Skip to main content
Erschienen in: Journal of Translational Medicine 1/2022

Open Access 01.12.2022 | Research

Antigenic molecular mimicry in viral-mediated protection from cancer: the HIV case

verfasst von: Carmen Manolio, Concetta Ragone, Beatrice Cavalluzzo, Angela Mauriello, Maria Lina Tornesello, Franco M. Buonaguro, Angelo Salomone Megna, Giovanna D’Alessio, Roberta Penta, Maria Tagliamonte, Luigi Buonaguro

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2022

Abstract

Background

People living with HIV/AIDS (PLWHA) show a reduced incidence for three cancer types, namely breast, prostate and colon cancers. In the present study, we assessed whether a molecular mimicry between HIV epitopes and tumor associated antigens and, consequently, a T cell cross-reactivity could provide an explanation for such an epidemiological evidence.

Methods

Homology between published TAAs and non-self HIV-derived epitopes have been assessed by BLAST homology. Structural analyses have been performed by bioinformatics tools. Immunological validation of CD8+ T cell cross-reactivity has been evaluated ex vivo by tetramer staining.

Findings

Sequence homologies between multiple TAAs and HIV epitopes have been found. High structural similarities between the paired TAAs and HIV epitopes as well as comparable patterns of contact with HLA and TCR α and β chains have been observed. Furthermore, cross-reacting CD8+ T cells have been identified.

Interpretation

This is the first study showing a molecular mimicry between HIV antigens an TAAs identified in breast, prostate and colon cancers. Therefore, it is highly reasonable that memory CD8+ T cells elicited during the HIV infection may play a key role in controlling development and progression of such cancers in the PLWHA lifetime. This represents the first demonstration ever that a viral infection may induce a natural “preventive” anti-cancer memory T cells, with highly relevant implications beyond the HIV infection.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-022-03681-4.
Carmen Manolio and Concetta Ragone contributed equally to this work and share first authorship
Maria Tagliamonte and Luigi Buonaguro contributed equally to this work and share last authorship

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
HIV
Human immunodeficiency virus
AIDS
Acquired Immunodeficiency Syndrome
PLWHA
People living with HIV /AIDS
TAA
Tumor associated antigen
TCR
T cell receptor
TME
Tumor microenvironment
TAA
Tumor-associated antigen
VirA
Viral antigen
MHC
Major histocompatibility complex
SIR
Standardized incidence ratio
DMSO
Dimethyl sulfoxide
PBS
Phosphate Buffered Saline
TAP
Transporter associated with antigen processing
OVA
Ovalbumin
FI
Fluorescence index
MFI
Mean fluorescence intensity
pMHC
PeptideMHC
PBMCs
Peripheral blood mononuclear cells
SB
Strong binder

Introduction

About 40 million people globally were living with HIV in 2020 (PLWHA) and a preventive vaccine is not yet available [1]. All of them have been exposed to multiple HIV epitopes eliciting a specific CD8+ T cell immunological memory. Strikingly, while the incidence rates of AIDS-defining cancers are significantly greater in PLWHA than in the general population, non-AIDS-defining cancers show variable standardized incidence ratios (SIRs) compared to the general population [2, 3]. In particular, reduced SIRs for three cancer types, namely breast, prostate and colon cancers have been reported by several meta-analyses [47]. So far, no biological explanations have been provided for such epidemiological evidence. A reduced implementation of population screenings has been proposed. But this seems to be unlikely, given that the lower risk is present for both early-stage tumors, primarily screen-detected, and larger tumors, clinically detected.
In the present study we wanted to verify whether the molecular mimicry between HIV antigens and TAAs expressed by breast, prostate and colon cancers could explain the reduced SIRs in PLWHA. In a word, whether the HIV infection could represent a natural “anti-cancer vaccination”.
Indeed, viruses are a natural source of non-self antigens (VirAs) expressed by host’s cells in the context of the HLA class I molecules, inducing a wide pool of specific memory CD8+ T cell clones. We and others have previously shown that the molecular mimicry between viral and tumor antigens may have a significant impact in controlling tumor growth and improving the clinical outcome in cancer patients [811]. More recently, we have shown that TAAs may show sequence homology as well as structural similarities with viral peptides and cross-reacting CD8 + T cells can be identified to drive the fate of cancer development and progression [12]. Therefore, a previous viral infection may represent a natural “anti-cancer vaccination” and anti-viral memory CD8 + T cells may be promptly recalled by cancer cells expressing TAAs similar or identical to VirAs. This is possible because the degeneracy of the TCR in antigen recognition allows each single receptor to cross-react against similar antigens, recognizing at least 106 different MHC-bound peptides [13, 14].
Indeed, striking sequence homologies between multiple TAAs and HIV epitopes have been found. High structural similarities between the paired TAAs and HIV epitopes as well as comparable patterns of contact with HLA and TCR α and β chains have been observed. Furthermore, cross-reacting CD8+ T cells have been identified with shared sequences in TCR α and β chains.
Overall, it is highly reasonable that memory CD8 + T cells elicited by HIV epitopes may play a key role in controlling development and progression of such cancers in the PLWHA lifetime.

Materials and methods

Protein expression analysis

Starting from downloadable data section of Human Protein Atlas database (https://​www.​proteinatlas.​org/​about/​download), Normal Tissue and Pathology dataset were downloaded. They contain expression profiles evaluated by immunohistochemistry on tissue micro array of proteins of different human tissues samples, non tumoral and tumoral respectively.
Normal Tissue dataset is an extremely wide list containing information about the expression of several proteins in different cell types of a specific human tissue. To identify tumor specific proteins, only those proteins who were defined as “not detected” in normal cells from the same tissues were selected. Subsequently, these proteins were searched in the Pathology dataset to select only those detected in “colon, breast, prostate, lung or brain cancer (ca)” tissue samples at “high”, “medium” or “low” expression level. The definition of expression levels was according to Human Protein Atlas database.

HLA class l epitope prediction

In order to predict MHC-class I epitopes, protein sequences were downloaded from UniProt database (https://​www.​uniprot.​org/​) and the entire sequence was analyzed with NetMHCpan4.1 tool (https://​services.​healthtech.​dtu.​dk/​service.​php?​NetMHCpan-4.​1), to predict the binding affinity [15]. Prediction analyses were performed for the 12 most frequent alleles in the world, selecting only peptides considered weak and strong binders, according to default parameters (WB; SB).
Peptides selected as SB according to NetMHCpan 4.1 prediction tool have been submitted to BLAST for a homology search against HIV-1 (taxid: 11,676) within the non-redundant protein sequences database (https://​blast.​ncbi.​nlm.​nih.​gov/​Blast.​cgi). HIV-derived homologous sequences were analyzed to predict their binding affinity and stability to all alleles as described above.

Epitope modelling and molecular docking

The 3D structure of interaction between peptides and HLA alleles was generated using Pymol software (PyMol Molecular graphics system, version 1.8.6.2) and Molsoft ICM (http://​www.​molsoft.​com/​, version 3.8-7d) software. The PDB format of complex between HLA molecules and reference peptides were downloaded from RCS Protein Data Bank (PDB) website (https://​www.​rcsb.​org). In particular, HLA-A*01:01 (3BO8), HLA-A*02:01 (1AO7), HLA-A*03:01 (3RL1), HLA-A*24:02 (7JYV), HLA-B*07:02 (7LGD), HLA-B*08:01 (7NUI), HLA-B*15:01 (6VB3), HLA-B*39:01 (4O2E), HLA-B*40:02 (5IEK), HLA-B*58:01 (5VWH). The original peptide sequences in the downloaded structures were replaced with individual predicted epitopes using Pymol visualization system and Molsoft ICM software was used to visualize the molecular docking.

