A METHOD TO MONITOR VIRUS-SPECIFIC T CELLS IN BIOLOGICAL SAMPLES

Information

  • Patent Application
  • 20230393121
  • Publication Number
    20230393121
  • Date Filed
    October 18, 2021
    2 years ago
  • Date Published
    December 07, 2023
    5 months ago
Abstract
The present invention relates to a method of diagnosing and/or monitoring of virus infection and/or response to vaccination by generating a profile of a virus-specific T cell response that can (i) discriminate between virus infected and uninfected individuals, (ii) determine the effect of vaccination on T cell response, and (iii) determine the effect of viral variants on T cell response. More particularly, the described virus-specific T cell profiling is based on the detection of activated antigen-specific T lymphocytes responding to pools of selected short peptides from virus proteins. These peptide sequences have been selected for their immunogenicity. The profiling is typically performed using ELISPOT, but may also be performed using other techniques such as qPCR, more particularly direct qPCR. The present invention also includes kits for use in the methods of the invention.
Description
FIELD OF THE INVENTION

The present invention relates to a method of diagnosing and/or monitoring of virus infection and/or response to vaccination by generating a profile of a virus-specific T cell response that can (i) discriminate between virus infected and uninfected individuals, (ii) determine the effect of vaccination on T cell response, and (iii) determine the effect of viral variants on T cell response. More particularly, the described virus-specific T cell profiling is based on the detection of activated antigen-specific T lymphocytes responding to pools of selected short peptides from virus proteins. These peptide sequences have been selected for their immunogenicity. The profiling is typically performed using ELISPOT, but may also be performed using other techniques such as qPCR, more particularly direct qPCR. The present invention also includes kits for use in the methods of the invention.


BACKGROUND OF THE INVENTION

The control and the long-term protection against viral infections require the coordinated activation of humoral (antibodies) and cellular (T cells) immunity. Viruses are intracellular pathogens, and CD8 T cells are necessary to recognize and lyse the infected cells. In addition, CD4 helper T cells are necessary to boost the maturation of antibody production.


Despite this important role in antiviral immunity, quantification of virus-specific T cells is technically complex in comparison to methods of antibody detection. As such virus-specific T cells are not routinely measured as a potential correlate of protection.


This is particularly problematic in the present COVID-19 pandemic, since measuring SARS-CoV-2-specific antibodies might not be sufficient to fully gauge the level of antiviral immunity induced by the infection.


The inventors and others have recently shown that SARS-CoV-2-specific T cells are present in 100% of COVID-19 convalescents (Grifoni A., et al., Cell Host Microbe 27(4): 671-680 (2020); Braun J., et al., 2020 medRxiv 1-12. doi:10.1101/2020.04.17.20061440; Dong T., et al., 2020 bioRxiv 1-36. doi:10.1101/2020.06.05.134551; Le Bert N., et al., Nature 2020; 584(7821): 457-62), while observations of SARS-CoV-2 infection with fading antibody titers have also been reported (Long Q.-X., et al., 2020 Nat Med 1-15. doi:10.1038/s41591-020-0897-1). A discrepancy between virus-specific T cell and antibody response was also reported in infections with the Middle East Respiratory Syndrome (MERS), where a quarter of MERS-infected patients showed only MERS-specific T cells (Zhao J., et al., 2017 Science Immunology 2, eaan5393. doi:10.1126/sciimmunol.aan5393).


However, quantification of SARS-CoV-2-specific T cells and, indeed, other virus-specific T cells in the general population is rarely performed since the methods of detection are complex and cumbersome and require very specific technical personnel and assays that require an initial step of separation of peripheral blood mononuclear cells (PBMC) from whole blood and experimental procedures (ELISPOT or Intracellular cytokine staining or qPCR) that necessitate specialized skills and equipment.


There is a need for a test that indicates a patient's level of virus-specific cellular immunity.


SUMMARY OF THE INVENTION

The present invention provides methods to quantify virus-specific T cell activation, which have several applications.

    • 1. A method of discriminating past or currently virus-infected subjects from virus un-infected subjects;
    • 2. A method of testing T cell responses in vaccinated subjects; and
    • 3. A method of testing whether T cells previously exposed to a virus or vaccine are activated by a virus variant, particularly a variant of concern (VOC).


SARS-CoV-2 and HBV are exemplified herein.


In a first aspect the invention provides an in vitro method of discriminating past or currently virus-infected subjects from virus un-infected subjects, comprising:

    • assaying a sample comprising or derived from blood, broncholavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject to determine whether it comprises T cells reactive to one or more virus peptide pools, wherein said peptide pools are separately derived from virus antigenic structural and/or non-structural proteins, wherein;
    • (a) if the sample T cells are reactive to a majority of the peptide pools derived from the virus antigenic proteins, in comparison to unstimulated or DMSO treated cells, the subject is identified as past or currently infected by the virus, or
    • (b) if the sample T cells are reactive to 0, or a minority of the peptide pools derived from the virus antigenic proteins, in comparison to unstimulated or DMSO treated cells, the subject is identified as having been uninfected by the virus.


In some embodiments the virus is an enveloped virus. The antigenic structural and non-structural proteins of these viruses are well-known. An example is the membrane (M), nucleoprotein (NP) and/or Spike (S) proteins of an enveloped virus such as a coronavirus. Other enveloped viruses include Hepatitis B virus (HBV), wherein the antigenic proteins are Polymerase (Pol), Envelope (E), Core (C) and X.


In some embodiments of any aspect of the invention, the virus is a coronavirus, such as SARS, MERS or SARS-CoV-2.


In some embodiments, the virus is SARS-CoV-2 or HBV or variant thereof.


In a second aspect the invention provides an in vitro method of determining whether a vaccinee or previously virus-infected subject has T cells whose activation may be reduced by a virus variant, such as a variant of concern (VOC), comprising:

    • assaying a sample comprising or derived from blood, bronchoalveolar lavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject to determine whether it comprises T cells reactive to one or more virus peptide pools, wherein said peptide pools are separately derived from (A) the whole virus antigenic protein present in the vaccine or corresponding to an antigenic protein from the virus that infected the subject, (B) non-conserved regions of said virus antigenic protein that are mutated in the virus variant, and (C) virus variant mutated non-conserved regions of the vaccine antigenic protein or corresponding to an antigenic protein from the virus that infected the subject, wherein;
    • the number or proportion of reactive T cells present in each pool is analyzed and utilized to derive in each single individual, the frequency of T cells directed towards the whole virus antigenic protein (PBMC stimulated with peptide pool A), the frequency of T cells directed toward non-conserved regions of said virus antigenic protein that are mutated in the virus variant (PBMC stimulated with Pool B) and the frequency of T cells inhibited by amino acid mutations present in virus variant mutated non-conserved regions (PBMC stimulated with pool C), wherein;
    • (a) if the sample T cells are reactive to peptide pool A, the subject has T cells responsive against the virus antigenic protein, and
    • (b) if the sample T cells are similarly reactive to pool B and pool C, the impact of the amino acid mutations in the variant are negligible on the total T cell response against the said virus antigenic protein;
    • (c) if the sample T cells react differently to pool B and pool C, the impact of the amino acid mutations in the variant on the total T cell response against the said virus antigenic protein can be estimated by the proportion of pool C against pool B response,
    • wherein the method provides an estimation of the ability of T cells of the subject to recognize the conserved and non-conserved region of different vaccine antigenic proteins or virus that infected the subject, and of the ability of mutations to reduce the T cell response towards variants.


In a third aspect the invention provides a method to quantify the presence of virus-specific T cells in a biological sample comprising or derived from blood, bronchoalveolar lavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject, comprising;

    • a) Mixing the biological sample with one or more virus peptide pools, wherein said peptide pools are separately derived from virus antigenic structural and/or non-structural proteins;
    • b) Incubating the mixture formed for a period to allow T cell activation;
    • c) Rupture the cells from b);
    • d) Aliquot a sample from c) into PCR reagents, ACTIN (or other internal control) forward and reverse primers, ACTIN (or other internal control) probe, CXCL10 forward and reverse primers and CXCL10 probe for dqPCR; and/or
    • e) Extract RNA from a sample from c) and add a portion into PCR reagents, ACTIN (or other internal control) forward and reverse primers, ACTIN (or other internal control) probe, CXCL10 forward and reverse primers and CXCL10 probe for qPCR;
    • f) Perform cycles of dqPCR and/or qPCR for d) and e), respectively; and
    • g) Quantitate the expression of CXCL10 in the sample and compare with a control,
    • wherein an elevated CXCL10 level indicates the presence of virus-specific T cells in the subject sample.


It would be understood that whether a single peptide pool is sufficient for an assay method may depend on whether a subject has only been exposed to a part of the virus (e.g. spike protein vaccine) or a whole virus (virus-infected). Using a peptide pool specific against a single viral protein only informs the response against that particular protein. Therefore, to know if an individual was infected before using the PCR method, one would still need to assess the response against multiple proteins. This logic does not change with different readouts like PCR, ELISPOT or cytokine release assay.


In a fourth aspect the invention provides a method of treatment comprising administering, to a subject with T cells reactive to a majority of peptide pools derived from virus antigenic proteins, an effective amount of a virus inhibitor.


In some embodiments of the method of treatment, the peptide pools are selected from:

    • i) one or more M, NP and S pools for a coronavirus, or
    • ii) one or more C, Pol, X, and E pools for HBV.


In some embodiments of the method of treatment, the peptide pools are selected from:

    • i) one or more M, NP and S pools listed in Tables 1-4 for SARS-CoV-2, or ii) one or more C, Pol, X, and E pools listed in Tables 20-27 for HBV.


In a fifth aspect the invention provides a method of prophylaxis comprising administering, to a subject with T cells reactive to a minority of peptide pools derived from virus antigenic proteins, an effective amount of a virus vaccine.


In a sixth aspect the invention provides a method of monitoring the efficacy of a virus vaccine, comprising testing whether the recipient of said vaccine has T cells reactive to a minority or majority of peptide pools derived from said virus antigenic proteins.


In a seventh aspect the invention provides a kit to discriminate past or currently virus-infected subjects from virus un-infected subjects, the kit comprising a plurality of virus antigenic peptides that stimulate virus-exposed T cells, wherein the virus antigenic peptides are in peptide pools derived from:

    • I) M, NP and/or S proteins or
    • ii) C, Pol, X and/or E proteins.


In some embodiments,

    • i) the M protein comprises the amino acid sequence set forth in SEQ ID NO: 793;
    • ii) the NP protein comprises the amino acid sequence set forth in SEQ ID NO: 794;
    • iii) the S protein comprises the amino acid sequence set forth in SEQ ID NO: 795;
    • iv) the C protein comprises the amino acid sequence set forth in SEQ ID NO: 798;
    • v) the E protein comprises the amino acid sequence set forth in SEQ ID NO: 797;
    • vi) the X protein comprises the amino acid sequence set forth in SEQ ID NO: 799;
    • vii) the Pol protein comprises the amino acid sequence set forth in SEQ ID NO: 796.


In some embodiments the kit can quantify virus-specific T cell activation in an isolated patient sample, comprising one or more peptide pools, wherein said peptide pools are separately derived from virus antigenic proteins, such as membrane (M), nucleoprotein (NP) and/or Spike (S) proteins; or Core (C), Polymerase (Pol), X and/or Envelope (E) proteins.


