaluation of a specific cyto-
the CK quantitatively. Although the idea of the method shows to kine response was necessary to identify, especially when ICS was
be very promising, until now it has no frequent applications. not an introduced method for cost reasons. Since nowadays the
Simultaneously the CFS method did not show any advantages majority of antigens have known structures pepscan and HTP
when compared to other methods such as the immuno- methods are frequently applied to screen the full genomes for T-
bioinformatics ones. cell epitopes in order to save time, control cost and reduce efforts.
Despite the fact that the majority of T-cell epitope mapping As long as the accurate quantification of cytokines is not a neces-
techniques are binding methods, recent advances in bioinformatics sary target in low resource research projects, the urge to use cell-
and reverse vaccinology strategies allowed scientists to implement ELISA and ICS shifted towards the semi-quantitative ELISPOT. This
in silico-based approaches for the large scale investigations of new confirms that the decision to select the best method to be used to
target antigens. Especially, that T-cell epitope are linear, and hence map an antigen is a case matter. It depends on the previous knowl-
accurate results are obtained from HTP methods. These methods edge of the antigens structure, the availability of sophisticated
focus only on promising target epitopes and it is now known that equipment and experienced professionals, the available fund, the
they speed the discovery process by about 1020-fold. Therefore, number of samples, the urgency to get results, and the type of cel-
they were usually introduced as primary step to unravel the lular responses desired from the designed vaccine. All the mapping
potency of novel unknown epitopes, especially those causing out- methods integrated to offer sufficient information to design a
breaks such as HIV, HCV, H1N1, H5N1, H3N2, Influenza A, Ebola, potent vaccine.
and Listeria. Pepscan was used as well to narrow the number of
tests.
Acknowledgments
(Table 1) displays the major criteria that can be used to com-
pare the appropriateness of a method to be used for T-cell epitope
The authors would like to thank Mr. Ahmed Issa and Mr. Derek
mapping. Preliminary screening of antigens of knows structures
Gulbranson for their efforts to edit this review.
T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322 19

References [33] N.S. Butler, N.W. Schmidt, J.T. Harty, Differential effector pathways regulate
memory CD8 T cell immunity against Plasmodium berghei versus P. yoelii
sporozoites, J. Immunol. 184 (2010) 25282538.
[1] P. Cresswell, M.J. Androlewicz, B. Ortmann, Assembly and transport of class I
[34] N.W. Schmidt, N.S. Butler, V.P. Badovinac, J.T. Harty, Extreme CD8 T cell
MHC-peptide complexes, Ciba Found. Symp. 187 (1994) 150162.
requirements for anti-malarial liver-stage immunity following immunization
[2] P. Cresswell, Assembly, transport, and function of MHC class II molecules,
with radiation attenuated sporozoites, PLoS Pathog. 6 (2010) e1000998.
Annu. Rev. Immunol. 12 (1994) 259293.
[35] A. Trimnell, A. Takagi, M. Gupta, T.L. Richie, S.H. Kappe, R. Wang, Genetically
[3] A.M. Lennon-Dumnil, A.H. Bakker, P. Wolf-Bryant, H.L. Ploegh, C.
attenuated parasite vaccines induce contact-dependent CD8+ T cell killing of
Lagaudrire-Gesbert, A closer look at proteolysis and MHC-class-II-
Plasmodium yoelii liver stage-infected hepatocytes, J. Immunol. 183 (2009)
restricted antigen presentation, Curr. Opin. Immunol. 14 (2002) 1521.
58705878.
[4] N. Ismail, J.P. Olano, H.M. Feng, D.H. Walker, Current status of immune
[36] I.A. Khan, K.H. Ely, L.H. Kasper, A purified parasite antigen (p30) mediates CD8
mechanisms of killing of intracellular microorganisms, FEMS Microbiol. Lett.
+ T cell immunity against fatal Toxoplasma gondii infection in mice, J.
207 (2002) 111120.
Immunol. 147 (1991) 35013506.
[5] K. Kedzierska, S.A. Valkenburg, P.C. Doherty, M.P. Davenport, V. Venturi, Use it
[37] J.S. Woodworth, S.M. Behar, Mycobacterium tuberculosis-specific CD8+ T cells
or lose it: establishment and persistence of T cell memory, Front. Immunol. 3
and their role in immunity, Crit. Rev. Immunol. 26 (2006) 317352.
(2012) 111.
[38] J.S. Woodworth, S.M. Fortune, S.M. Behar, Bacterial protein secretion is
[6] H.G. Rammensee, K. Falk, O. Rtzschke, Peptides naturally presented by MHC
required for priming of CD8+ T cells specific for the Mycobacterium
class I molecules, Annu. Rev. Immunol. 11 (1993) 213244.
tuberculosis antigen CFP10, Infect. Immun. 76 (2008) 41994205.
[7] T. Nagata, Y. Koide, Induction of specific CD8 T cells against intracellular
[39] M. Johansson, M. Ward, N. Lycke, B-cell-deficient mice develop complete
bacteria by CD8 T-cell-oriented immunization approaches, J. Biomed.
immune protection against genital tract infection with Chlamydia
Biotechnol. 2010 (2010) 764542.
trachomatis, Immunology 92 (1997) 422428.
[8] R.F. Wang, The role of MHC class II-restricted tumor antigens and CD4+ T cells
[40] S. Wu, T. Yu, X. Song, S. Yi, L. Hou, W. Chen, Prediction and identification of
in antitumor immunity, Trends Immunol. 22 (2001) 269276.
mouse cytotoxic T lymphocyte epitopes in Ebola virus glycoproteins, Virol. J.
[9] P.A. Castric, F.J. Cassels, Peptide epitope mapping in vaccine development:
9 (2012).
introduction, J. Ind. Microbiol. Biotechnol. 19 (1997) 5657.
[41] F. Fenner, D.A. Henderson, I. Arita, Z. Jezek, I.D. Ladnyi, Smallpox and its
[10] S.C. De Rosa, F.X. Lu, J. Yu, S.P. Perfetto, J. Falloon, S. Moser, et al., Vaccination
Eradication, The World Health Organization, Geneva, Switzerland, 1988.
in humans generates broad T cell cytokine responses, J. Immunol. 173 (2004)
[42] D.M. Knipe, P.M. Howley, in: J.J. Esposito (Ed.), Fields Virology, Lippincott
53725380.
Williams & Wilkins, Philadelphia, 2001.
[11] J.M. Gershoni, A. Roitburd-Berman, D.D. Siman-Tov, N.T. Freund, Y. Weiss,
[43] S.R. Hadrup, A.H. Bakker, C.J. Shu, R.S. Andersen, J. van Veluw, P. Hombrink,
Epitope mapping: the first step in developing epitope-based vaccines,
et al., Parallel detection of antigen-specific T-cell responses by
Biodrugs 21 (2007) 145156.
multidimensional encoding of MHC multimers, Nat. Methods 6 (2009) 520
[12] L.M. Childs, E.B. Baskerville, S. Cobey, Trade-offs in antibody repertoires to
526.
complex antigens, Philos. Trans. R. Soc. Lond. B Biol. Sci. 370 (2015).