Peptide synthesis and solubilization

All peptides were synthesized at a purity of ≥ 90% (GenScript; NJ, USA). Lyophilized powders were reconstituted in DMSO Solution (CARLO ERBA Reagents S.r.l., Cornaredo; Italy) and diluted in 90% of 1X PBS (HyClone, Thermo Fisher Scientific Inc., US).

Peptide binding affinity and BFA decay assays

Peptide binding affinity to HLA-A*02:01 molecule and BFA decay assays were performed for each candidate peptide. Human TAP-deficient T2 cell line (174xCEM.T2; ATCC CRL 1992™) was purchased from American Type Culture Collection (ATCC; https://​www.​atcc.​org/​) and cultured in Iscove's modified Dulbecco's medium (IMDM; Gibco Life Technologies) containing 25 mM HEPES and 2 mM L-Glut, supplemented with 20% fetal bovine serum (FBS; Capricorn Scientific GmbH), 100 IU/ml penicillin and 100 μg/ml streptomycin (Gibco Life Technologies). Cells were maintained at 37 °C in a humidified incubator with 5% CO2. Briefly, T2 were seeded at 3.5 × 105 cells per well in 24 well plates and incubated 16 h at 27°C with peptides (final concentrations: 5, 10, 20, 50 and 100 μM) in IMDM serum free medium. The next day, cells have been incubated for additional 2 h at 37 °C. Following incubation, cells were harvested and centrifuged at 200 × g for 5 min. Subsequently, cells were washed twice with phosphate buffered saline (1X PBS; Gibco Life Technologies) and stained with R-PE conjugated anti human HLA-A2 mono-clonal antibody (cat. 343,306; BioLegend), for 30 min at 4 °C, and analyzed with the Attune™ NxT flow cytometer (Thermo Fisher Scientific). Mouse H-2 Kb-specific OVA SIINFEKL peptide was used as negative control and T2 cells without any added peptide were used as a background control. A fluorescence index (FI) was calculated using the following formula: FI = [mean fluorescence intensity (MFI) sample – MFI background]/MFI background, where MFI background represents the value without peptide. A value of FI > 0.5 was set as threshold to indicate peptides with affinity for the HLA-A*02:01 molecule. For the brefeldin-A decay assay, T2 were incubated with peptides (50 μM) as described above, washed and treated with 1X BFA (brefeldin A solution; cat. 420,601; BioLegend) in IMDM serum free medium, for 1 h at 37°C. Cells were harvested every two hours (T0, T2, T4, T6, T8), washed with phosphate buffered saline (1X PBS; Gibco Life Technologies), stained with anti HLA-A*0201 fluorescent monoclonal antibody (cat. 343,306; BioLegend) and analyzed by flow cytometry. The stability of each peptide bound to HLA-A*02:01 was measured as the DC50 value, which was defined as an estimate of the time required for a 50% reduction of the MFI value at time 0. The DC50 value was calculated according to the formula: MFI at indicated time points/MFI at time 0 × 100. All the experiments were performed in triplicate [11, 16].

pMHC multimer preparation and T cell staining

pMHC complexes were generated by combining purified disulfide-stabilized HLA-A*0201 monomer (100 μg/ml) with 100 μM peptide for 30 min. in PBS at room temperature [17]. pMHC complexes were centrifuged for 5 min. at 3300 × g to sediment any aggregated MHC molecules. For each 100 μl pMHC, 9.02 μl (0.2 mg/ml stock, SA-PE (Bio-legend, 405,204), SA-APC (Biolegend, 405,207), SA-PE/Cy7 (Biolegend, 405,206), SA-PE-CF594 (BD, 562,284)) or 18.04 μl (0.1 mg/ml stock, SA-BV421 (Biolegend, 405,226), SA-BV650 (Biolegend, 405,231) streptavidin-conjugate was added and incubated for 30 min. on ice. D-biotin (Sigma-Aldrich) was added at a final concentration of 25 μM to block any free binding sites and multimers were stored at − 20 °C with 5% glycerol and 0.5% BSA. To stain for T cell reactivity, PBMCs from 16 HIV-1 patients and 10 healthy donors (2–5 × 106) were incubated with a pool of pMHC multimers (3 μl/multimer) and dasatinib (50 nM final, LC laboratories, D-3307) for 15 min. at 37 °C. Cells were then stained with antibodies CD3-FITC (1:40, BD, 345,763) and CD8-BV480 (1:100, BD, 566,121) and LIVE/DEAD Fixable Near-IR (1:1000, Invitrogen, L10119) for 30 min. on ice and washed twice in FACS buffer (PBS + 2% FCS). Gating for CD3+/CD8+ T cells was done on alive cells and binding to pMHCs was assessed by measuring specific fluorescence associated with each individual pMHC. Samples were acquired on a flow cytometer (LSRFortessa, BD) and analyzed by the Attune NxT software v3.1.2 (ThermoFisher scientific).

Statistical analysis

Comparison between individual data points were performed with the two-sided Student’s t-test and ANOVA, as appropriate. Paired, for the matched samples collected at the Istituto Nazionale Tumori—IRCCS—"Fond G. Pascale", Naples, Italy (INT); unpaired for the samples extracted by the publicly available data (http://​gent2.​appex.​kr/​gent2/​). Normally distributed data were represented as mean ± S.E.M. Two-way ANOVA and Bonferroni post-hoc analysis were used to examine the significance of differences among groups. All P values were two-tailed and considered significant if less than 0.05.

Results

Identification of tumor-related proteins

In order to identify proteins specifically overexpressed in breast, prostate and colon cancers and not detected in normal cells, the Normal Tissue dataset (15,170 unique proteins) and the Pathology dataset (15,313 unique proteins) available at the Human Protein Atlas were sequentially interrogated (https://​www.​proteinatlas.​org/​about/​download). The same analysis was performed for the lung and brain cancers, which show increased SIRs in PLWHA [5].
An average of 5,768 proteins showed no expression in normal cells of the five tissues, with a range from 4,814 (breast glandular cells) to 7,019 (lung pneumocytes). In parallel, an average of 4,134 proteins were found to be detected in cancer cells, with a range from 2,640 (lung ca) to 5,650 (colon ca). Of the latter proteins, an average of 213 proteins (5.15%) were found to be at high levels in cancer cells, as defined by the Human Protein Atlas, with a range from 86 (lung ca) to 408 (colon ca). Cancer-specific proteins were selected for each tissue as those not detected in normal cells and highly expressed in cancer cells,. The number of the latter proteins ranged from 1 to 2 for breast, prostate, lung and brain tumors. The only exception was the colon cancer for which 44 cancer-specific proteins were identified (Table 1). The subsequent filter was set for selecting proteins not detected in > 70% of normal cells of any tissue. Indeed, only such proteins should not be affected by immune tolerance impairing the immune system to mount responses against self antigens. According to such additional filter, six cancer-specific proteins in colon cancer (CEACAM8, CDH17, GLOD5, PPM1E, TRIM16, EPCAM) and one in breast cancer (CCR9) were identified. None of the cancer-specific proteins identified in prostate cancer, brain and lung cancer met these parameters (Additional file 1: Fig. S1). Nevertheless, they were selected for the subsequent analyses.
Table 1
Number of proteins identified in normal and cancer cells of indicated tissues. Cancer specific proteins are defined in the text
Tumor
Normal
Cancer
Cancer specific
 