In some embodiments, the kit further comprises:

    • i) PCR reagents; and/or
    • ii) primers and probes to detect CXCL10 and/or IFN-gamma expression by stimulated T cells.


In an eighth aspect the invention provides a set of 2 to 4 separate pools of peptides suitable to discriminate;

    • a) past or currently SARS-CoV-2-infected subjects from SARS-CoV-2 un-infected subjects;
    • b) past or currently HBV-infected subjects from HBV un-infected subjects,


      wherein the peptide pools are selected from those listed in Tables 1 to 4 and 8-19 for (a) and Tables 20-27 for (b).


In a ninth aspect the invention provides use of a kit of any one of aspects 7 to 9 in a method according to any one of aspects 1 to 6.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 shows cytokine secretion by T cells reactive to different pools of Spike peptides in COVID-19 convalescents. Spike is a long protein with 1276 amino acids, thus it requires 253 15-mer peptides overlapping by 10 amino acids to cover the whole protein, thus 7 pools of about 40 peptides. To reduce the number of peptides pools to test, we selected a single “Spike pool” comprised of 55 peptides. The 55 peptides cover 40.5% of the Spike protein. The frequency of reactive cells to the selected Spike pool (right) was compared to the 7 pools of 15-mers overlapping by 10 amino acids covering together the entire Spike protein (S1-S7) in 15 COVID-19 convalescents.



FIG. 2 shows a schematic representation of both approaches to profile SARS-CoV-2 specific T cells.



FIG. 3 shows (A) ELISPOT assay of PBMCs with SARS-CoV-2 M, NP1, NP2 and spike peptide pools can discriminate between infected and unexposed individuals; (B) Infected individuals are almost always positive for 3 or more peptide pools, while unexposed individuals occasionally have responses to 1-2 peptide pools. Grey shaded areas denote the threshold of positivity.



FIG. 4 shows (A) The concentration of IFN-γ in the plasma collected from whole blood of uninfected (n=9) and infected (n=6) individuals stimulated with the respective peptide pools was quantified. SARS-CoV-2 specific T cell response profile evaluated using this method can also discriminate between infected and uninfected individuals; (B) Infected individuals are positive for all peptide pools, while unexposed individuals occasionally have responses to 1-2 peptide pools. Grey shaded areas denote the threshold of positivity.



FIG. 5 shows (A) a schematic of how T cell responses against variants of concern (VOC) can be evaluated using the SARS-CoV-2 delta variant as an example. Vertical bar regions refer to amino acid mutations present in the delta variant compared to the wild-type SARS-CoV-2. Pool A contains peptides covering the whole Spike-Wuhan protein. Pool B contains peptides covering the non-conserved Spike-Wuhan regions affected by mutations present in the SARS-CoV-2 delta variant (B.1.617.2). Pool C contains peptides with the delta variant (B.1.617.2) amino acid mutations present in the non-conserved Spike-Wuhan regions. (B) Wells show the ELISPOT results from a vaccinee tested using the peptide pools described in (A). This vaccinee has a strong T cell response against the Spike protein as expected (Pool A, 133 spots/400,000 PBMC) and negligible T cell responses directed towards regions mutated in the delta variant (Pool B, 6 spots/400,000 PBMC). Since responses against these regions are very low, the impact of the amino acid mutations in the delta variant are negligible (Pool C VS Pool B). 2×Negative control wells are shown on the left.



FIG. 6 shows (A). Schematic of workflow for the three T cells activation (TACT) assays described. All assays begin with whole blood collection followed by overnight stimulation with nucleocapsid (NP) or spike (S) peptide pools. For TACTseq (FIG. 6), RNA was extracted and used for NGS using the Illumina system. For qTACT (FIG. 2), RNA was extracted and probe-based qPCR was performed using the BioRad CFX96/384 or Hyris bCUBE 2.0. For dqTACT (FIG. 3), blood was diluted and used directly for qPCR using the Hyris bCUBE 2.0. (B). TACTseq assay. Number of differentially expressed genes stimulated in whole blood by each peptide pool versus DMSO, grouped by subject COVID status. Significantly differentially expressed genes were defined as having p-value<0.05 and log 2FC>1. P-values were calculated using DESeq2 and adjusted using the Benjamini-Hochberg method. (C). Candidate genes selected for downstream validation based on differential expression versus DMSO, grouped by subject COVID status. Comparisons show significance calculated using DESeq2 and corrected using the Benjamini-Hochberg method.



FIG. 7 shows candidate genes selected for downstream validation based on differential expression versus DMSO. Comparisons show significance calculated using DESeq2 and corrected using the Benjamini-Hochberg method.



FIG. 8 shows (A). qPCR validation of 3 target genes (CXCL10, IFNG, IL2) on 11 naïve and 8 COVID-19 convalescent individuals. Results are plotted as gene expression relative to ACTIN minus DMSO control. Samples were run on the Hyris bCUBE 2.0. Comparisons show significance for the Wilcoxon Rank Sum two-sided test, corrected using the Benjamini-Hochberg method (*p<=0.05, **p<=0.01, ***p<=0.001, ****p<=0.0001). (B). Correlation between independent CXCL10, IFNG and ANKRD22 qPCR runs using samples from naïve and COVID-19 convalescent individuals treated with the spike protein. Values are calculated by comparing the final result (gene expression relative to ACTIN minus DMSO control) between all combinations of runs. Samples were run on the BioRad CFX96 or CFX384.



FIG. 9 shows qTACT assay. (A). Normalized CXCL10 mRNA expression (relative to ACTIN minus DMSO) before, 10 days, and 20 days after the first and second vaccine doses in SARS-CoV-2 naïve (black) and previously infected (grey) individuals. The qTACT assay was completed as shown in FIG. 6A (middle). The samples were run on a BioRad CFX384. Comparisons show significance for the Wilcoxon Rank Sum two-sided test, corrected using the Benjamini-Hochberg method (*p<=0.05, **p<=0.01, ***p<=0.001, ****p<=0.0001). (B). Normalized IFNG mRNA expression (relative to ACTIN minus DMSO) before, 10 days, and 20 days after the first and second vaccine doses in SARS-CoV-2 naïve (black) and previously infected (grey) individuals. The qTACT assay was completed as shown in FIG. 6A (middle). The samples were run on the Hyris bCUBE 2.0. Comparisons show significance for the Wilcoxon Rank Sum two-sided test, corrected using the Benjamini-Hochberg method (*p<=0.05, **p<=0.01, ***p<=0.001, ****p<=0.0001).



FIG. 10 shows (A). Correlation between CXCL10 mRNA expression (determined by the qTACT assay) and IFN-γ protein secretion (determined by ELLA) for the cohort described in FIG. 9 (B). Correlation between IFNG mRNA expression (determined by the qTACT assay) and IFN-γ protein secretion (determined by ELLA) for the cohort described in FIG. 9.



FIG. 11 shows a dqTACT assay. (A). Image showing all reagents and equipment needed to perform the dqTACT assay. With appropriate biosafety level 2 requirements and a cell culture incubator, this is the minimum reagents and equipment required to run the dqTACT assay which include in clockwise order: the Hyris bCube 2.0 qPCR machine, pipettes and tips, a heparin coated blood collection tube, Hyris 16/32 well cartridges, nuclease-free water, Jena Bioscience SCRIPT direct RT-qPCR ProbesMaster mix, PCR primer/probes, and RPMI medium. (B). Normalized CXCL10 expression (peptides stimulated relative to ACTIN minus DMSO control) of naïve and COVID-19 vaccinated individuals (FIG. 9). The dqTACT assay was completed as shown in FIG. 6A (bottom). Comparisons show significance for the Wilcoxon Rank Sum two-sided test (*p<=0.05, **p<=0.01, ***p<=0.001, ****p<=0.0001). (C). Normalized CXCL10 secretion as quantified by ELLA (peptides stimulated minus DMSO control) of naïve and COVID-19 vaccinated individuals. Comparisons show significance for the Wilcoxon Rank Sum two-sided test (*p<=0.05, **p<=0.01, ***p<=0.001, ****p<=0.0001). (D). Normalized CXCL10 expression (peptides stimulated relative to ACTIN minus DMSO control) of subjects enrolled in the CombiVacS trial. All subjects received a first dose of ChAdOx1s from AstraZeneca (Dose 1 (AZ)). The patients were then divided into two groups and received either a second placebo dose (Dose 1 (AZ)+Dose 2 (placebo)) or a second dose of BNT162b2 from Pfizer (Dose 1 (AZ)+Dose 2 (Pfizer)). The dqTACT assay was completed as shown in FIG. 6A (bottom). Comparisons show significance for the Wilcoxon Rank Sum two-sided test (*p<=0.05, **p<=0.01, ***p<=0.001, ****p<=0.0001).



FIG. 12 (A-C) show results from IFN-γ, IL2, and TNFα ELLA showing normalized protein secretion (minus DMSO control) of naïve and COVID-19 vaccinated individuals. Comparisons show significance for the Wilcoxon Rank Sum two-sided test, corrected using the Benjamini-Hochberg method (*p<=0.05, **p<=0.01, ***p<=0.001, ****p<=0.0001). (D-G) show correlation between data shown in A-C and FIG. 11C (CXCL10 mRNA quantified by dqTACT vs. CXCL10, IFN-γ, IL2, and TNFα protein quantified by ELLA). Correlation coefficients and p-values were calculated using the Spearman method.



FIG. 13 shows the application of a whole blood cytokine release assay for the detection of HBV-specific T cells. A) Schematic showing the 4 proteins of Hepatitis B virus and the corresponding peptide pools. B) Whole blood cytokine release assay performed by stimulation of whole blood from a chronically infected HBV patient and a vaccinated individual with the corresponding peptide pool. IFN-γ and IL-2 secretion was measure by ELLA. The level of cytokines present in the plasma of DMSO controls was subtracted from the corresponding peptide pool stimulated samples.





DEFINITIONS

Certain terms employed in the specification, examples and appended claims are collected here for convenience.


As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise.


The terms “amino acid” or “amino acid sequence,” as used herein, refer to an oligopeptide, peptide, polypeptide, or protein sequence, or a fragment of any of these, and to naturally occurring or synthetic molecules. Where “amino acid sequence” is recited herein to refer to an amino acid sequence of a naturally occurring protein molecule, “amino acid sequence” and like terms are not meant to limit the amino acid sequence to the complete native amino acid sequence associated with the recited protein molecule.


As used herein, the term “comprising” or “including” is to be interpreted as specifying the presence of the stated features, integers, steps or components as referred to, but does not preclude the presence or addition of one or more features, integers, steps or components, or groups thereof. However, in context with the present disclosure, the term “comprising” or “including” also includes “consisting of”. The variations of the word “comprising”, such as “comprise” and “comprises”, and “including”, such as “include” and “includes”, have correspondingly varied meanings.


As used herein, the term ‘majority’ refers to a value over 50%. Conversely, the term ‘minority’ refers to a value under 50%. For example, if 3 or 4 of a total of 4 peptide pools positively activate T cells in a sample, compared to a control, a majority of pools are positive and the sample is indicative of the subject having been exposed to SARS-CoV-2 infection. When 4 pools M, NP1, NP2 and S were used, 50% or more (2, 3, or 4 pools out of 4) positive pools was considered to indicate the subject had been infected by SARS-CoV-2.