[44] A.H. Bakker, T.N. Schumacher, MHC multimer technology: current status and
[13] T.A. Ahmad, The development of immunization trials, Edorium J. Biotechnol.
future prospects, Curr. Opin. Immunol. 17 (2005) 428433.
1 (2015) 16.
[45] G.T. Nepom, MHC class II tetramers, J. Immunol. 188 (2012) 24772482.
[14] T.A. Ahmad, A.E. Eweida, S.A. Sheweita, B-cell epitope mapping for the design
[46] V. Cecconi, M. Moro, S. Del Mare, P. Dellabona, G. Casorati, Use of MHC class II
of vaccine and effective diagnostics, Trials Vaccinol. 5 (2016) 7183.
tetramers to investigate CD4+ T cell responses: problems and solutions,
[15] A.S. De Groot, Immunomics: discovering new targets for vaccines and
Cytometry A 73A (2008) 10101018.
therapeutics, Drug Discov. Today 11 (2006) 203209.
[47] W.W. Kwok, J.A. Gebe, A. Liu, S. Agar, N. Ptacek, J. Hammer, et al., Rapid
[16] L. Malherbe, T-cell epitope mapping, Ann. Allergy Asthma Immunol. 103
epitope identification from complex class-II-restricted T-cell antigens, Trends
(2009) 7679.
Immunol. 22 (2001) 583588.
[17] P. Gilchuk, C.T. Spencer, S.B. Conant, T. Hill, J.J. Gray, X. Niu, et al., Discovering
[48] A. Geluk, K.E. van Meijgaarden, K.L. Franken, J.W. Drijfhout, S. DSouza, A.
naturally processed antigenic determinants that confer protective T cell
Necker, et al., Identification of major epitopes of Mycobacterium tuberculosis
immunity, J. Clin. Invest. 123 (2013) 19761987.
AG85B that are recognized by HLA-A0201-restricted CD8+ T cells in HLA-
[18] R.A. Seder, P.A. Darrah, M. Roederer, T-cell quality in memory and protection:
transgenic mice and humans, J. Immunol. 165 (2000) 64636471.
implications for vaccine design, Nat. Rev. Immunol. 8 (2008) 247258.
[49] W. Kuon, H.G. Holzhtter, H. Appel, M. Grolms, S. Kollnberger, A. Traeder,
[19] F. Kern, G. LiPira, J.W. Gratama, F. Manca, M. Roederer, Measuring Ag-specific
et al., Identification of HLA-B27-restricted peptides from the Chlamydia
immune responses: understanding immunopathogenesis and improving
trachomatis proteome with possible relevance to HLA-B27-associated
diagnostics in infectious disease, autoimmunity and cancer, Trends
diseases, J. Immunol. 167 (2001) 47384746.
Immunol. 26 (2005) 477484.
[50] A. Geluk, S.J. van den Eeden, K. Dijkman, L. Wilson, H.J. Kim, K.L. Franken,
[20] J.D. Doucet, D. Gauchat, R. Lapointe, MRNA PCR-based epitope chase method,
et al., ML1419c peptide immunization induces Mycobacterium leprae-specific
Methods Mol. Biol. 969 (2013) 305320.
HLA-A0201-restricted CTL in vivo with potential to kill live Mycobacteria, J.
[21] T. Midoro-Horiuti, R.M. Goldblum, Epitope mapping with random phage
Immunol. 187 (2011) 13931402.
display library, Methods Mol. Biol. 1131 (2014) 477484.
[51] K.B. Bond, B. Sriwanthana, T.W. Hodge, A.S. De Groot, T.D. Mastro, N.L. Young,
[22] P.H. Lambert, M. Liu, C.A. Siegrist, Can successful vaccines teach us
et al., An HLA-directed molecular and bioinformatics approach identifies new
how to induce efficient protective immune responses?, Nat Med. 11 (2005)
HLA-A11 HIV-1 subtype E cytotoxic T lymphocyte epitopes in HIV-1-infected
S54S62.
Thais, AIDS Res. Hum. Retroviruses 17 (2001) 703717.
[23] J.U. Igietseme, F.O. Eko, Q. He, C.M. Black, Antibody regulation of T-cell
[52] E.J. Novak, A.W. Liu, J.A. Gebe, B.A. Falk, G.T. Nepom, D.M. Koelle, et al.,
immunity: implications for vaccine strategies against intracellular pathogens,
Tetramer-guided epitope mapping: rapid identification and characterization
Expert Rev. Vaccines 3 (2004) 2334.
of immunodominant CD4+ T cell epitopes from complex antigens, J. Immunol.
[24] D.H. Walker, J.P. Olano, H.M. Feng, Critical role of cytotoxic T lymphocytes in
166 (2001) 66656670.
immune clearance of rickettsial infection, Infect. Immun. 69 (2001) 1841
[53] M. Terajima, L. Orphin, A.M. Leporati, P. Pazoles, J. Cruz, A.L. Rothman, et al.,
1846.
Vaccinia virus-specific CD8(+) T-cell responses target a group of epitopes
[25] J. Hess, C. Ladel, D. Miko, S.H. Kaufmann, Salmonella typhimurium
without a strong immunodominance hierarchy in humans, Hum. Immunol.
aroA-infection in gene-targeted immunodeficient mice: major role of
69 (2008).
CD4+ TCR-alpha beta cells and IFN-gamma in bacterial clearance
[54] A.M. Leen, A. Christin, M. Khalil, H. Weiss, A.P. Gee, M.K. Brenner, et al.,
independent of intracellular location, J. Immunol. 156 (1996) 33213326.
Identification of hexon-specific CD4 and CD8 T-cell epitopes for vaccine and
[26] E.R. Unanue, Inter-relationship among macrophages, natural killer cells and
immunotherapy, J. Virol. 82 (2008) 546s554s.
neutrophils in early stages of Listeria resistance, Curr. Opin. Immunol. 9
[55] L. Fong, Y. Hou, A. Rivas, C. Benike, A. Yuen, G.A. Fisher, et al., Altered peptide
(1997) 3543.
ligand vaccination with Flt3 ligand expanded dendritic cells for tumor
[27] J.L. Casanova, L. Abel, Genetic dissection of immunity to mycobacteria: the
immunotherapy, Proc. Natl. Acad. Sci. U. S. A. 98 (2001) 88098814.
human model, Annu. Rev. Immunol. 20 (2002) 581620.
[56] M. Schena, R.A. Heller, T.P. Theriault, K. Konrad, E. Lachenmeier, R.W. Davis,
[28] R.P. Morrison, C. HD, Immunity to murine chlamydial genital infection, Infect.