N.D
All
High
 
Breast
4814
4997
219
1
Colon
5823
5650
408
44
Prostate
5036
4543
187
1
Lung
7019
2640
86
2
Brain
6151
2841
169
2
Average
5768.6
4134.2
213.8
 

Prediction of Tumor-associated antigens—TAAs

The cancer-specific proteins selected in each cancer were submitted to the NetMHCpan software to predict 9 aa-long epitopes (nonamers) strong binders (SBs) to HLA alleles covering more than 70% of the World population. The analysis was performed considering all the overlapping nonamers with a one-amino-acid lateral shift covering the entire protein sequence. In order to select the best predicted epitopes, we focused our analysis only on those showing an affinity < 100 nM to HLA molecules, which we have been previously shown to have a 100% concordance with ex vivo binding assay [11, 12, 16].
The number of SBs with such characteristics predicted from the cancer-specific proteins was highly variable among the five cancers. The highest number of SBs (138) was predicted in colon cancers from the 6 cancer-specific proteins, covering all the evaluated HLA alleles; the lowest number [27] was predicted in breast cancer from the single CCR9 cancer-specific protein (Fig. 1A). Regardless the different numbers of SBs predicted for the cancer-specific proteins from the 5 cancers, their average affinity to the HLA alleles was very high (e.g. < 50 nM) (Fig. 1B). The number of SBs specific to each HLA allele was highly variable. The highest number was predicted for HLA-A*02:01 (51), the second and third highest numbers were predicted for HLA-B*15:01 (42) and HLA-B*58:01 (35). On the contrary, the lowest numbers by far were predicted for HLA-A*01:01 [7] and HLA-A*26:01 [10]. The number of predicted SBs for all other HLA alleles ranged between 13 and 30 (Additional file 1: Fig. S2). Furthermore, the average affinity to the HLA alleles was highly variable without reaching a statistical difference. The highest average affinity was observed for HLA-A*02:01 specific epitopes (e.g. 26 nM), the lowest was observed for HLA-B*08:01 specific epitopes (e.g. 52 nM) (Fig. 1C).
In order to verify the expression of the predicted SBs on normal cells, the HLA Ligand Atlas was interrogated. Interestingly, 271 (93.7%) of the 298 SBs predicted in the cancer-specific proteins were found not expressed in the HLA ligandome of normal cells (Table 2). All peptides found at the HLA Ligand Atlas were excluded from subsequent analyses. Moreover, some of such predicted TAAs have been previously described to be presented by cancer cells and/or be target of T cells [1822].
Table 2
Predicted TuAs from cancer-specific proteins and their presence in the HLA Ligand Atlas of normal tissues
MHC
Peptide
Aff (nM)
HLA Ligand Atlas
Breast: CCR9
HLA-A*02:01
AIADLLFLV
3,76
No match
HLA-A*02:01
FLPPLYWLV
2,67
No match
HLA-A*02:01
ILGFFLPFV
5,12
No match
HLA-A*03:01
KSSKHKALK
73,36
No match
HLA-A*24:02
LYVFVGERF
22,26
No match
HLA-A*24:02
CYTIIIHTL
40,22
No match
HLA-A*24:02
RYIAIAQAM
70,57
No match
HLA-A*26:01
QTIDAYAMF
19,7
No match
HLA-B*07:02
TPTDFTSPI
68,5
No match
HLA-B*08:01
STKLKSAVL
48,06
No match
HLA-B*39:01
FHSCLNPVL
5,92
No match
HLA-B*39:01
SQFPYNCIL
45,12
No match
HLA-B*40:01
MEDYVNFNF
46,12
No match
HLA-B*58:01
IAAADQWKF
13,32
No match
HLA-B*58:01
NSLVILVYW
28,08
No match
HLA-B*58:01
VTITVLTVF
48,71
No match
HLA-B*58:01
LTLKVILGF
31,09
No match
HLA-B*58:01
WAIAAADQW
7,46
No match
HLA-B*58:01
LGCISQAQW
22,22
No match
HLA-B*58:01
KTMTDMFLL
10,03
No match
HLA-B*58:01
SSMEDYVNF
96,02
No match
HLA-B*58:01
LTVFVLSQF
92,21
No match
HLA-B*15:01
FQVTQTIAF
11,07
No match
HLA-B*15:01
LLVQTIDAY
23,98
No match
HLA-B*15:01
LLYSKMVCF
42,55
No match
HLA-B*15:01
RVKTMTDMF
94,33
No match
HLA-B*15:01
VTITVLTVF
39,98
No match
Brain: DPYSL2
HLA-A*02:01
KMDENQFVA
37,21
Ovary
HLA-A*02:01
VLEDGTLHV
56,91
No match
HLA-A*02:01
ALAGGTTMI
98,73
No match
HLA-A*03:01
KVFNLYPRK
11,63
No match
HLA-A*03:01
ITSDRLLIK
83,95
No match
HLA-A*24:02
VYMAFKDRF
8,25
Liver, Lung
HLA-A*26:01
HVTEGSGRY
64,02
No match
HLA-B*07:02
YPRKGRIAV
2,87
Liver, Lung
HLA-B*07:02
IPRRTTQRI
19,79
No match
HLA-B*07:02
IPRITSDRL
26,75
No match
HLA-B*07:02
IPRKPFPDF
27,37
No match
HLA-B*07:02
KARSRLAEL
37,6
No match
HLA-B*07:02
SAKTHNSSL
74,97
No match
HLA-B*07:02
VPEPGTSLL
88,37
No match
HLA-B*08:01
SAKTHNSSL
40,66
No match
HLA-B*08:01
YPRKGRIAV
54,2
No match
HLA-B*15:01
FQLTDCQIY
53,36
No match
HLA-B*15:01
KTHNSSLEY
78,35
Tongue
HLA-B*27:05
KRIKARSRL
70,69
No match
HLA-B*27:05
ARSRLAELR
76,74
No match
HLA-B*27:05
SRMVIPGGI
76,85
No match
HLA-B*40:01
LEYNIFEGM
42,86
Multiple Tissues
HLA-B*40:01
IEAHSRMVI
86,33