As used herein, the term ‘a subject uninfected by SARS-CoV-2’ refers to a subject who is considered to not have been exposed to and infected by SARS-CoV-2, for the purpose of the invention.


Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is not to be taken as an admission that any or all of these matters form part of the prior art base, or were common general knowledge in the field relevant to the present disclosure as it existed before the priority date of each of the appended claims.


Bibliographic references mentioned in the present specification are for convenience listed at the end of the examples. The whole content of such bibliographic references are herein incorporated by reference.


DETAILED DESCRIPTION OF THE INVENTION

The present invention relates to a method of discriminating past or currently virus-infected subjects from virus un-infected subjects, based on the detection of activated antigen-specific T lymphocytes responding to selected peptide sequences from the virus in an isolated sample from said individual. These peptide sequences are selected for their immunogenicity and are represented in peptide pools separately derived from virus antigenic proteins, such as membrane (M), nucleoprotein (NP) and/or Spike (S) proteins; or Polymerase (Pol), Envelope (E), Core (C) and X proteins.


In addition, the present invention provides a method of testing T cell responses in vaccinated subjects. In this way, the efficacy of a vaccine to stimulate a T cell response can be determined.


Further, the invention provides a method of testing the impact of amino acid mutations in a virus strain on T cells that have previously been exposed to a parent or comparator strain. For example, different variants of concern (VOC) of SARS-CoV-2 have replaced world-wide the original SARS-CoV-2 Wuhan isolate. These VOCs are characterized by amino acid substitutions that provide biological advantages such as increased infectivity or escape humoral (Antibodies) but also cellular (T cells) immunity. We have designed a method based on specific combination of peptide pools covering both different SARS-CoV-2 proteins (i.e., Spike) and the corresponding regions affected by the amino acid mutations that are used to stimulate T cells, The results of this multiple stimulation strategy define with accuracy the impact of these amino acid mutations on the SARS-CoV-2-specific T cells induced by infection with Wuhan strain or by vaccination with the present vaccines based on the Wuhan strain. Presented herein as an example is a method to analyze the impact of amino acid mutations present in Spike in different VOCs (using Delta variant as an example) on the SPIKE specific T cells induced by vaccination or previous infection.


Discrimination can be achieved based on the number or proportion of peptide pools that stimulate the isolated T cells above a control value.


Each of the M, NP and S, or Pol, C, E and X, proteins may contribute at least one pool of immunogenic peptides. For example, the NP protein consists of 419 amino acids. It is possible that the NP protein could be divided into more than one pool comprising 15-mer peptides, such as 2 pools where 1 pool comprises 15-mer peptides which overlap adjacent peptides by 10 amino acids covering amino acids 1-215; and a second pool comprising 15-mers which overlap adjacent peptides by 10 amino acids covering amino acids 216-419. The peptide overlap can be seen, for example, in the NP peptides listed sequentially in Table 2. It would be understood that the degree of overlap between 15-mer peptides could be varied from 10 without substantially affecting the ability to activate T cells and obtain a valid result. Likewise, the S protein, which is 1273 amino acids in length, could be divided up into 1, 2, 3 or more pools of 15-mer peptides. The number and size of the pools needs to be balanced with practical considerations, such as the amount of blood sample available, the cost of generating peptide pools, and whether the number of pools improves discrimination.


In a first aspect the invention provides an in vitro method of discriminating past or currently virus-infected subjects from virus un-infected subjects, comprising:

    • assaying a sample comprising or derived from blood, broncholavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject to determine whether it comprises T cells reactive to one or more virus peptide pools, wherein said peptide pools are separately derived from virus antigenic structural and non-structural proteins, wherein;
    • (a) if the sample T cells are reactive to a majority of the peptide pools derived from the virus antigenic proteins, in comparison to unstimulated or DMSO treated cells, the subject is identified as past or currently infected by the virus, or
    • (b) if the sample T cells are reactive to 0, or a minority of the peptide pools derived from the virus antigenic proteins, in comparison to unstimulated or DMSO treated cells, the subject is identified as having been uninfected by the virus.


In some embodiments the virus is an enveloped virus or a non-enveloped virus. The antigenic structural and non-structural proteins of these viruses are well-known. An example is the membrane (M), nucleoprotein (NP) and/or Spike (S) proteins of an enveloped viruses such as coronaviruses, whereas and Hepatitis B viruses (HBV) comprise polymerase (Pol), envelope (E), core (C) and X antigenic structural and non-structural proteins.


In some embodiments of any aspect of the invention, the virus is a coronavirus.


In some embodiments, the virus is a coronavirus, selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, HKU1, OC43, NL63 and 229E or variant thereof.


In some embodiments, the virus is SARS-CoV-2 or variant thereof.


In some embodiments, the virus is HBV or variant thereof.


In some embodiments,

    • ai) an M peptide pool comprises or consists of at least one peptide derived from an M protein comprising the amino acid sequence set forth in SEQ ID NO: 793;
    • ii) an NP peptide pool comprises or consists of at least one peptide derived from an NP protein comprising the amino acid sequence set forth in SEQ ID NO: 794; and
    • iii) an S peptide pool comprises or consists of at least one peptide derived from an S protein comprising the amino acid sequence set forth in SEQ ID NO: 795; or
    • bi) a Pol peptide pool comprises or consists of at least one peptide derived from a Pol protein comprising the amino acid sequence set forth in SEQ ID NO: 796;
    • ii) an E peptide pool comprises or consists of at least one peptide derived from an E protein comprising the amino acid sequence set forth in SEQ ID NO: 797;
    • iii) a C peptide pool comprises or consists of at least one peptide derived from a C protein comprising the amino acid sequence set forth in SEQ ID NO: 798; and
    • iv) an X peptide pool comprises or consists of at least one peptide derived from an X protein comprising the amino acid sequence set forth in SEQ ID NO: 799.


In some embodiments;

    • ai) an M peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 1-43,
    • ii) an NP peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 44-125, and
    • iii) an S peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 126-454.


In some embodiments, the NP peptide pool is divided into 2 pools, NP1 and NP2.


In some embodiments, the S peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 126-180 (Table 4).


In some embodiments NP1 comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 44-84, and the NP2 peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 85-125.


In some embodiments;

    • ai) the M peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 1-43,
    • ii) the NP1 peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 44-84,
    • iii) the NP2 peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 85-125, and
    • iv) the S peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 126-180.


In some embodiments;

    • bi) a Pol peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 626-792 (Tables 24-27),
    • ii) an E peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 550-625 (Tables 22-23);
    • iii) a C peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 480-520 (Table 20); and
    • iv) an X peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 521-549 (Table 21).


In some embodiments, the Pol peptide pool is divided into a plurality of pools, such as 2, 3 or 4 pools of peptides. Preferably, each of the plurality of Pol pools has approximately equal numbers of peptides. In some embodiments, the Pol peptide pool is divided into 4 pools, Pol-1, Pol-2, Pol-3 and Pol-4. An example is shown in Tables 24-27.


In some embodiments, the E peptide pool is divided into 2 pools, E-1 and E-2. An example is shown in Tables 22-23.


In some embodiments,

    • bi) the Pol peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 626-792;
    • ii) the E peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 550-625;
    • iii) the C peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 480-520; and
    • iv) the X peptide pool consists of peptides having the amino acid sequences set forth in SEQ ID Nos: 521-549.


In some embodiments;

    • (a) if the sample T cells are reactive to 3 or 4 of the peptide pools derived from M, NP1, NP2 and S, in comparison to unstimulated or DMSO treated cells, the subject is identified as past or currently infected by SARS-CoV-2, or
    • (b) if the sample T cells are reactive to 0, 1 or 2 of the peptide pools derived from M, NP1, NP2 and S, in comparison to unstimulated or DMSO treated cells, the subject is identified as having been uninfected by SARS-CoV-2.


In some embodiments, the method comprises the steps of:

    • a) mixing the sample with each of said peptide pools to produce:
    • i) assay samples corresponding to M, NP and S; or
    • ii) assay samples corresponding to E, Pol, C and X;
    • b) incubating each mixture for a period to allow for T cell activation;
    • c) measuring the level of at least one secreted cytokine in each said mixture and determining whether the level of at least one secreted cytokine is above a threshold control value to indicate a positive T-cell reaction; and
    • d) counting the number of peptide pools that are positive.


In some embodiments, the method comprises the steps of:

    • a) mixing the sample with each of said peptide pools to produce:
    • i) 4 assay samples corresponding to M, NP1, NP2 and S; or
    • ii) 8 assay samples corresponding to C, Pol1, Po12, Po13, Po14, E1, E2 and X;
    • b) incubating each mixture for a period to allow for T cell activation;
    • c) measuring the level of at least one secreted cytokine in each said mixture and determining whether the level of at least one secreted cytokine is above a threshold control value to indicate a positive T-cell reaction; and
    • d) counting the number of peptide pools that are positive.


In some embodiments, if the sample T cells are reactive to 50% or more of the peptide pools derived from SARS-CoV-2 M, NP and S, in comparison to unstimulated or DMSO-treated cells, the subject is identified as past or currently infected by virus.


In a second aspect the invention provides an in vitro method of determining whether a vaccinee or previously virus-infected subject has T cells whose activation may be reduced by a virus variant, such as a variant of concern (VOC), comprising:

    • assaying a sample comprising or derived from blood, bronchoalveolar lavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject to determine whether it comprises T cells reactive to one or more virus peptide pools, wherein said peptide pools are separately derived from (A) the whole virus antigenic protein present in the vaccine or corresponding to an antigenic protein from the virus that infected the subject, (B) non-conserved regions of said virus antigenic protein that are mutated in the virus variant, and (C) virus variant mutated non-conserved regions of the vaccine antigenic protein or corresponding to an antigenic protein from the virus that infected the subject, wherein;
    • the number or proportion of reactive T cells present in each pool is analyzed and utilized to derive in each single individual, the frequency of T cells directed towards the whole virus antigenic protein (PBMC stimulated with peptide pool A), the frequency of T cells directed toward non-conserved regions of said virus antigenic protein that are mutated in the virus variant (PBMC stimulated with Pool B) and the frequency of T cells inhibited by amino acid mutations present in virus variant mutated non-conserved regions (PBMC stimulated with pool C), wherein;
    • (a) if the sample T cells are reactive to peptide pool A, the subject has T cells responsive against the virus antigenic protein, and
    • (b) if the sample T cells are similarly reactive to pool B and pool C, the impact of the amino acid mutations in the variant are negligible on the total T cell response against the said virus antigenic protein;
    • (c) if the sample T cells reacts differently to pool B and pool C, the impact of the amino acid mutations in the variant on the total T cell response against the said virus antigenic protein can be estimated by the proportion of pool C against pool B response,
      • wherein the method provides an estimation of the ability of T cells of the subject to recognize the conserved and non-conserved region of different vaccine antigenic proteins or virus that infected the subject, and of the ability of mutations to reduce the T cell response towards variants.


In some embodiments, the virus is a coronavirus, selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, KHU1, OC43, NL63 and 229E or variants thereof.


In some embodiments, the virus antigenic protein is an M, NP, or S protein.


In some embodiments, the virus is HBV and the virus antigenic protein is an E, Pol, C or X protein.