Microarrays: biotechnologys discovery platform for functional genomics,
Immun. 70 (2002) 27412751.
Trends Biotechnol. 16 (1998) 301316.
[29] J.U. Igietseme, F.O. Ekp, B. CM, Contemporary approaches to designing and
[57] J.D. Stone, W.E. Demkowicz Jr., S. LJ, HLA-restricted epitope identification and
evaluating vaccines against Chlamydia, Expert Rev. Vaccines 2 (2003) 129
detection of functional T cell responses by using MHC-peptide and
146.
costimulatory microarrays, Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 3744
[30] J.E. Kohlmeier, D.L. Woodland, Immunity to respiratory viruses, Annu. Rev.
3749.
Immunol. 27 (2009) 6182.
[58] T.M. Clay, A.C. Hobeika, P.J. Mosca, H.K. Lyerly, M.A. Morse, Assays for
[31] J. Goulding, R. Bogue, V. Tahiliani, M. Croft, S. Salek-Ardakani, CD8 T cells are
monitoring cellular immune responses to active immunotherapy of cancer,
essential for recovery from a respiratory vaccinia virus infection, J. Immunol.
Clin. Cancer Res. 7 (2001) 11271135.
189 (2012) 24322440.
[59] G. Repetto, A. del Peso, J.L. Zurita, Neutral red uptake assay for the estimation
[32] J.E. Epstein, K. Tewari, K.E. Lyke, B.K. Sim, P.F. Billingsley, M.B. Laurens, et al.,
of cell viability/cytotoxicity, Nat. Protoc. 3 (2008) 11251131.
Live attenuated malaria vaccine designed to protect through hepatic CD8+ T
[60] M.A. Bergman, L.A. Cummings, R.C. Alaniz, L. Mayeda, I. Fellnerova, B.T.
cell immunity, Science 334 (2011) 475480.
Cookson, CD4+-T-cell responses generated during murine Salmonella enterica
20 T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322

serovar Typhimurium infection are directed towards multiple epitopes within [86] M.G. Chaitra, M.S. Shaila, N.R. Chandra, R. Nayak, HLA-A0201-restricted
the natural antigen FliC, Infect. Immun. 73 (2005) 72267235. cytotoxic T-cell epitopes in three PE/PPE family proteins of Mycobacterium
[61] B.L. Talken, K.R. Schfermeyer, C.W. Bailey, D.R. Lee, R.W. Hoffman, T cell tuberculosis, Scand. J. Immunol. 67 (2008) 411417.
epitope mapping of the Smith antigen reveals that highly conserved Smith [87] R. Draenert, M. Altfeld, C. Brander, N. Basgoz, C. Corcoran, A.G. Wurcel, et al.,
antigen motifs are the dominant target of T cell immunity in systemic lupus Comparison of overlapping peptide sets for detection of antiviral CD8 and
erythematosus, J. Immunol. 167 (2001) 562580. CD4 T cell responses, J. Immunol. Methods 275 (2003) 1929.
[62] R.A. Court, K. Sitte, J.P. Opdebeeck, I.J. East, Mapping the T cell epitopes of the [88] J.A. Rutigliano, M.T. Rock, A.K. Johnson, J.E. Crowe Jr., B.S. Graham,
Babesia bovis antigen 12D3: implications for vaccine design, Parasite Identification of an H-2D(b)-restricted CD8+ cytotoxic T lymphocyte
Immunol. 20 (1998) 18. epitope in the matrix protein of respiratory syncytial virus, Virology 337
[63] H. Kobayashi, R. Omiya, M. Ruiz, E. Huarte, P. Sarobe, J.J. Lasarte, et al., (2005) 335343.
Identification of an antigenic epitope for helper T lymphocytes from [89] M. Maybeno, A. Redeker, S.P. Welten, B. Peters, S.M. Loughhead, S.P.
carcinoembryonic antigen, Clin. Cancer Res. 8 (2002) 32193225. Schoenberger, et al., Polyfunctional CD4+ T cell responses to immuno-
[64] B.Y. Lee, M.A. Horwitz, T-cell epitope mapping of the three most abundant dominant epitopes correlate with disease activity of virulent Salmonella,
extracellular proteins of Mycobacterium tuberculosis in outbred guinea pigs, PLoS One 7 (2012) e43481.
Infect. Immun. 67 (1999) 26652670. [90] S. Li, L. Peng, W. Zhao, H. Zhong, F. Zhang, Z. Yan, et al., Synthetic peptides
[65] P. Launois, R. DeLeys, M.N. Niang, A. Drowart, M. Andrien, P. Dierckx, et al., containing B- and T-cell epitope of dengue virus-2 E domain III provoked
T-cell-epitope mapping of the major secreted mycobacterial antigen Ag85A in B- and T-cell responses, Vaccine 29 (2011) 36953702.
tuberculosis and leprosy, Infect. Immun. 62 (1994) 36793687. [91] L.E. Wagar, L. Rosella, N. Crowcroft, B. Lowcock, P.C. Drohomyrecky, J. Foisy,
[66] D.G. McNeel, L.D. Nguyen, M.L. Disis, Identification of T helper epitopes from et al., Humoral and cell-mediated immunity to pandemic H1N1 influenza in a
prostatic acid phosphatase, Cancer Res. 61 (2001) 51615167. Canadian cohort one year post-pandemic: implications for vaccination, PLoS
[67] J. Marttila, H. Hyty, P. Vilja, T. Hrknen, A. Alho, M. Roivainen, et al., T cell One 6 (2011) e28063.
epitopes in coxsackievirus B4 structural proteins concentrate in regions [92] T.K. Hoffmann, D.J. Loftus, K. Nakano, M.J. Maeurer, K. Chikamatsu, E. Appella,
conserved between enteroviruses, Virology 293 (2002) 217224. et al., The ability of variant peptides to reverse the nonresponsiveness of T
[68] R. Aggarwal, R. Shukla, S. Jameel, S. Agrawal, P. Puri, V.K. Gupta, et al., T-cell lymphocytes to the wild-type sequence p53(264272) epitope, J. Immunol.
epitope mapping of ORF2 and ORF3 proteins of human hepatitis E virus, J. 168 (2002) 13381347.
Viral Hepat. 14 (2007) 283292. [93] X.D. Tang, Y. Wan, L. Chen, T. Chen, S.T. Yu, Z. Xiong, et al., H-2Kb-restricted
[69] K.L. Knutson, K. Schiffman, M.L. Disis, Immunization with a HER-2/neu helper CTL epitopes from mouse heparanase elicit an antitumor immune response
peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients, in vivo, Cancer Res. 68 (2008) 15291537.