No match
HLA-B*40:01
MEDGLIKQI
95,51
Multiple Tissues
HLA-B*58:01
AAFDQWREW
26,57
No match
HLA-B*58:01
LGTDGSHYW
27,85
No match
HLA-B*58:01
GSDADLVIW
38,13
No match
HLA-B*58:01
SLLAAFDQW
53,69
No match
Brain: SPARC
HLA-A*02:01
WLKNVLVTL
38,11
No match
HLA-A*24:02
YIFPVHWQF
79,77
Multiple tissues
HLA-B*08:01
WLKNVLVTL
91,35
No match
HLA-B*15:01
KTFDSSCHF
29,21
No match
HLA-B*27:05
MRAWIFFLL
9,57
No match
HLA-B*39:01
MRDWLKNVL
8,7
No match
HLA-B*39:01
MRAWIFFLL
15,81
No match
HLA-B*58:01
KTFDSSCHF
14,61
No match
Colon: CDH17BIS
HLA-A*01:01
VIEREGLLY
70,39
No match
HLA-A*01:01
FTGSSKILY
43,81
No match
HLA-A*01:01
NTANSFLNY
46,99
No match
HLA-A*02:01
ILQAHLHSL
12,25
No match
HLA-A*02:01
KLPRFPFSI
14,74
No match
HLA-A*02:01
IQLPMINNV
54,97
No match
HLA-A*02:01
LVIGIILAV
32,48
No match
HLA-A*02:01
GLFLIQTYA
43,93
No match
HLA-A*03:01
VMYFQINNK
10,12
No match
HLA-A*03:01
QLAKQSLKK
72,8
No match
HLA-A*03:01
AVVFIRIKK
79,68
No match
HLA-A*24:02
LFRGPHFTF
71,32
No match
HLA-A*26:01
NTANSFLNY
56,93
No match
HLA-B*07:02
HPLSAPGSL
11,9
No match
HLA-B*07:02
RPAGHQTGI
13,94
No match
HLA-B*07:02
APQFSQHVF
55,95
No match
HLA-B*07:02
RPGKPFLYV
59,59
No match
HLA-B*08:01
ILQAHLHSL
37,38
No match
HLA-B*08:01
MLQLAKQSL
92,53
No match
HLA-B*39:01
THNLQVAAL
16,43
No match
HLA-B*39:01
FKAENPEPL
13,19
No match
HLA-B*40:01
RETRSTHNL
35,21
No match
HLA-B*40:01
FEVQENERL
42,52
No match
HLA-B*40:01
GETDNIFVI
12,73
No match
HLA-B*40:01
FEEREYVVL
18,94
No match
HLA-B*40:01
GEIFSVAPL
5,02
No match
HLA-B*40:01
FETAAVSNI
52,35
No match
HLA-B*40:01
QEGKFSGPL
41,12
No match
HLA-B*58:01
KANPPAVTF
17,57
No match
HLA-B*58:01
LSAPGSLIF
12,51
No match
HLA-B*58:01
GSGSLQNDW
83,99
No match
HLA-B*58:01
SSLSSVSEF
88,68
No match
HLA-B*15:01
RLSTRHTEF
44,77
No match
HLA-B*15:01
GVKYNASSF
67,45
No match
HLA-B*15:01
LSAPGSLIF
34,98
No match
Colon: GLOD 5
MHC
HLA-A*02:01
RLDHIVMTV
5,77
No match
HLA-A*02:01
NLIEVSNYI
10,19
No match
HLA-A*03:01
KMWGRTLEK
3,88
No match
HLA-A*03:01
IVMTVKSIK
55,26
No match
HLA-B*08:01
MLRHLPSRL
90,51
No match
HLA-B*58:01
KSIKDTTMF
9,83
No match
HLA-B*15:01
SIKDTTMFY
60,19
No match
Colon: PPM1E
HLA-A*01:01
CSAPADLGY
31,65
No match
HLA-A*02:01
SVFSKLHEI
36,35
No match
HLA-A*02:01
FLAAALARA
5,94
No match
HLA-A*02:01
YLDLTQIEA
57,91
No match
HLA-A*02:01
SLSPVCSGL
61,33
No match
HLA-A*03:01
KLARSVFSK
9,37
No match
HLA-A*03:01
AIYASIHLH
98,98
No match
HLA-A*03:01
LINELMMEK
59,44
No match
HLA-A*03:01
VVFLRDMNK
32,23
No match
HLA-A*03:01
HLRHHYSKK
46,87
No match
HLA-A*03:01
RIRSSLPWR
53,88
No match
HLA-A*24:02
RFNPKFYSF
11,44
No match
HLA-A*24:02
HYSKKWHRF
20
No match
HLA-A*24:02
TYRRFLELF
14,81
No match
HLA-A*24:02
IYASIHLHV
38,63
No match
HLA-A*24:02
YYETSIHAI
83,38
No match
HLA-A*24:02
LYKYNCPSF
35,21
No match
HLA-B*07:02
FPLRRRPQL
9,87
No match
HLA-B*07:02
FPHDPAEAL
63,53
No match
HLA-B*07:02
SPGNRVSRL
35,08
No match
HLA-B*07:02
SPGSQINVL
51,81
No match
HLA-B*07:02
KPHSAQFLL
47,07
No match
HLA-B*07:02
LPRPLSERI
66,63
No match
HLA-B*07:02
CPSFLAAAL
22,27
No match
HLA-B*08:01
FPLRRRPQL
9,91
No match
HLA-B*08:01
MLVRKGQAV
26,5
No match
HLA-B*08:01
KTYRRFLEL
92,81
No match
HLA-B*15:01
LQSDLSAHY
11,02
No match
HLA-B*15:01
ASKPHSAQF
74,89
No match
HLA-B*15:01
AQFLLPVEM
90,51
No match
HLA-B*15:01
RLSHLRHHY
67,82
No match
HLA-B*27:05
HRFRFNPKF
21,58
No match
HLA-B*27:05
KRNRIRSSL
47,8
No match
HLA-B*27:05
YRRFLELFL
52,81
No match
HLA-B*39:01
WRVNGSLSV
42,89
No match
HLA-B*39:01
YRMQSLSPV
16,71
No match
HLA-B*40:01
GEFPTAFNL
8,41
No match
HLA-B*40:01
REEVEGESL
22,28
No match
HLA-B*40:01
LEDPGYLDL
59,85
No match
HLA-B*40:01
QEEQAYFAV
54,53
No match
HLA-B*58:01
SVQSSLPEW
44,83
No match
HLA-B*58:01
RGNMLHVAW
34,54
No match
Colon: TRIM16
HLA-A*02:01
YLHRYYFEV
2,33
No match
HLA-A*02:01
VLSQQSLYL
58,02
No match
HLA-A*02:01
RMAAISNTV
13,5
No match
HLA-A*03:01
GTYVGLTCK
29,93
No match
HLA-A*03:01
SVYVGLKDK
83,52
No match
HLA-A*24:02
RYYFEVEIF
10,7
No match
HLA-A*26:01
DTMTLVHKF
63,32
No match
HLA-A*26:01
NTVQFLEEY
32,67
No match
HLA-B*08:01
SLYLHRYYF
95,86
No match
HLA-B*15:01
LQYAYDITF
8,56
No match
HLA-B*15:01
HLIQLLENY
64,75
No match
HLA-B*15:01
RQVLSQQSL
84,65
No match
HLA-B*15:01
ILSFYGVEY
57,78
No match
HLA-B*15:01
SLYLHRYYF
75,83
No match
HLA-B*27:05
RRLGVYIDF
56,62
No match
HLA-B*27:05
SRFLHWRQV
54,5
No match
HLA-B*40:01
AEMQFGELL
4,59
Thymus
HLA-B*40:01
SEVKAVAEM
28,88
No match
HLA-B*40:01
LEEKEQAAL
40,08
No match
HLA-B*40:01
QEHSGHTIV
61,14
No match
HLA-B*40:01
QELERMAAI
97,6
No match
HLA-B*58:01
KAQANVMLF
13,36
No match
HLA-B*58:01
CISGNNFSW
31,21
No match
Colon: CEACAM8
HLA-A*01:01
FSDPVTLNV
44,46
Bone marrow
HLA-A*02:01
IMIGVLARV