In some embodiments, peptide pool A and pool B are derived from a wild-type virus.


It would be understood that peptide pool A and pool B could be derived from a variant if it became a reference point due to becoming endemic or if future vaccines employ the variant sequence instead of the original wildtype virus.


In some embodiments of the method;

    • ai) the M protein comprises the amino acid sequence set forth in SEQ ID NO: 793;
    • ii) the NP protein comprises the amino acid sequence set forth in SEQ ID NO: 794; and
    • iii) the S protein comprises the amino acid sequence set forth in SEQ ID NO: 795; or
    • bi) the Pol protein comprises the amino acid sequence set forth in SEQ ID NO: 796;
    • ii) the E protein comprises the amino acid sequence set forth in SEQ ID NO: 797;
    • iii) the C protein comprises the amino acid sequence set forth in SEQ ID NO: 798; and
    • iv) the X protein comprises the amino acid sequence set forth in SEQ ID NO: 799.


In some embodiments, the wildtype virus is SARS-CoV-2 wildtype and the variant is selected from the group comprising B.1.617.2 (Delta), B.1.1.7 (Alpha V1), B.1.351 (Beta V2), P.1 (Gamma, V3), B.1.617.1 (Kappa), P.2, B.1.427/9 (Epsilon), B.1.525 (Eta), B.1.526 (Iota), C.37 (Lambda), B.1.621 and B.1.620; or the virus is HBV C.


In some embodiments, the method comprises the steps of:

    • a) mixing the sample with each of said peptide pools A, B, and C to produce assay samples;
    • b) incubating each mixture formed for a period to allow T cell activation;
    • c) measuring the level of at least one secreted cytokine in each said mixture and determining whether the level of at least one secreted cytokine is above a threshold control value to indicate a positive T-cell reaction; and
    • d) determining the number or proportion of reactive T cells present in each pool.


In some embodiments, the secreted cytokine is selected from the group comprising IFN-gamma (IFN-γ), IL-2, CXCL9, CXCL10, TNF-alpha, IL-6, IL-10 and IL-1. Preferably IFN-gamma, IL-2, or CXCL10 levels are measured. Detection of the cytokine CXCL10 is preferred if qPCR is used to quantify T cell activation.


In some embodiments, the said cytokine level is determined by immunoassay, such as ELISA or ELISPOT, or by qPCR or direct qPCR.


According to the method, a sample is tested by separately mixing aliquots from the sample with a peptide pool representing at least a portion of M, NP or S protein, or with a peptide pool representing at least a portion of E, Pol, C or X protein to determine whether the sample comprises T cells reactive to M, NP and/or S peptides; or E, Pol, C and/or X peptides, respectively.


In some embodiments, the sample comprises whole blood, broncholavage (BAL fluid), nasal swabs, nasopharyngeal aspirate, or isolated peripheral blood mononuclear cells (PBMCs).


In some embodiments, when whole blood, broncholavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate is used, the incubation period in step b) may be for between about 6 to 24 h.


In some embodiments of the method:

    • a) whole blood is mixed with each of said peptide pools;
    • b)i) each mixture is incubated for at least 6 h;
    • b)ii) a plasma fraction of the mixture is isolated;
    • c) the level of at least one secreted cytokine in each said plasma fraction is measured and compared to a threshold control value to indicate a positive or negative T cell reaction.


In some embodiments, the sample also comprises a concentration of DMSO and/or heparin. Heparin may be required particularly if whole blood is to be assayed, to inhibit coagulation.


In some embodiments, the control sample or threshold control value may be derived from an assay sample comprising a subject sample that is unstimulated or DMSO-treated.


In some embodiments of the method:

    • a) whole blood is mixed with heparin, DMSO and each of said peptide pools to produce:
    • i) 4 assay samples corresponding to M, NP1, NP2 and S; or
    • ii) 8 assay samples corresponding to E1, E2, Pol1, Po12, Po13, Po14, C and X;
    • b)i) each mixture is incubated overnight;
    • b)ii) a plasma fraction of the mixture is isolated;
    • c) the level of at least one secreted cytokine, selected from the group comprising IFN-gamma, IL-2, CXCL9, CXCL10, TNF-alpha, IL-6, IL-10 and IL-1, in each said plasma fraction is measured and compared to a threshold control value, derived from an assay sample comprising a subject sample that is unstimulated or DMSO-treated, to indicate a positive or negative T cell reaction.


In a third aspect the invention provides a method to quantify the presence of virus-specific T cells in a biological sample comprising or derived from blood, broncholavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject, comprising;

    • a) Mixing the biological sample with one or more virus peptide pools, wherein said peptide pools are separately derived from virus antigenic structural or non-structural proteins;
    • b) incubating the mixture formed for a period to allow T cell activation;
    • c) Rupture the cells from b);
    • d) Aliquot a sample from c) into PCR reagents, ACTIN (or other internal control) forward and reverse primers, ACTIN (or other internal control) probe, CXCL10 forward and reverse primers and CXCL10 probe for dqPCR; and/or
    • e) extract RNA from a sample from c) and add a portion into PCR reagents, ACTIN (or other internal control) forward and reverse primers, ACTIN (or other internal control) probe, CXCL10 forward and reverse primers and CXCL10 probe for qPCR;
    • f) perform cycles of dqPCR and/or qPCR for d) and e), respectively; and
    • g) quantitate the expression of CXCL10 in the stimulated T cells and compare with a control,
    • wherein an elevated CXCL10 level indicates the presence of virus-specific T cells in the subject sample.


In some embodiments the virus is an enveloped virus. The antigenic structural and non-structural proteins of these viruses are well-known. An example is the membrane (M), nucleoprotein (NP) and/or Spike (S) proteins of a coronavirus. The antigenic proteins of Hepatitis B virus (HBV) are Pol, E, C and X proteins.


In some embodiments, the virus is a coronavirus, selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, KHU1, OC43, NL63 and 229E or variant thereof; or a non-enveloped virus, such as HBV.


In some embodiments, the virus is SARS-CoV-2 or HBV.


In some embodiments:

    • ai) the M protein comprises the amino acid sequence set forth in SEQ ID NO: 793;
    • ii) the NP protein comprises the amino acid sequence set forth in SEQ ID NO: 794; and
    • iii) the S protein comprises the amino acid sequence set forth in SEQ ID NO: 795; or
    • bi) the Pol protein comprises the amino acid sequence set forth in SEQ ID NO: 796;
    • ii) the E protein comprises the amino acid sequence set forth in SEQ ID NO: 797;
    • iii) the C protein comprises the amino acid sequence set forth in SEQ ID NO: 798; and
    • iv) the X protein comprises the amino acid sequence set forth in SEQ ID NO: 799.


In some embodiments, the peptide pools comprise one or more M, NP and S peptides listed in Tables 1-4 and 7-19; or one or more Pol, E, C and X peptides listed in Tables 20-27.


In a fourth aspect, the invention provides a method of treatment comprising administering, to a subject with T cells reactive to:

    • i) a majority of peptide pools derived from virus antigenic structural or non-structural proteins, an effective amount of a virus inhibitor; or
    • ii) 0, or a minority of the peptide pools M, NP and S; or E, Pol, C and X, an effective amount of a coronavirus or HBV vaccine, respectively.


In some embodiments, the virus is a coronavirus such as a coronavirus selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, HKU1, OC43, NL63 and 229E or variants thereof.


In some embodiments, the virus is SARS-CoV-2.


In some embodiments the virus is HBV.


In some embodiments, the invention provides a method of treatment comprising administering, to a subject with T cells reactive to 3 or 4 of the peptide pools M, NP1, NP2 and S listed in Tables 1-4, an effective amount of a SARS-CoV-2 inhibitor.


In some embodiments, the invention provides a method of treatment comprising administering, to a subject with T cells reactive to 3 or 4 of the peptide pools M, NP1, NP2 and S listed in Tables 1-4, an effective amount of a SARS-CoV-2 inhibitor.


In some embodiments, the invention provides a method of treatment comprising administering, to a subject with T cells reactive to 5 to 8 of the pools E1, E2, Pol1, Pol2, Pol3, Pol4, C and X listed in Tables 20-27, an effective amount of a HBV inhibitor.


In a fifth aspect, the invention provides a method of prophylaxis comprising administering, to a subject with T cells reactive to 0, or a minority of peptide pools derived from virus antigenic structural and non-structural proteins, an effective amount of a virus vaccine.


In some embodiments the virus is an enveloped virus.


In some embodiments, the virus is a coronavirus such as a coronavirus selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, HKU1, OC43, NL63 and 229E or variants thereof.


In some embodiments, the virus is SARS-CoV-2. In some embodiments the virus is HBV.


In some embodiments, the invention provides a method of prophylaxis comprising administering, to a subject with T cells reactive to 0, 1 or 2 of the peptide pools M, NP1, NP2 and S listed in Tables 1-4, an effective amount of a SARS-CoV-2 vaccine.


In some embodiments, the invention provides a method of prophylaxis comprising administering, to a subject with T cells reactive to 0, 1, 2, 3 or 4 of the peptide pools E1, E2, Pol1, Po12, Po13, Po14, C and X listed in Tables 20-27, an effective amount of a HBV vaccine.


In a sixth aspect the invention provides a method of monitoring the efficacy of a virus vaccine, comprising testing whether the recipient of said vaccine has T cells reactive to a minority, 50%, or majority of peptide pools derived from virus antigenic structural and non-structural proteins, such as virus M, NP and S proteins; or virus E, Pol, C and X proteins.


In some embodiments, the virus is a coronavirus such as a coronavirus selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, HKU1, OC43, NL63 and 229E or variants thereof. In some embodiments, the virus is SARS-CoV-2.


In some embodiments the virus is HBV.


In some embodiments, the invention provides a method of monitoring the efficacy of a SARS-CoV-2 vaccine, comprising testing whether the recipient of said vaccine has T cells reactive to 0, 1, 2, 3 or 4 of the peptide pools M, NP1, NP2 and S listed in Tables 1-4 and 7-19.


In some embodiments, the invention provides a method of monitoring the efficacy of a HBV vaccine, comprising testing whether the recipient of said vaccine has T cells reactive to 0, 1, 2, 3, 4, 5, 6, 7 or 8 of the peptide pools E1, E2, Pol1, Pol2, Pol3, Pol4, C and X listed in Tables 20-27.


It would be understood that the pools used may depend on which of the virus antigenic proteins is/are used in the vaccine. The pool may be for a particular protein from a wildtype, or variant virus. Table 4 contains selected peptides of the spike protein that were tested and demonstrated to be good enough to estimate the total spike T cell response. This table of peptides do not cover the entire spike protein. This is used in conjunction with peptides from Tables 1-3 to detect if the subject is infected or not infected. Table 7 is the reference peptides that may be used to assess the T cell response against the delta VOC with the wildtype Wuhan as a reference. Tables 8-19 contain peptides derived from the VOC that are different from the Wuhan wildtype SARS-CoV-2 virus.


In a seventh aspect the invention provides a kit to discriminate past or currently virus-infected subjects from virus un-infected subjects, the kit comprising a plurality of virus structural or non-structural peptides that stimulate virus-exposed T cells, wherein the virus peptides are in peptide pools derived from virus antigenic structural or non-structural proteins.


In some embodiments the virus is an enveloped virus or a non-enveloped virus.