J. Clin. Invest. 107 (2001) 477484. [94] M.A. Altfeld, B. Livingston, N. Reshamwala, P.T. Nguyen, M.M. Addo, A. Shea,
[70] S.J. McSorley, B.T. Cookson, M.K. Jenkins, Characterization of CD4+ T cell et al., Identification of novel HLA-A2-restricted human immunodeficiency
responses during natural infection with Salmonella typhimurium, J. Immunol. virus type 1-specific cytotoxic T-lymphocyte epitopes predicted by the HLA-
164 (2000) 986993. A2 supertype peptide-binding motif, J. Virol. 75 (2001) 13011311.
[71] J. Schmitz, E. Reali, J.W. Hodge, A. Patel, G. Davis, J. Schlom, et al., [95] Y. Hou, Y. Guo, C. Wu, N. Shen, Y. Jiang, J. Wang, Prediction and identification
Identification of an interferon-gamma-inducible carcinoembryonic antigen of T Cell epitopes in the H5N1 influenza virus nucleoprotein in chicken, PLoS
(CEA) CD8(+) T-cell epitope, which mediates tumor killing in CEA transgenic One 7 (2012) e39344.
mice, Cancer Res. 62 (2002) 50585064. [96] J. Lu, E. Celis, Recognition of prostate tumor cells by cytotoxic T lymphocytes
[72] J.L. Vissers, I.J. De Vries, L.P. Engelen, N.M. Scharenborg, J. Molkenboer, C.G. specific for prostate-specific membrane antigen, Cancer Res. 62 (2002) 5807
Figdor, et al., Renal cell carcinoma-associated antigen G250 encodes a 5812.
naturally processed epitope presented by human leukocyte antigen-DR [97] L.X. Wang, T. Nagata, K. Tsujimura, M. Uchijima, S. Seto, Y. Koide,
molecules to CD4(+) T lymphocytes, Int. J. Cancer 100 (2002) 441444. Identification of HLA-DR4-restricted T-cell epitope on MPT51 protein, a
[73] G. Consogno, S. Manici, V. Facchinetti, A. Bachi, J. Hammer, B.M. Conti-Fine, major secreted protein derived from Mycobacterium tuberculosis using MPT51
et al., Identification of immunodominant regions among promiscuous HLA- overlapping peptides screening and DNA vaccination, Vaccine 28 (2010)
DR-restricted CD4+ T-cell epitopes on the tumor antigen MAGE-3, Blood 101 20262031.
(2003) 10381044. [98] T. Aoshi, T. Nagata, M. Suzuki, M. Uchijima, D. Hashimoto, A. Rafiei, et al.,
[74] C.C. Wilson, B. Palmer, S. Southwood, J. Sidney, Y. Higashimoto, E. Appella, Identification of an HLA-A0201-restricted T-cell epitope on the MPT51
et al., Identification and antigenicity of broadly cross-reactive and conserved protein, a major secreted protein derived from Mycobacterium tuberculosis,
human immunodeficiency virus type 1-derived helper T-lymphocyte by MPT51 overlapping peptide screening, Infect. Immun. 76 (2008) 1565
epitopes, J. Virol. 75 (2001) 41954207. 1571.
[75] M. Pascal, G.N. Konstantinou, M. Masilamani, J. Lieberman, H.A. Sampson, In [99] H. Gahery, S. Figueiredo, C. Texier, S. Pouvelle-Moratille, L. Ourth, C. Igea,
silico prediction of Ara h 2 T cell epitopes in peanut-allergic children, Clin. et al., HLA-DR-restricted peptides identified in the Nef protein can induce HIV
Exp. Allergy 43 (2013) 116127. type 1-specific IL-2/IFN-gamma-secreting CD4+ and CD4+/CD8+ T cells in
[76] S.L. Waldrop, C.J. Pitcher, D.M. Peterson, V.C. Maino, L.J. Picker, Determination humans after lipopeptide vaccination, AIDS Res. Hum. Retroviruses 23 (2007)
of antigen-specific memory/effector CD4+ T cell frequencies by flow 427437.
cytometry: evidence for a novel, antigen-specific homeostatic mechanism [100] D.W. Liu, Y.C. Yang, H.F. Lin, M.F. Lin, Y.W. Cheng, C.C. Chu, et al., Cytotoxic
in HIV-associated immunodeficiency, J. Clin. Invest. 99 (1997) 17391750. T-lymphocyte responses to human papillomavirus type 16 E5 and E7
[77] A. Letsch, C. Scheibenbogen, Quantification and characterization of specific proteins and HLA-A0201-restricted T-cell peptides in cervical cancer
T-cells by antigen-specific cytokine production using ELISPOT assay or patients, J. Virol. 81 (2007) 28692879.
intracellular cytokine staining, Methods 31 (2003) 143149. [101] F. Neumann, C. Wagner, K. Preuss, B. Kubuscho, C. Schormann, S. Stevanovic,
[78] A. Scardino, P. Alves, D.A. Gross, S. Tourdot, S. Graff-Dubois, E. Angevin, et al., et al., Identification of an epitope derived from the cancer testis antigen
Identification of HER-2/neu immunogenic epitopes presented by renal cell HOM-TES-14/SCP1 and presented by dendritic cells to circulating CD4 T cells,
carcinoma and other human epithelial tumors, Eur. J. Immunol. 31 (2001) Blood 106 (2006) 31053113.
32613270. [102] D. Cossu, G. Mameli, G. Galleri, E. Cocco, S. Masala, J. Frau, et al., Human
[79] F. Kern, I.P. Surel, N. Faulhaber, C. Frmmel, J. Schneider-Mergener, C. interferon regulatory factor 5 homologous epitopes of Epstein-Barr virus and
Schnemann, et al., Target structures of the CD8(+)-T-cell response to human Mycobacterium avium subsp. paratuberculosis induce a specific humoral and
cytomegalovirus: the 72-kilodalton major immediate-early protein revisited, cellular immune response in multiple sclerosis patients, Mult. Scler. 21
J. Virol. 73 (1999) 81798184. (2015) 984995.
[80] V. Pancr, B. Georges, G. Angyalosi, F. Castelli, A. Delanoye, M. Delacre, et al., [103] G. Li Pira, F. Ivaldi, C. Dentone, E. Righi, V. Del Bono, C. Viscoli, et al.,
Novel promiscuous HLA-DQ HIV Nef peptide that induces IFN-gamma- Evaluation of antigen-specific T-cell responses with a miniaturized and
producing memory CD4+ T cells, Clin. Exp. Immunol. 129 (2002) 429437. automated method, Clin. Vaccine Immunol. 15 (2008) 18111818.
[81] F. Micheletti, P. Monini, C. Fortini, P. Rimessi, M. Bazzaro, M. Andreoni, et al., [104] D.M. McKinney, R. Skvoretz, M. Qin, G. Ishioka, A. Sette, Characterization of an
Identification of cytotoxic T lymphocyte epitopes of human herpesvirus 8, in situ IFN-gamma ELISA assay which is able to detect specific peptide
Immunology 106 (2002) 395403. responses from freshly isolated splenocytes induced by DNA minigene
[82] Y. Umano, T. Tsunoda, H. Tanaka, K. Matsuda, H. Yamaue, H. Tanimura, immunization, J. Immunol. Methods 237 (2000) 105117.