5,67
No match
HLA-A*02:01
FSDPVTLNV
65,43
No match
HLA-A*02:01
KLNLMSEEV
39,39
No match
HLA-A*03:01
GTFQQYTQK
35,5
Bone marrow
HLA-A*03:01
GSYTLQVIK
70,59
No match
HLA-A*24:02
LYGPDAPTI
83,9
No match
HLA-A*24:02
IYPNASLLM
53,31
No match
HLA-A*26:01
ETIYPNASL
71,49
No match
HLA-B*07:02
LPVSPRLQL
9,45
No match
HLA-B*15:01
QQITPGPAY
19,97
Bone marrow
HLA-B*15:01
IQNPASANF
9,34
Bone marrow
HLA-B*15:01
SVNGTFQQY
83,79
Bone marrow
HLA-B*15:01
FQQYTQKLF
52,48
No match
HLA-B*15:01
VTRNDTGSY
94,41
No match
HLA-B*27:05
WRIPWQGLL
18,87
No match
HLA-B*39:01
YHAGVNLNL
9,24
No match
HLA-B*39:01
WRIPWQGLL
91,33
No match
HLA-B*40:01
AEGKEVLLL
78,97
No match
HLA-B*58:01
LTASLFTFW
3,13
No match
HLA-B*58:01
ETQNTTYLW
36,36
No match
HLA-B*58:01
YSWSVNGTF
20,84
No match
Colon: EPCAM
HLA-A*02:01
GLKAGVIAV
69,41
No match
HLA-A*02:01
VVAGIVVLV
48,75
No match
HLA-A*03:01
KSLRTALQK
50,33
No match
HLA-A*26:01
DIADVAYYF
90,16
No match
HLA-B*40:01
KEITTRYQL
25,58
No match
HLA-B*40:01
AEIKEMGEM
66,18
No match
HLA-B*58:01
CSERVRTYW
23,26
No match
HLA-B*15:01
LQKEITTRY
14,52
No match
HLA-B*15:01
LLAAATATF
4,39
No match
Prostate: NDUFS2
HLA-A*01:01
VTAEEALNY
58,86
No match
HLA-A*01:01
VSDGSSRPY
72,62
No match
HLA-A*02:01
RLLNHIMAV
2,59
No match
HLA-A*02:01
KMFEFYERV
3,05
No match
HLA-A*02:01
KLIEYKTYL
4,96
No match
HLA-A*02:01
MLADVVAII
5,94
No match
HLA-A*02:01
KLYTEGYQV
6,73
No match
HLA-A*02:01
YQFSKNFSL
7,18
Multiple tissues
HLA-A*02:01
HMLADVVAI
16,16
No match
HLA-A*02:01
VLFGEITRL
19,13
No match
HLA-A*02:01
ALNYGFSGV
23,23
No match
HLA-A*02:01
IMAVTTHAL
27,34
No match
HLA-A*02:01
RLDELEELL
29,47
No match
HLA-A*02:01
GLMDDIYQF
49,65
No match
HLA-A*02:01
KMPPGEIKV
84,76
No match
HLA-A*03:01
KTYLQALPY
18,63
No match
HLA-A*03:01
RIIAQCLNK
25,84
No match
HLA-A*03:01
GLLHRGTEK
52,91
No match
HLA-A*24:02
TYLQALPYF
10,57
No match
HLA-A*24:02
AMTPFFWLF
20,73
No match
HLA-A*24:02
RYLCRVEEM
59,38
No match
HLA-B*07:02
RPGGVHQDL
21,45
No match
HLA-B*07:02
RVEEMRQSL
90,96
No match
HLA-B*15:01
YQVPPGATY
9,56
No match
HLA-B*15:01
AQQFGGAVM
15,81
No match
HLA-B*15:01
SMMCNEQAY
16,28
No match
HLA-B*15:01
QQFGGAVMY
23,49
No match
HLA-B*15:01
KTYLQALPY
41,5
No match
HLA-B*15:01
YQFSKNFSL
56,91
No match
HLA-B*15:01
IVKNITLNF
58,22
No match
HLA-B*15:01
SLIHHFKLY
68,72
No match
HLA-B*27:05
ARMHAAYIR
37,13
No match
HLA-B*39:01
YQFSKNFSL
5,33
No match
HLA-B*39:01
FRGVAAQVL
45,59
No match
HLA-B*39:01
THALDLGAM
81,98
No match
HLA-B*40:01
AEEALNYGF
88,35
No match
HLA-B*58:01
LGAMTPFFW
6,09
No match
HLA-B*58:01
RAQWIRVLF
56,87
No match
Lung: CNIH4
HLA-A*02:01
LLMSLHWFI
3,25
No match
HLA-A*02:01
YLYSMILAL
3,82
No match
HLA-A*02:01
SLHWFIFLL
10,12
No match
HLA-A*02:01
FLLNLPVAT
15,76
No match
HLA-A*02:01
AMIKLGFHL
21,24
No match
HLA-A*02:01
LIGHTIVTV
69,56
No match
HLA-A*24:02
LYSMILALI
32,32
No match
HLA-B*07:02
VPSGNMGVF
77,38
No match
HLA-B*08:01
MIKLGFHLL
25,27
No match
HLA-B*58:01
LLNLPVATW
99,66
No match
Lung: PRPF40A
HLA-A*02:01
YLMDNPTFA
4,03
No match
HLA-A*02:01
ALDLFKFYV
8,07
No match
HLA-A*02:01
MMMSHMSQA
9,12
No match
HLA-A*02:01
RIFKDFMHV
15,96
No match
HLA-A*02:01
NILDNMANV
34,74
No match
HLA-A*02:01
TLDAGNIKL
84,81
Ovary
HLA-A*03:01
KMTSTTRYK
11,36
No match
HLA-A*03:01
KSNLHAMIK
29,28
No match
HLA-A*03:01
IVAGSLITK
36,6
Multiple tissues
HLA-A*03:01
KQAFKELLK
63,33
No match
HLA-A*03:01
LAFNSLLEK
65,36
No match
HLA-A*03:01
TVADFTPKK
91,15
No match
HLA-A*03:01
MTSTTRYKK
98,8
No match
HLA-A*24:02
SWMELYPTI
12,79
No match
HLA-A*24:02
QYLMDNPTF
54,19
No match
HLA-A*24:02
RYKKAEQMF
69,86
No match
HLA-A*26:01
EIPTTMSTM
42,38
No match
HLA-A*26:01
EIYEDVLFF
48,08
No match
HLA-A*26:01
FVVEVNTTF
90,52
Multiple tissues
HLA-B*07:02
HPMGQRANM
5,68
No match
HLA-B*07:02
RPSMGHPGM
6,99
No match
HLA-B*07:02
HPGMHYAPM
8,62
No match
HLA-B*07:02
MPGMMSSVM
11,35
No match
HLA-B*07:02
MPPMGGPPM
28,69
Spleen
HLA-B*07:02
MPPVPHGMM
60,4
No match
HLA-B*07:02
VPHGMMPQM
70,06
No match
HLA-B*08:01
NIKLAFNSL
30,71
No match
HLA-B*08:01
ELEKRRRTL
57,75
No match
HLA-B*15:01
RQRKNRESF
38,7
No match
HLA-B*15:01
FVVEVNTTF
39,02
No match
HLA-B*15:01
KMKRKESAF
42,12
No match
HLA-B*15:01
SMGHPGMHY
45,42
No match
HLA-B*15:01
SMSSWMELY
55,07
No match
HLA-B*15:01
SQASMQPAL
93,92
No match
HLA-B*27:05
RRRTLLEQL
35,63
No match
HLA-B*27:05
SRWAKPKEL
70,18
No match
HLA-B*39:01
SQASMQPAL
33,18
No match
HLA-B*39:01
NRESFQIFL
87,31
No match
HLA-B*40:01
GEMEVWNAI
5,84
No match
HLA-B*40:01
KESAFKSML
27,62
No match
HLA-B*58:01
GSSLCSGSW
22,83
No match
HLA-B*58:01
QMFGEMEVW
23,31
No match
HLA-B*58:01
KTGKDSGNW
25,86
No match
HLA-B*58:01
TASGAKSMW
39,94
No match
HLA-B*58:01
STALDLFKF
58,82
No match