An example of the antigenic proteins is the membrane (M), nucleoprotein (NP) and/or Spike (S) proteins of an enveloped virus.


In some embodiments, the virus is a coronavirus such as a coronavirus selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, HKU1, OC43, NL63 and 229E or variants thereof.


In some embodiments, the virus is SARS-CoV-2. In some embodiments the virus is HBV.


In some embodiments of the kit:

    • ai) the M protein comprises the amino acid sequence set forth in SEQ ID NO: 793;
    • ii) the NP protein comprises the amino acid sequence set forth in SEQ ID NO: 794; and
    • iii) the S protein comprises the amino acid sequence set forth in SEQ ID NO: 795; or
    • bi) the Pol protein comprises the amino acid sequence set forth in SEQ ID NO: 796;
    • ii) the E protein comprises the amino acid sequence set forth in SEQ ID NO: 797;
    • iii) the C protein comprises the amino acid sequence set forth in SEQ ID NO: 798; and
    • iv) the X protein comprises the amino acid sequence set forth in SEQ ID NO: 799.


In some embodiments:

    • the M peptide pool comprises peptides having amino acid sequences set forth in SEQ ID Nos: 1-43;
    • the NP peptide pool comprises peptides having amino acid sequences set forth in SEQ ID Nos: 44-125;
    • the S peptide pool comprising peptides selected from peptides having amino acid sequences set forth in SEQ ID Nos: 126-454.


In some embodiments the kit further comprises one or more reagents to detect cytokines and/or chemokines secreted from activated T cells.


In some embodiments the kit further comprises:

    • i) PCR reagents and/or primers and probes to detect CXCL10 and/or IFN-gamma expression; and/or
    • ii) ELISPOT reagents.


In some embodiments, suitable primers and probes are as shown in Table 5.


In some embodiments, the kit comprises one or more peptide pools selected from the pools in Tables 7-19 rather than the pools in Tables 1-4. Such pools could be used to analyse the effect of virus variants, including variants of concern (VOC), on T cell activation in vaccinated or previously infected subjects.


In some embodiments the VOC are selected from the group comprising B.1.617.2 (Delta), B.1.1.7 (Alpha V1), B.1.351 (Beta V2), P.1 (Gamma, V3), B.1.617.1 (Kappa), P.2, B.1.427/9 (Epsilon), B.1.525 (Eta), B.1.526 (Iota), C.37 (Lambda), B.1.621 and B.1.620.


Herein is presented a method based on specific combination of peptide pools covering both different SARS-CoV-2 proteins (i.e., Spike) and the corresponding regions affected by the amino acid mutations that are used to stimulate T cells, The results of this multiple stimulation strategy define with accuracy the impact of these amino acid mutations on the SARS-CoV-2-specific T cells induced by infection with Wuhan strain or by vaccination with the present vaccines based on the Wuhan strain. Peptide pools directed to non-conserved regions of the Wuhan strain variants are shown in Tables 8 to 19, while the sequences of the regions of the Wuhan strain that correspond to the regions of the Delta variant (B.1.617.2) are shown in Table 7.


Presented herein is an example of a method to analyze the impact of AA mutations present in the Spike protein in different VOCs (using Delta variant as an example) on the SPIKE specific T cells induced by vaccination. It would be understood that pools derived from non-conserved regions of M or NP may be used to analyse the effect of VOCs, depending on the antigens the subject's T cells have been exposed to.


In an eighth aspect the invention provides a set of at least 2, at least 3, or at least 4 separate pools of peptides suitable to discriminate:

    • i) past or currently SARS-CoV-2-infected subjects from SARS-CoV-2 un-infected subjects, wherein the peptide pools are selected from those listed in Tables 1 to 4 and 7-19; or
    • ii) past or currently HBV-infected subjects from HBV un-infected subjects, wherein the peptide pools are selected from those listed in Tables 20-27.


In a ninth aspect the invention provides a use of a kit of aspect 7 in a method according to any one of aspects 1 to 6.


Having now generally described the invention, the same will be more readily understood through reference to the following examples which are provided by way of illustration, and are not intended to be limiting of the present invention.


EXAMPLES

Standard molecular biology techniques known in the art and not specifically described were generally followed as described in Green and Sambrook, Molecular Cloning: A Laboratory Manual, Cold Springs Harbor Laboratory, New York (2012).


Example 1
SARS-CoV-2 Peptide Pools

An integral part of our invention is the selection of peptide pools necessary to define a profile of T cell responses that can differentiate individuals that have been primed by SARS-CoV-2 infection or individuals that were infected by other common cold coronaviruses. The SARS-CoV-2 specific T cell responses were profiled in individuals who were in contact with SARS-CoV-2 and tested antibody positive (Abbot test/anti-NP antibody) or were positive in a surrogate virus neutralization assay (sVNT) at the time of the T cell test (symptomatic n=35; asymptomatic n=73), and compared the profile to that obtained from healthy donors (n=51) without any history of SARS-CoV-2 contact and negative for anti-NP SARS-CoV-2 antibodies.


Four SARS-CoV-2 peptide pools of 15-mers (Tables 1-4) covering NP (NP-1, NP-2), membrane (M), and one pool of 55 peptides covering the most immunogenic regions of Spike (S)(FIG. 1) were generated. Spike is a long protein with 1276 amino acids, so it requires 253 15-mer peptides overlapping by 10 amino acids to cover the whole protein, thus 7 pools of about 40 peptides. To reduce the number of peptides pools to test, we selected a single “Spike pool” comprised of 55 peptides. For the selection, all sequences of published SARS-CoV-1 epitopes (wwwdotiedbdotorg; positive assays only, T cells assays, host: human) were aligned with the library of Spike-SARS-CoV-2 15-mers. The 15-mer peptides cover the homologue sequence of the described SARS-CoV-1 epitope sequences. In addition, we added the 15-mer peptides that cover the predicted SARS-CoV-2 Spike epitopes published by Grifoni et al., [Cell Host Microbe. 27(4): 671-680 (2020)]. The 55 peptides cover 40.5% of the Spike protein. The frequency of reactive cells to the selected Spike pool (red) was compared to the 7 pools of 15-mers overlapping by 10 amino acids covering together the entire Spike protein (S1-S7) in 15 COVID-19 convalescents. It would be understood that the invention is not limited to use of pools having specific peptide sequences disclosed herein, and that pools comprising peptides corresponding to a shift of one or only a few amino acids along the virus protein sequence may generate useful diagnostic data, given there are overlaps in the peptides.









TABLE 1







Summary of Peptide Pool M.










POOL M





Peptide


SEQ ID


Number
A.A Sequence
A.A Position
Number













M_1
MADSNGTITVEELKK
 1-15
1





M_2
GTITVEELKKLLEQW
 6-20
2





M_3
EELKKLLEQWNLVIG
11-25
3





M_4
LLEQWNLVIGFLFLT
16-30
4





M_5
NLVIGFLFLTWICLL
21-35
5





M_6
FLFLTWICLLQFAYA
26-40
6





M_7
WICLLQFAYANRNRF
31-45
7





M_8
QFAYANRNRFLYIIK
36-50
8





M_9
NRNRFLYIIKLIFLW
41-55
9





M_10
LYIIKLIFLWLLWPV
46-60
10





M_11
LIFLWLLWPVTLACF
51-65
11





M_12
LLWPVTLACFVLAAV
56-70
12





M_13
TLACFVLAAVYRINW
61-75
13





M_14
VLAAVYRINWITGGI
66-80
14





M_15
YRINWITGGIAIAMA
71-85
15





M_16
ITGGIAIAMACLVGL
76-90
16





M_17
AIAMACLVGLMWLSY
81-95
17





M_18
CLVGLMWLSYFIASF
 86-100
18





M_19
MWLSYFIASFRLFAR
 91-105
19





M_20
FIASFRLFARTRSMW
 96-110
20





M_21
RLFARTRSMWSFNPE
101-115
21





M_22
TRSMWSFNPETNILL
106-120
22





M_23
SFNPETNILLNVPLH
111-125
23





M_24
TNILLNVPLHGTILT
116-130
24





M_25
NVPLHGTILTRPLLE
121-135
25





M_26
GTILTRPLLESELVI
126-140
26





M_27
RPLLESELVIGAVIL
131-145
27





M_28
SELVIGAVILRGHLR
136-150
28





M_29
GAVILRGHLRIAGHH
141-155
29





M_30
RGHLRIAGHHLGRCD
146-160
30





M_31
IAGHHLGRCDIKDLP
151-165
31





M_32
LGRCDIKDLPKEITV
156-170
32





M_33
IKDLPKEITVATSRT
161-175
33





M_34
KEITVATSRTLSYYK
166-180
34





M_35
ATSRTLSYYKLGASQ
171-185
35





M_36
LSYYKLGASQRVAGD
176-190
36





M_37
LGASQRVAGDSGFAA
181-195
37





M_38
RVAGDSGFAAYSRYR
186-200
38





M_39
SGFAAYSRYRIGNYK
191-205
39





M_40
YSRYRIGNYKLNTDH
196-210
40





M_41
IGNYKLNTDHSSSSD
201-215
41





M_42
LNTDHSSSSDNIALL
206-220
42





M_43
SSSSDNIALLVQ
211-222
43
















TABLE 2







Summary of Peptide Pool NP-1.










POOL 





NP-1





Peptide

A.A
SEQ ID


Number
A.A Sequence
Position
Number





NP_1
MSDNGPQNQRNAPRI
 1-15
44





NP_2
PQNQRNAPRITFGGP
 6-20
45





NP_3
NAPRITFGGPSDSTG
11-25
46





NP_4
TFGGPSDSTGSNQNG
16-30
47





NP_5
SDSTGSNQNGERSGA
21-35
48





NP_6
SNQNGERSGARSKQR
26-40
49





NP_7
ERSGARSKQRRPQGL
31-45
50





NP_8
RSKQRRPQGLPNNTA
36-50
51





NP_9
RPQGLPNNTASWFTA
41-55
52





NP_10
PNNTASWFTALTQHG
46-60
53





NP_11
SWFTALTQHGKEDLK
51-65
54





NP_12
LTQHGKEDLKFPRGQ
56-70
55





NP_13
KEDLKFPRGQGVPIN
61-75
56





NP_14
FPRGQGVPINTNSSP
66-80
57





NP_15
GVPINTNSSPDDQIG
71-85
58





NP_16
TNSSPDDQIGYYRRA
76-90
59





NP_17
DDQIGYYRRATRRIR
81-95
60





NP_18
YYRRATRRIRGGDGK
 86-100
61





NP_19
TRRIRGGDGKMKDLS
 91-105
62





NP_20
GGDGKMKDLSPRWYF
 96-110
63





NP_21
MKDLSPRWYFYYLGT
101-115
64





NP_22
PRWYFYYLGTGPEAG
106-120
65





NP_23
YYLGTGPEAGLPYGA
111-125
66





NP_24
GPEAGLPYGANKDGI
116-130
67





NP_25
LPYGANKDGIIWVAT
121-135
68





NP_26
NKDGIIWVATEGALN
126-140
69





NP_27
IWVATEGALNTPKDH
131-145
70





NP_28
EGALNTPKDHIGTRN
136-150
71





NP_29
TPKDHIGTRNPANNA
141-155
72





NP_30
IGTRNPANNAAIVLQ
146-160
73





NP_31
PANNAAIVLQLPQGT
151-165
74





NP_32
AIVLQLPQGTTLPKG
156-170
75





NP_33
LPQGTTLPKGFYAEG
161-175
76





NP_34
TLPKGFYAEGSRGGS
166-180
77





NP_35
FYAEGSRGGSQASSR
171-185
78





NP_36
SRGGSQASSRSSSRS
176-190
79





NP_37
QASSRSSSRSRNSSR
181-195
80





NP_38
SSSRSRNSSRNSTPG
186-200
81





NP_39
RNSSRNSTPGSSRGT
191-205
82





NP_40
NSTPGSSRGTSPARM
196-210
83





NP_41
SSRGTSPARMAGNGG
201-215
84
















TABLE 3







Summary of Peptide Pool NP-2.