Generation of cytotoxic T cell responses to an HLA-A24 restricted epitope [105] G. Li Pira, F. Ivaldi, L. Bottone, F. Manca, High throughput functional
peptide derived from wild-type p53, Br. J. Cancer 84 (2001) 10521057. microdissection of pathogen-specific T-cell immunity using antigen and
[83] G. Francini, A. Scardino, K. Kosmatopoulos, F.A. Lemonnier, G. Campoccia, M. lymphocyte arrays, J. Immunol. Methods 326 (2007) 2232.
Sabatino, et al., High-affinity HLA-A()02.01 peptides from parathyroid [106] G. Li Pira, F. Ivaldi, P. Moretti, F. Manca, High throughput T epitope mapping
hormone-related protein generate in vitro and in vivo antitumor CTL and vaccine development, J. Biomed. Biotechnol. 2010 (2010) 325720.
response without autoimmune side effects, J. Immunol. 169 (2002) 4840 [107] N. Gohain, W.D. Tolbert, P. Acharya, L. Yu, T. Liu, P. Zhao, et al., Cocrystal
4849. structures of antibody N60i3 and antibody JR4 in complex with gp120
[84] N. Caccamo, S. Milano, C. Di Sano, D. Cigna, J. Ivanyi, A.M. Krensky, et al., define more cluster A epitopes involved in effective antibody-dependent
Identification of epitopes of Mycobacterium tuberculosis 16-kDa protein effector function against HIV-1, J. Virol. 89 (2015) 88408854.
recognized by human leukocyte antigen-A0201 CD8(+) T lymphocytes, J. [108] A. Ramanathan, C. Immanuel, D.N. Rao, P. Kaliraj, Dissecting the immune
Infect. Dis. 186 (2002) 991998. response elicited by WbALT-2, ALT MAP in clinical populations and mouse
[85] A. Rafiei, Y. Kiutade, Identification of Mycobacterium tuberculosis CTL epitopes model: a prophylactic measure against lymphatic filariasis, Lymphat. Res.
restricted by HLA-A0201 in HHD mice, Iran. Biomed. J. 11 (2007) 2331. Biol. 13 (2015) 120125.
T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322 21

[109] D. Cossu, S. Masala, J. Frau, G. Mameli, M.G. Marrosu, E. Cocco, et al., Antigenic [131] J.W. Zinckgraf, M. Sposato, V. Zielinski, D. Powell, J.J. Treanor, E. von Hofe,
epitopes of MAP2694 homologous to T-cell receptor gamma-chain are highly Identification of HLA class II H5N1 hemagglutinin epitopes following
recognized in multiple sclerosis Sardinian patients, Mol. Immunol. 57 (2014) subvirion influenza A (H5N1) vaccination, Vaccine 27 (2009) 53935401.
138140. [132] E.A. Claassen, G.M. van Bleek, Z.S. Rychnavska, R.J. de Groot, E.J. Hensen, E.J.
[110] N. Harashima, K. Tanaka, T. Sasatomi, K. Shimizu, Y. Miyagi, A. Yamada, et al., Tijhaar, et al., Identification of a CD4 T cell epitope in the pneumonia virus of
Recognition of the Lck tyrosine kinase as a tumor antigen by cytotoxic T mice glycoprotein and characterization of its role in protective immunity,
lymphocytes of cancer patients with distant metastases, Eur. J. Immunol. 31 Virology 368 (2007) 1725.
(2001) 323332. [133] M. Widhe, C. Ekerfelt, S. Jarefors, B.H. Skogman, E.M. Peterson, S. Bergstrm,
[111] Q.J. Wang, X.L. Huang, G. Rappocciolo, F.J. Jenkins, W.H. Hildebrand, Z. Fan, et al., T-cell epitope mapping of the Borrelia garinii outer surface protein A in
et al., Identification of an HLA A0201-restricted CD8(+) T-cell epitope for the lyme neuroborreliosis, Scand. J. Immunol. 70 (2009) 141148.
glycoprotein B homolog of human herpesvirus 8, Blood 99 (2002) 3360 [134] D. Suzuki, T. Nagata, G. Eweda, S. Matsumoto, M. Matsumoto, K. Tsujimura,
3366. et al., Characterization of murine T-cell epitopes on mycobacterial
[112] J.A. Hural, R.S. Friedman, A. McNabb, S.S. Steen, R.A. Henderson, M. Kalos, DNA-binding protein 1 (MDP1) using DNA vaccination, Vaccine 28 (2010)
Identification of naturally processed CD4 T cell epitopes from the prostate- 20202025.
specific antigen kallikrein 4 using peptide-based in vitro stimulation, J. [135] E. Ferris, F. Connan, F. Pags, J. Gaston, A.M. Hagnr, A. Vieillefond, et al.,
Immunol. 169 (2002) 557565. Identification of p53 peptides recognized by CD8(+) T lymphocytes from
[113] C. Czerkinsky, G. Andersson, H.P. Ekre, L.A. Nilsson, L. Klareskog, O. patients with bladder cancer, Hum. Immunol. 62 (2001) 791798.
Ouchterlony, Reverse ELISPOT assay for clonal analysis of cytokine [136] A. Kiessling, M. Schmitz, S. Stevanovic, B. Weigle, K. Hlig, M. Fssel, et al.,
production. I. Enumeration of gamma-interferon-secreting cells, J. Prostate stem cell antigen: identification of immunogenic peptides and
Immunol. Methods 110 (1988) 2936. assessment of reactive CD8+ T cells in prostate cancer patients, Int. J. Cancer
[114] A. Gazagne, E. Claret, J. Wijdenes, H. Yssel, F. Bousquet, E. Levy, et al., A 102 (2002) 390397.
Fluorospot assay to detect single T lymphocytes simultaneously producing [137] R. Konopitzky, U. Knig, R.G. Meyer, W. Sommergruber, T. Wlfel, T.
multiple cytokines, J. Immunol. Methods 283 (2003) 9198. Schweighoffer, Identification of HLA-A0201-restricted T cell epitopes
[115] J.D. Altman, P.A. Moss, P.J. Goulder, D.H. Barouch, M.G. McHeyzer-Williams, J. derived from the novel overexpressed tumor antigen calcium-activated
I. Bell, et al., Phenotypic analysis of antigen-specific T lymphocytes, Science chloride channel 2, J. Immunol. 169 (2002) 540547.