Identification of homologous HIV-1 epitopes

The following step was to verify the molecular mimicry between TAAs predicted for the cancer-specific proteins and HIV-1 antigens. Indeed, this might explain a cross-protective anti-cancer immune response elicited by HIV-1 antigens in HIV-1 patients. To this aim, the predicted 298 TAAs were screened in BLAST for homology to HIV-1 peptide sequences. In total, 25 HIV-1 peptides were identified with sequence homology to predicted TAAs and showing high binding affinity to HLA alleles (i.e. < 100 nM) (Table 3). Interestingly, the highest number of HIV-1 peptides were identified for colon cancer (eleven) and breast cancer (six). Five peptides were identified for lung cancer, two for prostate cancer and only one for brain cancer (Fig. 2A). The broadest HLA coverage (6 alleles) was observed for the HIV peptides homologous to colon-specific TAAs, while the narrowest one (1 allele) was observed for the HIV peptides homologous to prostate-specific and brain-specific TAAs. Most importantly, only HIV peptides homologous to colon-, breast- and prostate-specific TAAs showed strong binding to HLA-A alleles (01:01; 02:01; 03:01 and 24:02) which are the most frequent in the world population (Fig. 2 B; Additional file 1: Fig. S3A; Additional file 1: Fig. S4). Considering the individual cancer-specific proteins, the highest number of HIV peptides homologous to TAAs (six) were predicted for the CCR9 (breast ca) and EPCAM (colon ca) proteins, covering the two most frequent HLA-A alleles in the world population (02:01 and 24:02) (Fig. 2 C; Additional file 1: Fig. S3B). The average binding affinity of such HIV peptides to the HLA alleles was very high (18.23 nM for A*02:01; 43,35 nM for A*24:02) (Fig. 2 D).
Table 3
HIV epitopes with homology to TuAs derived from cancer-specific proteins
Breast CA—CCR9
HLA
TAA
HIV
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-A*02:01
AIADLLFLV
3,76
RLRDLLFLV
10,01
env
AEN20914.1
HLA-B*58:01
LGCISQAQW
22,22
YCNISQAQW
30,01
env
QBF53490.1
HCNISEAQW
66,16
env
AFE02648.1
HCNISQTQW
61,58
env
QGJ15916.1
HLA-B*58:01
NSLVILVYW
28,08
ASLVIITYW
11,65
vif
AAW57764.1
HLA-A*24:02
CYTIIIHTL
40,22
NYTEIIHTL
43,35
env
AAX97247.1
Brain CA—DPYSL2
HLA
TAA
HIV
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-B*07:02
IPRRTTQRI
19,79
IPRRIRQRI
25,38
env
AET80771.1
Colon CA—CEACAM8
HLA
TAA
HIV
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-B*15:01
IQNPASANF*
9,34
RQGPASSNF
20,55
pol
QIC94847.1
HLA-B*58:01
YSWSVNGTF
20,84
SSWNVNGTW
5,6
env
ANO43474.1
HLA-A*03:01
GTFQQYTQK*
35,5
RTFQQYKKK
42
env
ABA08367.1
HLA-B*15:01
VTRNDTGSY
94,41
VTRNDTSTY
71,3
env
AAO20547.1
Colon CA—GLOD5
HLA
TAA
HIV
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-A*03:01
KMWGRTLEK
3,88
TQWGRTLEK
90,05
env
ACD34677.1
Colon CA—EPCAM
HLA
TAA
HIV
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-B*58:01
CSERVRTYW
23,26
VSERLKTYW
20,72
env
AAO20591.1
HLA-A*02:01
VVAGIVVLV
48,75
VVAGIIALV
24,48
vpu
AUO72667.1
VIAGIVALV
17,96
vpu
ABI50903.1
VVASIVALV
36,96
vpu
QGU21448.1
HLA-B*40:01
AEIKEMGEM
66,18
AELGEMGEM
87,18
vpu
AEW28038.1
HLA-B*39:01
THSPNSRAV
81,49
THSPSSREL
51,06
pol
AMX26992.1
Lung CA—PRPF40A
HLA
TAA
HIV
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-B*08:01
QLRKRNWEA
57,55
LLRRRGWEA
22,01
env
ACE65673.1
VLRRRGWEA
43,08
env
AXP17656.1
TLRRRGWEA
27,86
env
QMX89949.1
ILRRRGWEA
35,96
env
QPK37756.1
Lung CA—CNIH4
HLA
TAA
HIV
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-B*58:01
LLNLPVATW
99,66
HCNLSVATW
38,73
env
ACR83664.1
Prostate CA—NDUFS2
 
HLA
TAA
HIV
 
PEPTIDE
AFF (nM)
PEPTIDE
AFF (nM)
Protein
GeneBank CODE
HLA-A*01:01
VSDGSSRPY
72,62
SSDNSSRPY
60,07
env
ANC69009.1
GSDSTSRPY
99,17
env
AFV81777.1
*peptide found at the HLA Ligand Atlas eluted from normal tissues.

In vitro analysis of binding affinity and stability to HLA-A*02:01 molecule

The confirmation of predicted binding of peptides to HLA alleles could be experimentally confirmed only for the A*0201. The TAP-deficient T2 cell line was loaded with the individual peptides from paired TAA and HIV peptides. In particular, the VVAGIVVLV TAA derived from the colon ca-associated EPCAM protein was compared with the VVAGIIALV and VIAGIVALV epitopes derived from the HIV-1 vpu protein. The analysis confirmed that each peptide bound the HLA-A*02:01 molecule inducing a dose-dependent increase in the HLA surface expression on T2 cells over the background.
The results confirmed the comparable binding to HLA-A*02:01 of the three peptides as suggested by the predicted affinity values (Fig. 3A). Likewise, the peptide—MHC dissociation kinetics showed that the 50% dissociation value (Thalf) was reached 4 h after the peptide loading (T4), for the EPCAM TAA, and 6 – 8 h after the peptide loading (T8) for the two HIV peptides (Fig. 3B and C). Such results were representative of affinity and stability of the other HLA-A*02:01 associate peptides (data not shown).