POOL 





NP-2





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





NP_42
SPARMAGNGGDAALA
206-220
 85





NP_43
AGNGGDAALALLLLD
211-225
 86





NP_44
DAALALLLLDRLNQL
216-230
 87





NP_45
LLLLDRLNQLESKMS
221-235
 88





NP_46
RLNQLESKMSGKGQQ
226-240
 89





NP_47
ESKMSGKGQQQQGQT
231-245
 90





NP_48
GKGQQQQGQTVTKKS
236-250
 91





NP_49
QQGQTVTKKSAAEAS
241-255
 92





NP_50
VTKKSAAEASKKPRQ
246-260
 93





NP_51
AAEASKKPRQKRTAT
251-265
 94





NP_52
KKPRQKRTATKAYNV
256-270
 95





NP_53
KRTATKAYNVTQAFG
261-275
 96





NP_54
KAYNVTQAFGRRGPE
266-280
 97





NP_55
TQAFGRRGPEQTQGN
271-285
 98





NP_56
RRGPEQTQGNFGDQE
276-290
 99





NP_57
QTQGNFGDQELIRQG
281-295
100





NP_58
FGDQELIRQGTDYKH
286-300
101





NP_59
LIRQGTDYKHWPQIA
291-305
102





NP_60
TDYKHWPQIAQFAPS
296-310
103





NP_61
WPQIAQFAPSASAFF
301-315
104





NP_62
QFAPSASAFFGMSRI
306-320
105





NP_63
ASAFFGMSRIGMEVT
311-325
106





NP_64
GMSRIGMEVTPSGTW
316-330
107





NP_65
GMEVTPSGTWLTYTG
321-335
108





NP_66
PSGTWLTYTGAIKLD
326-340
109





NP_67
LTYTGAIKLDDKDPN
331-345
110





NP_68
AIKLDDKDPNFKDQV
336-350
111





NP_69
DKDPNFKDQVILLNK
341-355
112





NP_70
FKDQVILLNKHIDAY
346-360
113





NP_71
ILLNKHIDAYKTFPP
351-365
114





NP_72
HIDAYKTFPPTEPKK
356-370
115





NP_73
KTFPPTEPKKDKKKK
361-375
116





NP_74
TEPKKDKKKKADETQ
366-380
117





NP_75
DKKKKADETQALPQR
371-385
118





NP_76
ADETQALPQRQKKQQ
376-390
119





NP_77
ALPQRQKKQQTVTLL
381-395
120





NP_78
QKKQQTVTLLPAADL
386-400
121





NP_79
TVTLLPAADLDDFSK
391-405
122





NP_80
PAADLDDFSKQLQQS
396-410
123





NP_81
DDFSKQLQQSMSSAD
401-415
124





NP_82
QLQQSMSSADSTQA
406-419
125
















TABLE 4







Summary of Peptide Pool SP.












POOL 






SP






Peptide 

A.A
SEQ ID



Number
A.A Sequence
Position
Number







SP_21
IRGWIFGTTLDSKTQ
101-115
126







SP_22
FGTTLDSKTQSLLIV
106-120
127







SP_34
CTFEYVSQPFLMDLE
166-180
128







SP_35
VSQPFLMDLEGKQGN
171-185
129







SP_48
TRFQTLLALHRSYLT
236-250
130







SP_49
LLALHRSYLTPGDSS
241-255
131







SP_50
RSYLTPGDSSSGWTA
246-260
132







SP_59
CALDPLSETKCTLKS
291-305
133







SP_60
LSETKCTLKSFTVEK
296-310
134







SP_61
CTLKSFTVEKGIYQT
301-315
135







SP_62
FTVEKGIYQTSNFRV
306-320
136







SP_63
GIYQTSNFRVQPTES
311-325
137







SP_70
RFASVYAWNRKRISN
346-360
181







SP_71
YAWNRKRISNCVADY
351-365
182







SP_75
SASFSTFKCYGVSPT
371-385
138







SP_76
TFKCYGVSPTKLNDL
376-390
139







SP_85
YNYKLPDDFTGCVIA
421-435
140







SP_88
WNSNNLDSKVGGNYN
436-450
141







SP_89
LDSKVGGNYNYLYRL
441-455
142







SP_90
GGNYNYLYRLFRKSN
446-460
143







SP_91
YLYRLFRKSNLKPFE
451-465
144







SP_92
FRKSNLKPFERDIST
456-470
145







SP_93
LKPFERDISTEIYQA
461-475
146







SP_106
GPKKSTNLVKNKCVN
526-540
147







SP_107
TNLVKNKCVNFNFNG
531-545
148







SP_109
FNFNGLTGTGVLTES
541-555
149







SP_110
LTGTGVLTESNKKFL
546-560
150







SP_130
RAGCLIGAEHVNNSY
646-660
151







SP_131
IGAEHVNNSYECDIP
651-665
152







SP_138
SVASQSIIAYTMSLG
686-700
153







SP_139
SIIAYTMSLGAENSV
691-705
154







SP_140
TMSLGAENSVAYSNN
696-710
155







SP_150
STECSNLLLQYGSFC
746-760
156







SP_151
NLLLQYGSFCTQLNR
751-765
157







SP_156
KNTQEVFAQVKQIYK
776-790
158







SP_157
VFAQVKQIYKTPPIK
781-795
159







SP_158
KQIYKTPPIKDFGGF
786-800
160







SP_159
TPPIKDFGGFNFSQI
791-805
161







SP_161
NFSQILPDPSKPSKR
801-815
162







SP_167
AGFIKQYGDCLGDIA
831-845
163







SP_168
QYGDCLGDIAARDLI
836-850
164







SP_179
GAALQIPFAMQMAYR
891-905
165







SP_181
QMAYRFNGIGVTQNV
901-915
166







SP_182
FNGIGVTQNVLYENQ
906-920
167







SP_188
DSLSSTASALGKLQD
936-950
168







SP_189
TASALGKLQDVVNQN
941-955
169







SP_192
AQALNTLVKQLSSNF
956-970
170







SP_196
VLNDILSRLDKVEAE
976-990
171







SP_200
LITGRLQSLQTYVTQ
 996-1010
172







SP_203
QLIRAAEIRASANLA
1011-1025
173







SP_204
AEIRASANLAATKMS
1016-1030
174







SP_212
APHGVVFLHVTYVPA
1056-1070
175







SP_221
HWFVTQRNFYEPQII
1101-1115
176







SP_237
KEIDRLNEVAKNLNE
1181-1195
177







SP_238
LNEVAKNLNESLIDL
1186-1200
178







SP_239
KNLNESLIDLQELGK
1191-1205
179







SP_244
IWLGFIAGLIAIVMV
1216-1230
180










Example 2
Sample Processing and Testing
Human Samples

Whole blood samples (8 ml) were collected from individuals who were in contact with SARS-CoV-2 and tested antibody positive (Abbot test/anti-NP antibody), or were positive in a surrogate virus neutralization assay (sVNT) at the time of the T cell test (symptomatic n=35; asymptomatic n=73), or were healthy donors (n=51) without any history of SARS-CoV-2 contact and negative for anti-NP SARS-CoV-2 antibodies.


Schematic representations of suitable assays of the invention are presented in FIG. 2.


ELISpot Testing of PBMCs

SARS-CoV-2-specific T cells were tested as described previously [Le Bert N, et al., Nature 584(7821): 457-62 (2020)]. Briefly, peripheral blood mononuclear cells (PBMCs) were isolated from 8 ml whole blood by density-gradient centrifugation using Ficoll-Paque™ (Sigma-Aldrich). Isolated PBMC were either studied directly or cryopreserved and stored in liquid nitrogen until used in the assays.


ELISpot plates (Millipore) were coated with human IFNγ antibody (1-D1K, Mabtech; 5 μg/ml) overnight at 4° C. Then, 4×105 PBMCs were seeded per well and stimulated for 18 h with the different pools of SARS-CoV-2 peptides (2 μg/ml final concentration per peptide) described in Tables 1-4. For stimulation with peptide matrix pools or single peptides, a concentration of 5 μg/ml was used. Subsequently, the plates were developed with human biotinylated IFNγ detection antibody (7-B6-1, Mabtech; 1:2,000), followed by incubation with streptavidin-AP (Mabtech) and KPL BCIP/NBT Phosphatase Substrate (SeraCare). Spot forming units (SFU) were quantified with ImmunoSpot. To quantify positive peptide-specific responses, the highest number of spots of the unstimulated (or DMSO stimulated) wells was subtracted from the peptide-stimulated wells, and the results expressed as SFU/106 PBMCs. We excluded the results if negative control wells had >30 SFU/106 PBMCs or positive control wells (phorbol 12-myristate 13-acetate/ionomycin) were negative.


The ELISPOT assays showed that patients who have been infected by SARS-CoV-2 and cleared the virus up to 3 months ago have T cells that are reactive to peptide pools covering Membrane, Nucleoprotein and Spike (FIG. 3A). In contrast, individuals who are antibody anti-NP negative and without a history of SARS-CoV-2 infection (healthy donors) present only occasional responses to 1-2 peptide pools (FIG. 3B).


Whole Blood Testing

Whole blood was isolated from each subject and tested within 24 hours after blood draw. 400 μl aliquots were separately mixed with 100 μl RPMI containing each of the SARS-CoV-2 peptide pools (2 μg/ml final concentration per peptide) or a DMSO control and incubated for a period extending overnight, to allow activation of responsive T cells. A plasma fraction was isolated from each of the incubated samples (about 30 minutes) and the level of cytokines in the sample measured using an EIIa™ multi-analyte ELISA machine (ProteinSimple, CA, USA).


It would be understood that the parameters described in the example may be changed and still achieve a valid assay. For example, the ratio of blood to RMPI media can range from 100% blood to 50% blood; 100 μl aliquots or more than 400 μl might be used, but a larger blood sample would be required to test multiple peptide pools. RPMI may be exchanged with other cell culture media. The final concentration of peptides in an assay mixture may be from 1 to 5 μg/ml. The control sample may contain DMSO or may be an unstimulated blood sample. The incubation period may range between about 6-24 h.


Having showed that the distinct peptide pools are able to define individuals that were recently in contact with SARS-CoV-2 (FIG. 3), we then demonstrated that the SARS-CoV-2 T cell response profile can also be delineated through the direct activation of antigen-specific T cells in whole blood using the same peptide pools and measuring the secreted cytokines in the plasma. We activated whole blood obtained from uninfected individuals (n=9) and individuals who have been infected with SARS-CoV-2 (n=6), as confirmed by the detection of anti-NP antibodies or by SARS-CoV-2 sVNT assays, using the respective peptide pools and measured the concentration of IFN-γ in the plasma collected after 24 hours.