274 (1996) 9496. [138] H.M. Zarour, B. Maillere, V. Brusic, K. Coval, E. Williams, S. Pouvelle-Moratille,
[116] S.H. van der Burg, M.E. Ressing, K.M. Kwappenberg, A. de Jong, K. Straathof, J. et al., NY-ESO-1 119143 is a promiscuous major histocompatibility complex
de Jong, et al., Natural T-helper immunity against human papillomavirus class II T-helper epitope recognized by Th1- and Th2-type tumor-reactive
type 16 (HPV16) E7-derived peptide epitopes in patients with HPV16- CD4+ T cells, Cancer Res. 62 (2002) 213218.
positive cervical lesions: identification of 3 human leukocyte antigen [139] M. Sedegah, Y. Kim, H. Ganeshan, J. Huang, M. Belmonte, E. Abot, et al.,
class II-restricted epitopes, Int. J. Cancer 91 (2001) 612618. Identification of minimal human MHC-restricted CD8+ T-cell epitopes within
[117] M. Terajima, J. Cruz, G. Raines, E.D. Kilpatrick, J.S. Kennedy, A.L. Rothman, the Plasmodium falciparum circumsporozoite protein (CSP), Malar. J. 185
et al., Quantitation of CD8+ T cell responses to newly identified HLA-A0201- (2013), http://dx.doi.org/10.1186/1475-2875-1112-1185.
restricted T cell epitopes conserved among vaccinia and variola (smallpox) [140] K. Sundar, A. Boesen, R. Coico, Computational prediction and identification of
viruses, J. Exp. Med. 197 (2003) 927932. HLA-A2.1-specific Ebola virus CTL epitopes, Virology 360 (2007) 257263.
[118] P.T. Tan, A.T. Heiny, O. Miotto, J. Salmon, E.T. Marques, F. Lemonnier, et al., [141] M. Wang, M.V. Larsen, M. Nielsen, M. Harndahl, S. Justesen, M.H. Dziegiel,
Conservation and diversity of influenza A H1N1 HLA-restricted T cell epitope et al., HLA class I binding 9mer peptides from influenza A virus induce CD4 T
candidates for epitope-based vaccines, PLoS One 5 (2010) e8754. 8710.1371/ cell responses, PLoS One 5 (2010) e10533.
journal.pone.0008754. [142] Z. Guo, H. Zhang, H. Rao, D. Jiang, X. Cong, B. Feng, et al., DCs pulsed with
[119] P.A. Gillis, N. Hernandez-Alvarado, J.S. Gnanandarajah, F. Wussow, D.J. novel HLA-A2-restricted CTL epitopes against hepatitis C virus induced a
Diamond, M.R. Schleiss, Development of a novel, guinea pig-specific IFN- broadly reactive anti-HCV-specific T lymphocyte response, PLoS One 7 (2012)
gamma ELISPOT assay and characterization of guinea pig cytomegalovirus e38390.
GP83-specific cellular immune responses following immunization with a [143] M. Wang, S.T. Tang, A. Stryhn, S. Justesen, M.V. Larsen, M.H. Dziegiel, et al.,
modified vaccinia virus Ankara (MVA)-vectored GP83 vaccine, Vaccine 32 Identification of MHC class II restricted T-cell-mediated reactivity against
(2014) 39633970. MHC class I binding Mycobacterium tuberculosis peptides, Immunology 132
[120] C.A. Semeniuk, R.E. Capina, M.G. Mendoza, J. Kimani, T.B. Ball, M. Luo, et al., (2011) 482491.
Identification and characterization of HLA-A0301 epitopes in HIV-1 gag [144] A.S. De Groot, A. Bosma, N. Chinai, J. Frost, B.M. Jesdale, M.A. Gonzalez, et al.,
proteins using a novel approach, J. Immunol. Methods 352 (2010) 118125. From genome to vaccine: in silico predictions, ex vivo verification, Vaccine 19
[121] B. Ondondo, S. Abdul-Jawad, A. Bridgeman, T. Hanke, Characterization of (2001) 43854395.
T-cell responses to conserved regions of the HIV-1 proteome in BALB/c mice, [145] A. Jaramillo, K. Majumder, P.P. Manna, T.P. Fleming, G. Doherty, J.F. Dipersio,
Clin. Vaccine Immunol. 21 (2014) 15651572. et al., Identification of HLA-A3-restricted CD8+ T cell epitopes derived from
[122] J. Zhang, J. Yang, D. Fan, H. Tao, H. Wang, T. Yu, Peptide FLNPDVLDI of mammaglobin-A, a tumor-associated antigen of human breast cancer, Int. J.
heparanase is a novel HLA-A2-restricted CTL epitope and elicits potent Cancer 102 (2002) 499506.
immunological antitumor effects in vitro with an 8-branched design, Oncol. [146] A. Kather, A. Ferrara, M. Nonn, M. Schinz, J. Nieland, A. Schneider, et al.,
Rep. 29 (2013) 19551961. Identification of a naturally processed HLA-A0201 HPV18 E7 T cell epitope
[123] K. Horvati, S. Bosze, H.P. Gideon, B. Bacsa, T.G. Szabo, R. Goliath, et al., by tumor cell mediated in vitro vaccination, Int. J. Cancer 104 (2003) 345353.
Population tailored modification of tuberculosis specific interferon-gamma [147] T. Chen, X.D. Tang, Y. Wan, L. Chen, S.T. Yu, Z. Xiong, et al., HLA-A2-restricted
release assay, J. Infect. 72 (2016) 179188. cytotoxic T lymphocyte epitopes from human heparanase as novel targets for
[124] D. Hinz, C. Oseroff, J. Pham, J. Sidney, B. Peters, A. Sette, Definition of a pool of broad-spectrum tumor immunotherapy, Neoplasia 10 (2008) 977986.
epitopes that recapitulates the T cell reactivity against major house dust mite [148] S.D. Xiang, Q. Gao, K.L. Wilson, A. Heyerick, M. Plebanski, Mapping T and B
allergens, Clin. Exp. Allergy 45 (2015) 16011612. cell epitopes in sperm protein 17 to support the development of an ovarian
[125] M.H. Andersen, L.O. Pedersen, B. Capeller, E.B. Brcker, J.C. Becker, Thor cancer vaccine, Vaccine 33 (2015) 59505959.
Straten P, Spontaneous cytotoxic T-cell responses against survivin-derived [149] X. Li, S. Yuan, S. He, J. Gao, H. Chen, Identification and characterization of the
MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer antigenic site (epitope) on bovine beta-lactoglobulin: common residues in
patients, Cancer Res. 61 (2001) 59645968. linear and conformational epitopes, J. Sci. Food Agric. 95 (2015) 29162923.