HIV peptide sequence conservation across viral isolates

HIV-1 is characterized by an extensive intra- and inter-host genome variability with generation of molecular “quasi-species” [23]. Therefore, the biological significance of the described homology between TAAs and HIV peptides is relevant only if the amino acid sequences are conserved across viral isolates. In order to assess such a sequence conservation, about 5000 HIV-1 sequences deposited in Blast were aligned to generate sequence logos. Indeed, logos provide the consensus sequence of all aligned peptides sequences as well as the relative frequency of amino acids at every position of the peptides. The results showed that the HIV peptides with the highest homology to TAAs identified in breast (CCR9), colon (EPCAM, GLOD5, CEACAM8) as well as prostate (NDUFS2) cancer and linked to most frequent HLA alleles (01:01; 02:01 and 24:02), are highly conserved across HIV isolates (Additional file 1: Fig. S5). In particular, HIV peptides RLRDLLFLV and NYTEIIHTL (homologous to breast CCR9) and VVAGIIALV (homologous to colon EPCAM) show 8 of 9 amino acids conserved in the consensus sequence. The HIV peptide SSDNSSRPY (homologous to prostate NDUFS2) shows identity in 7 of 9 amino acids compared to the consensus sequence.
Additional HIV peptides homologous to TAAs from breast and colon cancer and linked to less frequent HLA alleles show high conservation across HIV isolates. Finally, similar results are observed for the HIV peptide IPRRTTQRI (homologous to brain DPYSL2) showing identity in 7 of 9 amino acids compared to the consensus sequence. On the contrary, HIV peptides homologous to lung TAAs show poor identity (3 or 5 of 9 amino acids) to the consensus sequence (Additional file 1: Fig. S6).

Peptide modelling and molecular docking

In order to verify that predicted paired TAAs and HIV-1 epitopes, including the HIV-1 consensus sequences, share contact residues with both the HLA molecule and the TCR α and β chains, epitope modelling and molecular docking were performed.
Epitopes crystallized with HLA alleles showing sequence homology with TAAs or HIV-1 peptides described in the present study were not found and examples of epitopes were used as templates to conduct the analyses. Crystallized pMHC complexes including also the TCR chains were available for the HLA-A*02:01, HLA-A*01:01, B*07:02, B*08:01.
As predictable from the sequence homology, the HIV peptides homologous to TAAs derived from breast, colon and prostate cancers showed highly similar, if not identical, conformation and contact points with HLA molecule and TCR chains as compared to corresponding TAAs. In detail, the breast CCR9 CYTIIIHTL and the HIV- 1 NYTEIIHTL show a conservative Cys to Asn and a non-conservative Ile to Glu substitutions in p1 and p4, respectively. The colon EPCAM VVAGIVVLV and the HIV- 1 VVAGIIALV show conservative Ile to Val and Val to Ala substitutions in p6 and p7, respectively. The prostate NDFUS2 VSDGSSRPY and the HIV-1 SSDNSSRPY show non-conservative Val to Ser and Gly to Asn substitutions in p1 and p4, respectively. They all do not induce significant conformational changes in the structure of paired epitopes. Moreover, the consensus sequences highly conserved across HIV isolates showed identical structure to individual HIV-1 peptides. In detail, the CCR9-like NYTEIIHTL peptide and the consensus NYTEIIYTL show a single non-conservative His to Tyr substitution in p7. The EPCAM-like VVAGIIALV peptide and the consensus VVAAIIALV show a single conservative Gly to Ala substitution in p4. The NDFUS2-like SSDNSSRPY peptide and the consensus SSNSSSRPY show a non-conservative Asp to Asn and a conservative Asn to Ser substitutions in p3 and p4, respectively (Fig. 4).
On the contrary, HIV peptides homologous to TAAs derived from brain and lung cancers did not show such high conformational similarities. In particular, the brain DPYSL2 IPRRTTQRI and the HIV-1 IPRRIRQRI show two non-conservative Thr to Ile and Thr to Arg substitutions in p5 and p6, respectively. The lung CNIH4 LLNLPVATW and the HIV-1 HCNLSVATW show three non-conservative Leu to His, Leu to Cys and Pro to Ser substitutions in p1, p2 and p5, respectively. They all induce significant conformational changes in the structure of paired epitopes. Moreover, the HIV consensus sequences showed structures significantly different from the individual HIV-1 peptides. In detail, the DPYSL2-like IPRRIRQRI peptide and the consensus IPRRLRQRV show two conservative Ile to Leu and Ile to Val substitutions in p5 and p9, respectively. The CNIH4-like HCNLSVATW peptide and the consensus SCNTSVITQ show three non- conservative His to Ser; Leu to Thr; Trp to Gln substitutions in p1, p4 and p9, respectively; and a conservative Ala to Ile substitution in p7 (Fig. 4). The Los Alamos HIV immunological database was interrogated for experimentally validated HIV epitopes showing sequence homology to those paired to TAAs. Indeed, validated epitopes with such characteristics were found for the breast ca CCR9 CYTIIIHTL epitope (NYTDIIYRL) [24], the brain ca DPYSL2 IPRRTTQRI epitope (IPRRIRQGL) [25] and the colon ca EPCAM VVAGIVVLV epitope (VVAAIIAIV) [26] (Fig. 4). In particular, the latter validated HIV epitope shows a relevant structural and conformational homology to the EPCAM TAA, confirming the biological relevance of the predictive findings. All other HIV peptides homologous to TAAs showed limited and variable similarity in conformation and contact points with HLA molecule and TCR chains as compared to corresponding TAAs (Additional file 1: Fig. S7–S9).

Identification of cross-reactive epitope-specific CD8 + T cells

In order to confirm the antigenicity of the predicted peptides and the cross-reactivity with the paired TAAs and HIV-1 peptides, we performed an analysis to detect reactive CD8+ specific T cells in PBMCs from HIV patients and healthy subjects (Additional file 1: Fig. S10).
An “ex vivo” expansion of HIV-specific T cells was induced culturing PBMCs with HIV peptides for 7 days. Subsequently, cells were incubated with tetramers loaded with the corresponding TAAs and analysed at cytofluorimeter.
Given the overwhelming prevalence of HLA-A*02:01 and 24:02 alleles in the cohort of enrolled HIV patients (16/22, 73%) (Additional file 2: Table S1), the analysis was performed with the paired HIV/TAA peptides from the EPCAM colon ca (HLA-A*02:01) and from the CCR9 breast ca (HLA-A*24:01) proteins.
Strikingly, results show that HIV-positive subjects cross-react against HLA-A matched TAAs at baseline, even without any prior “ex vivo” expansion, while HIV-negative subjects do not (Fig. 5A and C). Such cross-reactivity is further increased only in HIV-positive subjects after the “ex vivo” expansion with the paired HIV peptide (Fig. 5B and D). The difference between the HIV-positive and -negative groups reaches the statistical significance for the EPCAM colon cancer HLA-A*02:01 paired peptides. The limited number of HLA-A*24:02 HIV-positive subjects does not provide the sufficient statistical power for the trend observed with CCR9 breast cancer paired peptides. The increased cross-reactivity against the TAA in HIV-positive subjects, prior and after the “ex vivo” expansion with HIV peptides, shows a clear trend that does not reach the statistical significance (Fig. 5E–L). No cross-reactivity was observed when T cells from HIV patients were reacted with HLA-matched not-homologous epitope (data not shown).