SARS-CoV-2 reactive T cells in infected individuals can be detected by quantifying the amount of secreted IFN-γ after the direct addition of the peptide pools into whole blood (FIG. 4A). Similar to the results obtained with the ELISPOT assay, uninfected individuals occasionally have 1-2 responding peptide pools while infected individuals are simultaneously reactive to all peptide pools tested (FIG. 4B).


qPCR and dqPCR Testing of PBMCs


The complexity of quantifying the presence of virus-specific T cell has so far prevented large scale studies of the cellular immune response to viral infection or, more recently, the vaccine. To address this problem, we have implemented a qPCR-based rapid T cell Activation (qTACT) assay, based on in vitro stimulation of whole blood samples with a pool of viral peptides covering the spike or other SARS-CoV-2 viral proteins (i.e., nucleoprotein [N]) [Le Bert, N., et al. Nature 584: 457-462 (2020); Kalimuddin S, et al., Med (N Y). 2021 Jun. 11; 2(6):682-688.e4. doi: 10.1016/j.medj.2021.04.003; Le Bert, N. et al., J Exp Med 218(5):e20202617 (2021)], followed by direct amplification of IFN-γ or IL-2 (directly produced by SARS-CoV-2 antigen-specific T cells) or CXCL10, a molecule expressed by monocytes in response to T cell activation. A further technical implementation of the assay allows quantification of T cell immunity directly from blood, bypassing the need for red blood cell (RBC) lysis or RNA purification. We call this latter assay direct qPCR-based rapid T cell Activation (dqTACT) assay


Summary of Methods





    • 1. Collect blood (sodium heparin collection tube) & stimulate (320 μl blood+80 μl RPMI/peptide) overnight.

    • 2. The next morning, collect 25 μl of serum in Eppendorf tubes for ELLA/ELISA (freeze −80° C.).

    • 3. Replace serum with 25 μl of RPMI.

    • 4. Vortex or pipette up and down aggressively.

    • 5. Split remaining sample into two more tubes:

    • a. Tube A (whole blood—dqTACT): 180 μl blood+540 μl buffer A (process immediately or freeze −80° C.).

    • b. Tube B (RNA—qTACT): 180 μl blood+180 μl RNA/DNA shield+3.6 μl blood proteinase K (incubate at room temperature for 30 minutes then process immediately or freeze −80° C.).

    • i. To extract, add Trizol reagent (1:1) then proceed with Direct-zol 96 extraction kit (Zymo).





Primers and probes should be resuspended at 100 μM and stored at −20° C. Keep probes away from light when working with them.









TABLE 5







Primer & probe sequences 


(for qTACT or dqTACT)













SEQ



Primer/

ID



probe
Primer/probe sequence
No







ACTIN 
/5HEX/TCATCCATG/ZEN/
455



probe 
GTGAGCTGGCGG/3IABkFQ/




HEX









CXCL10 
/56-FAM/AGTGGCATT/ZEN/
456



probe
CAAGGAGTACCTCTCTCT/




FAM
3IABkFQ/








ACTIN 
CCTTGCACATGCCGGAG
457



primer 1









ACTIN 
ACAGAGCCTCGCCTTTG
458



primer 2









CXCL10 
CCATTCTGATTTGCTGCCTTATC
459



primer 1









CXCL10 
TACTAATGCTGATGCAGGTACAG
460



primer 2











dqTACT PCR 1× Mix:


10 μl SCRIPT Direct RT-qPCR ProbesMaster mix; 0.1 μl ACTIN primer 1; 0.1 μl ACTIN primer 2; 0.1 μl CXCL10 primer 1; 0.1 μl CXCL10 primer 2; 0.05 μl ACTIN probe (HEX); 0.05 μl CXCL10 probe (FAM); 2 μl blood/buffer A mixture; 7.5 μl PEC-1


qTACT PCR 1× Mix:


5 μl TaqPath 1 Step Multiplex Master Mix (no ROX); 0.1 μl ACTIN primer 1; 0.1 μl ACTIN primer 2; 0.1 μl CXCL10 primer 1; 0.1 μl CXCL10 primer 2; 0.05 μl ACTIN probe (HEX); 0.05 μl CXCL10 probe (FAM); 5 μl RNA; 9.5 μl PEC-1


dqTACT Run Conditions (Hyris bCUBE ONLY):

    • Reverse transcription: 53° C. for 15 minutes
    • Initial denaturation: 95° C. for 5 minutes
    • PCR (X45 cycles):
    • 95° C. 15 seconds
    • 60° C. 30 seconds


      qTACT Run Conditions (Hyris bCUBE or CFX96/384)
    • UNG incubation: 25° C. for 2 minutes
    • Reverse transcription: 53° C. for 10 minutes
    • Initial denaturation: 95° C. for 2 minutes
    • PCR (X45 cycles):
    • 95° C. for 15 seconds
    • 60° C. for 30 seconds


Protocol Descriptions

Whole Blood Culture with SARS-CoV-2 Peptide Pools


320 μl of whole blood drawn on the same day into sodium heparin tubes (BD) were mixed with 80 μl RPMI and stimulated with pools of SARS-CoV-2 peptides (S or NP; 2 μg/ml) or DMSO control at 37° C. After 15-17 hours of stimulation, the supernatant (plasma) was collected and stored at −80° C. until quantification of cytokines.


qTACT Assay


Samples used for RNA extraction were diluted 1:1 with RNA/DNA shield (Zymo) and incubated at room temperature with proteinase K at a 1:100 dilution (20 mg/ml stock). Samples were then frozen at −80° C. until RNA extraction could be performed. Samples stored in RNA/DNA shield were thawed at room temperature prior to RNA extraction. Samples were vortexed and mixed with Trizol reagent (Life Technologies) at a 1:1 dilution. After vortexing, samples were processed using the Direct-zol 96 well extraction kit (Zymo) as per the manufacturer's instructions. Eluted RNA was diluted in TE buffer, aliquoted, and stored at −80° C. or used immediately for qPCR analysis. Real-time quantification was performed on a BioRad CFX96/CFX384 or Hyris bCUBE 2.0. 5 μl of diluted RNA was used with the TaqPath 1-Step Multiplex MasterMix (Applied Biosystems) and primers/probes targeting ACTIN (internal control) and other target genes, as described.


dqTACT Assay


Samples used for direct amplification from whole blood were diluted 1:3 with Buffer A and stored at −80° C. or used immediately for qPCR analysis. 2 μl of diluted whole blood was mixed with SCRIPT Direct RT-qPCR ProbesMaster (Jena Bioscience) and primers/probes targeting ACTIN (internal control) and other target genes, as described. Quantification was performed using the Hyris bCUBE 2.0.


Reagents:

    • Buffer A: 2% Tween-20 in RNAse free water
    • SCRIPT Direct RT-qPCR ProbesMaster mix: worldwidewebdotjenabiosciencedotcom/molecular-biology/reversetranscription-rt-pcr/direct-rt-qpcr-robust-amplification/pcr-528-script-direct-rt-qpcr-probesmaster
    • PEC-1: worldwidewebdotklentaqdotcom/products/pcr-enhancer-cocktail-1
    • TaqPath 1 Step Multiplex Master Mix: worldwidewebdotthermofisherdotcom/order/catalog/product/A28526#/A28526


Algorithm Representing T Cell Response Profile

The present study demonstrates that distinct SARS-CoV-2 peptide pools are able to define the individuals that were recently infected with SARS-CoV-2. This T cell response profile can be evaluated using other laboratory techniques capable of detecting T cell activation after peptide stimulation, including the direct activation of antigen-specific T cells in whole blood. A proposed algorithm to interpret the SARS-CoV-2 T cell response profile is summarized in Table 6.


Table 6 summarizes the interpretation of the SARS-CoV-2 T cell response profile in Example 1. When 50% or more of the pools (thus 2, 3 or 4 out of 4) are positive, the subject is categorized as having SARS-COV2 specific T cells induced by SARS-COV-2 infection (thus previously or currently SARS-COV-2 infected).









TABLE 6







Proposed algorithm to interpret SARS-


CoV-2 T cell response profile












No. of positive pools
Spike
M
NP1
NP2
Interpretation





4
+
+
+
+
Infected


3

+
+
+
Infected



+

+
+



+
+

+



+
+
+



2
+
+


Infected



+

+




+


+




+
+





+

+





+
+


1
+



Uninfected




+







+







+


0




Uninfected









Example 3
Impact of Variants of Concern on T Cells Induced by SARS-CoV-2 Wuhan Infection or SARS-CoV-2 Vaccines

Different variants of concern (VOC) of SARS-CoV-2 have replaced world-wide the original SARS-CoV-2 Wuhan isolate. These VOCs are characterized by amino acid substitutions that provide biological advantages like increased infectivity or escape humoral (antibodies) but also cellular (T cells) immunity. We have designed a method based on specific combination of peptide pools covering both different SARS-CoV-2 proteins (i.e., Spike) and the corresponding regions affected by the amino acid mutations that are used to stimulate T cells, The results of this multiple stimulation strategy define with accuracy the impact of these amino acid mutations on the SARS-CoV-2-specific T cells induced by infection with Wuhan strain or by vaccination with the present vaccines based on the Wuhan strain. Peptide pools directed to non-conserved regions of the Wuhan strain variants are shown in Tables 8 to 19, while the sequences of the regions of the Wuhan strain that correspond to the regions of the Delta variant (B.1.617.2) are shown in Table 7.


The inventors present here as an example the method to analyze the impact of AA mutations present in Spike in different VOCs (using Delta variant as an example) on the SPIKE specific T cells induced by vaccination.









TABLE 7







Peptide sequences of non-conserved regions 


in Wuhan strain corresponding to Delta


mutation peptides shown in Table 8.