[126] V. Novitsky, N. Rybak, M.F. McLane, P. Gilbert, P. Chigwedere, I. Klein, et al., [150] M.L. Selenica, H. Davtyan, S.B. Housley, L.J. Blair, A. Gillies, B.A. Nordhues,
Identification of human immunodeficiency virus type 1 subtype C Gag-, Tat-, et al., Epitope analysis following active immunization with tau proteins
Rev-, and Nef-specific elispot-based cytotoxic T-lymphocyte responses for reveals immunogens implicated in tau pathogenesis, J. Neuroinflammation
AIDS vaccine design, J. Virol. 75 (2001) 92109228. 11 (2014) 152.
[127] J.R. Currier, M. deSouza, P. Chanbancherd, W. Bernstein, D.L. Birx, J.H. Cox, [151] J.J. Zaunders, M.L. Munier, N. Seddiki, S. Pett, S. Ip, M. Bailey, et al., High levels
Comprehensive screening for human immunodeficiency virus type 1 of human antigen-specific CD4+ T cells in peripheral blood revealed by
subtype-specific CD8 cytotoxic T lymphocytes and definition of degenerate stimulated coexpression of CD25 and CD134 (OX40), J. Immunol. 183 (2009)
epitopes restricted by HLA-A0207 and -C(W)0304 alleles, J. Virol. 76 (2002) 28272836.
49714986. [152] M. Frentsch, O. Arbach, D. Kirchhoff, B. Moewes, M. Worm, M. Rothe, et al.,
[128] N. Frahm, B.T. Korber, C.M. Adams, J.J. Szinger, R. Draenert, M.M. Addo, et al., Direct access to CD4+ T cells specific for defined antigens according to CD154
Consistent cytotoxic-T-lymphocyte targeting of immunodominant regions in expression, Nat. Med. 11 (2005) 11181124.
human immunodeficiency virus across multiple ethnicities, J. Virol. 78 (2004) [153] N.H.R. Litjens, E.A. de Wit, C.C. Baan, M.G.H. Betjes, Activation-induced CD137
21002187. is a fast assay for the identification and multi-parameter flow cytometric
[129] J.R. Currier, U. Visawapoka, S. Tovanabutra, C.J. Mason, D.L. Birx, F.E. analysis of alloreactive t cells, Clin. Exp. Immunol. 174 (2013) 179191.
McCutchan, et al., CTL epitope distribution patterns in the Gag and Nef [154] I.Z. Hartman, A. Kim, R.J. Cotter, K. Walter, S.K. Dalai, T. Boronina, et al., A
proteins of HIV-1 from subtype A infected subjects in Kenya: use of multiple reductionist cell-free major histocompatibility complex class II antigen
peptide sets increases the detectable breadth of the CTL response, BMC processing system identifies immunodominant epitopes, Nat. Med. 16
Immunol. 7 (2006) 8. (2010) 13331340.
[130] N. Frankenberg, S. Pepperl-Klindworth, R.G. Meyer, B. Plachter, Identification [155] T.J. Messitt, F. Terry, L. Moise, W. Martin, A.S. De Groot, A comparison of two
of a conserved HLA-A2-restricted decapeptide from the IE1 protein (pUL123) methods for T cell epitope mapping: cell free in vitro versus
of human cytomegalovirus, Virology 295 (2002) 202216. immunoinformatics, Immunome Res. 7 (2011) e6.
22 T.A. Ahmad et al. / Vaccine Reports 6 (2016) 1322

[156] M. Marcilla, I. Alvarez, A. Ramos-Fernandez, M. Lombardia, A. Paradela, J.P. [177] L. Levitz, O.A. Koita, K. Sangare, M.T. Ardito, C.M. Boyle, J. Rozehnal, et al.,
Albar, Comparative analysis of the endogenous peptidomes displayed by Conservation of HIV-1 T cell epitopes across time and clades: validation of
HLA-B27 and mamu-B08: Two MHC class I alleles associated with elite immunogenic HLA-A2 epitopes selected for the GAIA HIV vaccine, Vaccine 30
control of HIV/SIV infection, J. Proteome Res. 15 (2016) 10591069. (2012) 75477560.
[157] M.N. Davies, D.R. Flower, Harnessing bioinformatics to discover new [178] A.S. De Groot, L. Levitz, M.T. Ardito, G. Skowron, K.H. Mayer, S. Buus, et al.,
vaccines, Drug Discov. Today 12 (2007) 389395. Further progress on defining highly conserved immunogenic epitopes for a
[158] A. Patronov, I. Doytchinova, T-cell epitope vaccine design by global HIV vaccine: HLA-A3-restricted GAIA vaccine epitopes, Hum. Vaccin.
immunoinformatics, Open Biol. 3 (2013) 120139. Immunother. 8 (2012) 9871000.
[159] A.S. De Groot, V. Nene, N.R. Hegde, S. Srikumaran, J. Rayner, W. Martin, T cell [179] M. Mori, K. Matsuki, T. Maekawa, M. Tanaka, B. Sriwanthana, M. Yokoyama,
epitope identification for bovine vaccines: an epitope mapping method for et al., Development of a novel in silico docking simulation model for the fine
BoLA A-11, Int. J. Parasitol. 33 (2003) 641653. HIV-1 cytotoxic T lymphocyte epitope mapping, PLoS One 7 (2012) e41703.
[160] W. Martin, H. Sbai, A.S. De Groot, Bioinformatics tools for identifying [180] C.M. Diez-Rivero, P.A. Reche, CD8 T cell epitope distribution in viruses reveals
class I-restricted epitopes, Methods 29 (2003) 289298. patterns of protein biosynthesis, PLoS One 7 (2012) e43674. 43610.41371/
[161] R. Vita, J.A. Overton, J.A. Greenbaum, J. Ponomarenko, J.D. Clark, J.R. Cantrell, journal.pone.0043674.
et al., The immune epitope database (IEDB) 3.0, Nucleic Acids Res. 43 (2015) [181] V.R. Duvvuri, A. Marchand-Austin, A. Eshaghi, S.N. Patel, D.E. Low, J.B.
D405D412. Gubbay, T. Potential, Cell epitopes within swine-origin triple reassortant
[162] L. Moise, A. Gutierrez, F. Kibria, R. Martin, R. Tassone, R. Liu, et al., IVAX: An influenza A (H3N2) variant virus which emerged in, an immunoinformatics
integrated toolkit for the selection and optimization of antigens and the study, Vaccine 30 (2012) (2011) 60546063.
design of epitope-driven vaccines, Hum. Vaccin. Immunother. 11 (2015) [182] F. Wen, J.H. Ma, H. Yu, F.R. Yang, M. Huang, Y.J. Zhou, et al., A novel M2e
23122321. multiple antigenic peptide providing heterologous protection on mice, J. Vet.
[163] T. Sturniolo, E. Bono, J. Ding, L. Raddrizzani, O. Tuereci, U. Sahin, et al., Sci. (2015).