Discussion

In the present study we aimed at identifying evidences for a molecular mimicry possibly explaining the strikingly reduced occurrence of breast, prostate and colon cancers in PLWHA reported by several meta-analyses. Indeed, a sequence and structural homology between HIV epitopes and tumor-antigens expressed by such cancers would provide an experimental proof of immunological cross-reactivity. Ultimately, the HIV infection would represent a natural “anti-cancer vaccination” eliciting a memory T cells response able to cross-react with tumor antigens and prevent/delay tumor growth.
The entire Normal Tissue and the Pathology datasets at the Human Protein Atlas (15,170 and 15,313 unique proteins, respectively) were sequentially interrogated to identify relevant cancer-specific proteins. In particular, such proteins should be highly expressed in cancer cells, not expressed in the normal cellular counterpart as well as not detected in > 70% of normal cells of any tissue. The latter characteristic is relevant, considering the aim of focusing on target antigens specifically expressed on cancer cells and not on normal cells. According to such parameters, six cancer-specific proteins in colon cancer (CEACAM8, CDH17, GLOD5, PPM1E, TRIM16, EPCAM), one in breast cancer (CCR9) and none in prostate cancer were identified. In parallel, no cancer-specific proteins matching such characteristics were identified in brain and lung cancers, selected as control tumors that do not show reduced occurrence in PLWHA.
The NetMHCpan software was used to predict strong binders (SBs) to the 12 HLA alleles that altogether cover more than 70% of the World population (http://​www.​allelefrequencie​s.​net). Only SBs with a very high affinity to HLA molecules (< 100 nM) have been selected for the present analysis, according to our previous studies [11, 12, 16]. A total of 298 SBs were predicted from the cancer-specific proteins selected for each cancer, with a high variability among cancers and the highest number was predicted for the HLA-A*02:01 allele. The vast majority (93.7%) of such SBs have never been identified in the HLA ligandome of normal cells, suggesting they should be considered bona fide tumor-associated antigens (TAAs).
The homology search in BLAST returned 25 HIV-1 peptides with high binding affinity to HLA alleles, showing sequence homology to predicted TAAs. Ninenteen (76%) of these HIV-1 peptides were identified for colon [11], breast [6] and prostate [2] cancers. Only 5 were identified in the control lung [5] and brain [1] cancers. More importantly, only HIV peptides homologous to colon-, breast- and prostate-specific TAAs showed strong binding to HLA-A alleles (01:01; 02:01; 03:01 and 24:02) which are the most frequent in the world population. Experimental binding and stability assays in TAP-deficient T2 cells confirmed the predictive bioinformatics analysis. These results show that a high number of HIV-1 epitopes, homologous to TAAs, are found only for the three cancers with reduced occurrence in PLWHA. Moreover, they are associated with the most frequent HLA alleles, supporting the concept that their immunological protective role would cover most of the World population. In addition, the biological relevance of such observation is further supported by the sequence logos showing that TAAs-like HIV peptides in colon, breast and prostate cancers are highly conserved across HIV isolates. Indeed, this would confirm that the immune response elicited by HIV peptides would have an anti-cancer protective role regardless the high genetic variability of HIV-1 isolates.
Epitope modelling and molecular docking bioinformatics analyses confirmed that HIV peptides, and the corresponding consensus sequences, homologous to TAAs derived from breast, colon and prostate cancers showed highly similar, if not identical, conformation and contact points with HLA molecule and TCR chains as compared to corresponding TAAs. Such a similarity was not observed for the HIV peptides homologous to TAAs derived from lung and brain cancers.
The cross-reactivity to TAAs was ultimately demonstrated in ex vivo binding assays. Indeed, only PBMCs from HIV-positive patients, and not those from HIV-negative controls, reacted against HLA-matched TAAs. This confirmed that only HIV-positive patients have a circulating memory T cell population elicited by HIV peptides homologous to cancer-specific TAAs. The identification of validated HIV epitopes with a relevant structural and conformational homology to predicted epitopes, strongly confirms the biological relevance of our findings.
Overall, the present study shows for the first time the molecular mimicry between HIV epitopes and TAAs derived from cancer-specific proteins associated with colon, breast and prostate cancers that show reduced occurrence in PLWHA. All the experimental evidences strongly suggest that this could be, in fact, the immunological explanation for such epidemiological observation. Therefore, HIV infection may represent a “natural anti-cancer vaccination” eliciting a memory T cell compartment able to cross-react with cancer cells and providing a protection from cancer development and progression. This represents a breakthrough in cancer immunology with highly relevant implications beyond HIV infection.
Two independent studies have recently reported evidence for cancer regression in SARS-CoV2 infected or vaccinated individuals. No experimental evidence for SARS-CoV2 epitopes sharing sequence or structural homology with TAAs has been shown. Nevertheless, such clinical evidences support for a broader interplay between viral and cancer T cell responses [27, 28].
Other more ubiquitous viral infections may play a role as “natural anti-cancer vaccination” and a much larger population may be protected from development and progression of different cancers. In addition, viral antigens homologous to TAAs may be used in anti-cancer preventive/therapeutic vaccine formulations with higher antigenicity and immunogenicity than over-expressed tumor self-antigens.
The present study ultimately provides a definitive explanation of the reduced occurrence of breast, prostate and colon cancers in PLWHA and represents a new way of thinking in cancer immunology.

Contribution to the field statement

Evidence before this study

We and others have previously shown that the molecular mimicry between viral and tumor antigens may have a significant impact in controlling tumor growth and improving the clinical outcome in cancer patients. More recently, we have shown that TAAs may show sequence homology as well as structural similarities with viral peptides and cross-reacting CD8+ T cells can be identified to drive the fate of cancer development and progression. In particular, such homologies must be found at the residues facing the T cell receptor (TCR). Therefore, a previous viral infection may represent a natural “anti-cancer vaccination”.

Added value of this study

This is the first study showing a molecular mimicry between HIV antigens and TAAs identified in breast, prostate and colon cancers. T cells cross-reacting with paired antigens has been demonstrated.

Implications of all the available evidence

Therefore, it is highly reasonable that memory CD8+ T cells elicited during the HIV infection may play a key role in controlling development and progression of such cancers in the PLWHA lifetime. This represents the first demonstration ever that a viral infection may induce natural “preventive” anti-cancer memory T cells, with highly relevant implications beyond the HIV infection.

Acknowledgements

We thank patients for their willingness to participate.

Declarations

Approval and consent were obtained at the AORN San Pio Hospital, Benevento, Italy.
This was obtained by each participant at the AORN San Pio Hospital, Benevento, Italy.

Competing interests

No competing interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
19.
Metadaten
Titel
Antigenic molecular mimicry in viral-mediated protection from cancer: the HIV case
verfasst von
Carmen Manolio
Concetta Ragone
Beatrice Cavalluzzo
Angela Mauriello
Maria Lina Tornesello
Franco M. Buonaguro
Angelo Salomone Megna
Giovanna D’Alessio
Roberta Penta
Maria Tagliamonte
Luigi Buonaguro
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2022
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03681-4

Weitere Artikel der Ausgabe 1/2022

Journal of Translational Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.