Generation of tissue-specific and promiscuous HLA ligand databases using [183] C.T. Su, C. Schnbach, C.K. Kwoh, Molecular docking analysis of 2009-H1N1
DNA microarrays and virtual HLA class II matrices, Nat. Biotechnol. 17 (1999) and 2004-H5N1 influenza virus HLA-B4405-restricted HA epitope
555561. candidates: implications for TCR cross-recognition and vaccine
[164] A.S. De Groot, B.M. Jesdale, E. Szu, J.R. Schafer, R.M. Chicz, G. Deocampo, An development, BMC Bioinformatics 14 (Suppl. 2:S21) (2013), http://dx.doi.
interactive Web site providing major histocompatibility ligand predictions: org/10.1186/1471-2105-1114-S1182-S1121. Epub 2013 Jan 1121.
application to HIV research, AIDS Res. Hum. Retroviruses 13 (1997) 529531. [184] S. Nitipan, T. Sritrakul, A. Kunjantarachot, S. Prapong, Identification of
[165] A.S. De Groot, B. Jesdale, W. Martin, C. Saint Aubin, H. Sbai, A. Bosma, et al., epitopes in Leptospira borgpetersenii leucine-rich repeat proteins, Infect.
Mapping cross-clade HIV-1 vaccine epitopes using a bioinformatics Genet. Evol. 14 (2013) 4657.
approach, Vaccine 21 (2003) 44864504. [185] M. Panigada, T. Sturniolo, G. Besozzi, M.G. Boccieri, F. Sinigaglia, G.G. Grassi,
[166] A.S. De Groot, E.A. Bishop, B. Khan, M. Lally, L. Marcon, J. Franco, et al., et al., Identification of a promiscuous T-cell epitope in Mycobacterium
Engineering immunogenic consensus T helper epitopes for a cross-clade HIV tuberculosis Mce proteins, Infect. Immun. 70 (2002) 7985.
vaccine, Methods 34 (2004) 476487. [186] T.A. Ahmad, B.A. El-Sayed, L.H. El-Sayed, Development of immunization trials
[167] A.S. De Groot, W. Martin, From immunome to vaccine: epitope mapping and against Eimeria spp, Trials Vaccinol. 5 (2016) 3847.
vaccine design tools, Novartis Found. Symp. 254 (2003) 5772. [187] A. Rana, A. Rub, Y. Akhter, Proteome-wide B and T cell epitope repertoires in
[168] H. Rammensee, J. Bachmann, N.P. Emmerich, O.A. Bachor, S. Stevanovic, outer membrane proteins of Mycobacterium avium subsp. paratuberculosis
SYFPEITHI: database for MHC ligands and peptide motifs, Immunogenetics 50 have vaccine and diagnostic relevance: a holistic approach, J. Mol. Recognit.
(1999) 213219. 28 (2015) 506520.
[169] P.A. Reche, J.P. Glutting, E.L. Reinherz, Prediction of MHC class I binding [188] A. Rana, Y. Akhter, A multi-subunit based, thermodynamically stable model
peptides using profile motifs, Hum. Immunol. 63 (2002) 701709. vaccine using combined immunoinformatics and protein structure based
[170] A.S. De Groot, L. Marcon, E.A. Bishop, D. Rivera, M. Kutzler, D.B. Weiner, et al., approach, Immunobiology 221 (2016) 544557.
HIV vaccine development by computer assisted design: the GAIA vaccine, [189] M. Bello, R. Campos-Rodriguez, S. Rojas-Hernandez, A. Contis-Montes de Oca,
Vaccine 23 (2005) 21362148. J. Correa-Basurto, Predicting peptide vaccine candidates against H1N1
[171] A. Fridman, A.C. Finnefrock, D. Peruzzi, I. Pak, N. La Monica, A. Bagchi, et al., influenza virus through theoretical approaches, Immunol. Res. 62 (2015)
An efficient T-cell epitope discovery strategy using in silico prediction and 315.
the iTopia assay platform, Oncoimmunology 1 (2012) 12581270. [190] S. Momtaz, A. Rahman, M. Sultana, M.A. Hossain, Evolutionary analysis and
[172] L. Moise, A.H. Gutierrez, C. Bailey-Kellogg, F. Terry, Q. Leng, K.M. Abdel Hady, prediction of peptide vaccine candidates for Foot-and-Mouth-Disease Virus
et al., The two-faced T cell epitope: examining the host-microbe interface types A and O in Bangladesh, Evol. Bioinform. Online 10 (2014) 187196.
with JanusMatrix, Hum. Vaccin. Immunother. 9 (2013) [Epub ahead of print]. [191] H.M. Geysen, R.H. Meloen, S.J. Barteling, Use of peptide synthesis to probe
[173] X. Song, L. Xu, R. Yan, X. Huang, X. Li, Construction of Eimeria tenella multi- viral antigens for epitopes to a resolution of a single amino acid, Proc. Natl.
epitope DNA vaccines and their protective efficacies against experimental Acad. Sci. 81 (1984) 39984002.
infection, Vet. Immunol. Immunopathol. 166 (2015) 7987. [192] B.C. Barnett, D.S. Burt, C.M. Graham, A.P. Warren, J.J. Skehel, D.B. Thomas, I-Ad
[174] P. Carlos, V. Roupie, S. Holbert, F. Ascencio, K. Huygen, G. Gomez-Anduro, restricted T cell recognition of influenza hemagglutinin. Synthetic peptides
et al., In silico epitope analysis of unique and membrane associated proteins identify multiple epitopes corresponding to antibody-binding regions of the
from Mycobacterium avium subsp. paratuberculosis for immunogenicity and HA1 subunit, J. Immunol. 143 (1989) 26632669.
vaccine evaluation, J. Theor. Biol. 384 (2015) 19. [193] I. Jelcic, L. Aly, T.M. Binder, I. Jelcic, S. Bofill-Mas, R. Planas, et al., T cell epitope
[175] R.S. Salvat, A.S. Parker, Y. Choi, C. Bailey-Kellogg, K.E. Griswold, Mapping the mapping of JC polyoma virus-encoded proteome reveals reduced T cell
Pareto optimal design space for a functionally deimmunized biotherapeutic responses in HLA-DRB104:01+ donors, J. Virol. 87 (2013) 33933408.
candidate, PLoS Comput. Biol. 11 (2015) e1003988. [194] S. Tu, H. Huang, S.I. Lin, H.Y. Liu, Y. Sher, S. Chiang, et al., A novel
[176] O.A. Koita, D. Dabitao, I. Mahamadou, M. Tall, S. Dao, A. Tounkara, et al., HLA-A2 -restricted CTL epitope of tumor-associated antigen L6 can inhibit
Confirmation of immunogenic consensus sequence HIV-1 T-cell epitopes in tumor growth In Vivo, J. Immunother. 35 (2012) 235244.
Bamako, Mali and Providence, Rhode Island, Hum. Vaccin. 2 (2006).

View publication stats

Das könnte Ihnen auch gefallen