POOL 





SP





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





SP_002
VLLPLVSSQCVNLTT
 6-20
461





SP_003
VSSQCVNLTTRTQLP
11-25
462





SP_004
VNLTTRTQLPPAYTN
16-30
463





SP_027
CEFQFCNDPFLGVYY
131-145
464





SP_028
CNDPFLGVYYHKNNK
136-150
465





SP_029
LGVYYHKNNKSWMES
141-155
466





SP_030
HKNNKSWMESEFRVY
146-160
467





SP_031
SWMESEFRVYSSANN
151-165
468





SP_032
EFRVYSSANNCTFEY
156-170
469





SP_089
LDSKVGGNYNYLYRL
441-455
142





SP_090
GGNYNYLYRLFRKSN
446-460
143





SP_091
YLYRLFRKSNLKPFE
451-465
144





SP_094
RDISTEIYQAGSTPC
466-480
470





SP_095
EIYQAGSTPCNGVEG
471-485
471





SP_096
GSTPCNGVEGFNCYF
476-490
472





SP_121
GTNTSNQVAVLYQDV
601-615
473





SP_122
NQVAVLYQDVNCTEV
606-620
474





SP_123
LYQDVNCTEVPVAIH
611-625
475





SP_135
CASYQTQTNSPRRAR
671-685
476





SP_136
TQTNSPRRARSVASQ
676-690
477





SP_137
PRRARSVASQSIIAY
681-695
478





SP_188
DSLSSTASALGKLQD
936-950
168





SP_189
TASALGKLQDVVNQN
941-955
169





SP_190
GKLQDVVNQNAQALN
946-960
479
















TABLE 8







Peptide sequences of non-conserved regions 


in SEQUENCE_21A (Delta) (B.1.617.2)










POOL 





SP





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





SP_002
VLLPLVSSQCVNLRT
 6-20
183





SP_003
VSSQCVNLRTRTQLP
11-25
184





SP_004
VNLRTRTQLPPAYTN
16-30
185





SP_027
CEFQFCNDPFLDVYY
131-145
186





SP_028
CNDPFLDVYYHKNNK
136-150
187





SP_029
LDVYYHKNNKSWMES
141-155
188





SP_030
HKNNKSWMESGVYSS
146-160
189





SP_031
SWMESGVYSSANNCT
151-165
190





SP_032
GVYSSANNCTFEYVS
156-170
191





SP_089
LDSKVGGNYNYRYRL
441-455
192





SP_090
GGNYNYRYRLFRKSN
446-460
193





SP_091
YRYRLFRKSNLKPFE
451-465
194





SP_094
RDISTEIYQAGSKPC
466-480
195





SP_095
EIYQAGSKPCNGVEG
471-485
196





SP_096
GSKPCNGVEGFNCYF
476-490
197





SP_121
GTNTSNQVAVLYQGV
601-615
198





SP_122
NQVAVLYQGVNCTEV
606-620
199





SP_123
LYQGVNCTEVPVAIH
611-625
200





SP_135
CASYQTQTNSRRRAR
671-685
201





SP_136
TQTNSRRRARSVASQ
676-690
202





SP_137
RRRARSVASQSIIAY
681-695
203





SP_188
DSLSSTASALGKLQN
936-950
204





SP_189
TASALGKLQNVVNQN
941-955
205





SP_190
GKLQNVVNQNAQALN
946-960
206
















TABLE 9







Peptide sequences of non-conserved regions 


in SEQUENCE_20I (Alpha, V1) (B.1.1.7)












POOL 






SP






Peptide 

A.A
SEQ ID



Number
A.A Sequence
Position
Number







SP_012
LPFFSNVTWFHAISG
56-70
207







SP_013
NVTWFHAISGTNGTK
61-75
208







SP_014
HAISGTNGTKRFDNP
66-80
209







SP_027
CEFQFCNDPFLGVYH
131-145
210







SP_028
CNDPFLGVYHKNNKS
136-150
211







SP_029
LGVYHKNNKSWMESE
141-155
212







SP_095
EIYQAGSTPCNGVKG
471-485
213







SP_096
GSTPCNGVKGFNCYF
476-490
214







SP_097
NGVKGFNCYFPLQPY
481-495
215







SP_098
FNCYFPLQPYGFQPT
486-500
216







SP_099
PLQPYGFQPTYGVGY
491-505
217







SP_100
GFQPTYGVGYQPYRV
496-510
218







SP_101
YGVGYQPYRVVVLSF
501-515
219







SP_112
NKKFLPFQQFGRDID
556-570
220







SP_113
PFQQFGRDIDDTTDA
561-575
221







SP_114
GRDIDDTTDAVRDPQ
566-580
222







SP_121
GTNTSNQVAVLYQGV
601-615
223







SP_122
NQVAVLYQGVNCTEV
606-620
224







SP_123
LYQGVNCTEVPVAIH
611-625
225







SP_135
CASYQTQTNSHRRAR
671-685
226







SP_136
TQTNSHRRARSVASQ
676-690
227







SP_137
HRRARSVASQSIIAY
681-695
228







SP_142
AYSNNSIAIPINFTI
706-720
229







SP_143
SIAIPINFTISVTTE
711-725
230







SP_144
INFTISVTTEILPVS
716-730
231







SP_195
GAISSVLNDILARLD
971-985
232







SP_196
VLNDILARLDKVEAE
976-990
233







SP_197
LARLDKVEAEVQIDR
981-995
234







SP_222
QRNFYEPQIITTHNT
1106-1120
235







SP_223
EPQIITTHNTFVSGN
1111-1125
236







SP_224
TTHNTFVSGNCDVVI
1116-1130
237







SP-237
KEIDRLNEVANNLNE
1181-1195
238







SP_238
LNEVANNLNESLIDL
1186-1200
239







SP_239
NNLNESLIDLQELGK
1191-1205
240

















TABLE 10







Peptide sequences of non-conserved regions 


in SEQUENCE_20H (Beta, V2) (B.1.351)










POOL





SP





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





SP_002
VLLPLVSSQCVNFTT
 6-20
241





SP_003
VSSQCVNFTTRTQLP
11-25
242





SP_004
VNFTTRTQLPPAYTN
16-30
243





SP_014
HAIHVSGTNGTKRFA
66-80
244





SP_015
SGTNGTKRFANPVLP
71-85
245





SP_016
TKRFANPVLPFNDGV
76-90
246





SP_041
FKIYSKHTPINLVRG
201-215
247





SP_042
KHTPINLVRGLPQGF
206-220
248





SP_043
NLVRGLPQGFSALEP
211-225
249





SP_047
IGINITRFQTLHRSY
231-245
250





SP_048
TRFQTLHRSYLTPGD
236-250
251





SP_049
LHRSYLTPGDSSSGW
241-255
252





SP_082
EVRQIAPGQTGNIAD
406-420
253





SP_083
APGQTGNIADYNYKL
411-425
254





SP_084
GNIADYNYKLPDDFT
416-430
255





SP_095
EIYQAGSTPCNGVKG
471-485
256





SP_096
GSTPCNGVKGFNCYF
476-490
257





SP_097
NGVKGFNCYFPLQSY
481-495
258





SP_099
PLQSYGFQPTYGVGY
491-505
259





SP_100
GFQPTYGVGYQPYRV
496-510
260





SP_101
YGVGYQPYRVVVLSF
501-515
261





SP_121
GTNTSNQVAVLYQGV
601-615
262





SP_122
NQVAVLYQGVNCTEV
606-620
263





SP_123
LYQGVNCTEVPVAIH
611-625
264





SP_139
SIIAYTMSLGVENSV
691-705
265





SP_140
TMSLGVENSVAYSNN
696-710
266





SP_141
VENSVAYSNNSIAIP
701-715
267
















TABLE 11







Peptide sequences of non-conserved regions 


in SEQUENCE_20J (Gamma, V3) (P.1)












POOL 






SP






Peptide 

A.A
SEQ ID



Number
A.A Sequence
Position
Number







SP_002
VLLPLVSSQCVNFTN
 6-20
268







SP_003
VSSQCVNFTNRTQLP
11-25
269







SP_004
VNFTNRTQLPSAYTN
16-30
270







SP_005
RTQLPSAYTNSFTRG
21-35
271







SP_006
SAYTNSFTRGVYYPD
26-40
272







SP_026
VVIKVCEFQFCNYPF
126-140
273







SP_027
CEFQFCNYPFLGVYY
131-145
274







SP_028
CNYPFLGVYYHKNNK
136-150
275







SP_036
LMDLEGKQGNFKNLS
176-190
276







SP_037
GKQGNFKNLSEFVFK
181-195
277







SP_038
FKNLSEFVFKNIDGY
186-200
278







SP_082
EVRQIAPGQTGTIAD
406-420
279







SP_083
APGQTGTIADYNYKL
411-425
280







SP_084
GTIADYNYKLPDDFT
416-430
281







SP_095
EIYQAGSTPCNGVKG
471-485
282







SP_096
GSTPCNGVKGFNCYF
476-490
283







SP_097
NGVKGFNCYFPLQSY
481-495
284







SP_099
PLQSYGFQPTYGVGY
491-505
285







SP_100
GFQPTYGVGYQPYRV
496-510
286







SP_101
YGVGYQPYRVVVLSF
501-515
287







SP_121
GTNTSNQVAVLYQGV
601-615
288







SP_122
NQVAVLYQGVNCTEV
606-620
289







SP_123
LYQGVNCTEVPVAIH
611-625
290







SP_129
NVFQTRAGCLIGAEY
641-655
291







SP_130
RAGCLIGAEYVNNSY
646-660
292







SP_131
IGAEYVNNSYECDIP
651-665
293







SP_204
AEIRASANLAAIKMS
1016-1030
294







SP_205
SANLAAIKMSECVLG
1021-1035
295







SP_206
AIKMSECVLGQSKRV
1026-1040
296







SP_234
LGDISGINASFVNIQ
1166-1180
297







SP_235
GINASFVNIQKEIDR
1171-1185
298







SP_236
FVNIQKEIDRLNEVA
1176-1190
299

















TABLE 12







Peptide sequences of non-conserved regions 


in SEQUENCE_21B (Kappa) (B.1.617.1)












POOL 






SP






Peptide 

A.A
SEQ ID



Number
A.A Sequence
Position
Number







SP_017
NPVLPFNDGVYFASI
81-95
300







SP_018
FNDGVYFASIEKSNI
 86-100
301







SP_019
YFASIEKSNIIRGWI
 91-105
302







SP_027
CEFQFCNDPFLDVYY
131-145
303







SP_028
CNDPFLDVYYHKNNK
136-150
304







SP_029
LDVYYHKNNKSWMKS
141-155
305







SP_030
HKNNKSWMKSEFRVY
146-160
306







SP_031
SWMKSEFRVYSSANN
151-165
307







SP_089
LDSKVGGNYNYRYRL
441-455
308







SP_090
GGNYNYRYRLFRKSN
446-460
309







SP_091
YRYRLFRKSNLKPFE
451-465
310







SP_095
EIYQAGSTPCNGVQG
471-485
311







SP_096
GSTPCNGVQGFNCYF
476-490
312







SP_097
NGVQGFNCYFPLQSY
481-495
313







SP_121
GTNTSNQVAVLYQGV
601-615
314







SP_122
NQVAVLYQGVNCTEV
606-620
315







SP_123
LYQGVNCTEVPVAIH
611-625
316







SP_135
CASYQTQTNSRRRAR
671-685
317







SP_136
TQTNSRRRARSVASQ
676-690
318







SP_137
RRRARSVASQSIIAY
681-695
319







SP_213
VFLHVTYVPAHEKNF
1061-1075
320







SP_214
TYVPAHEKNFTTAPA
1066-1080
321







SP_215
HEKNFTTAPAICHDG
1071-1085
322

















TABLE 13







Peptide sequences of non-conserved regions 


in SEQUENCE_20B/S.484K (P.2)












POOL 






SP






Peptide 

A.A
SEQ ID



Number
A.A Sequence
Position
Number







SP_095
EIYQAGSTPCNGVKG
471-485
323







SP_096
GSTPCNGVKGFNCYF
476-490
324







SP_097
NGVKGFNCYFPLQSY
481-495
325







SP_121
GTNTSNQVAVLYQGV
601-615
326







SP_122
NQVAVLYQGVNCTEV
606-620
327







SP_123
LYQGVNCTEVPVAIH
611-625
328







SP_234
LGDISGINASFVNIQ
1166-1180
329







SP_235
GINASFVNIQKEIDR
1171-1185
330







SP_236
FVNIQKEIDRLNEVA
1176-1190
331

















TABLE 14







Peptide sequences of non-conserved regions 


in SEQUENCE_21C (Epsilon) (B.1.427/9)










POOL 





SP





Peptide 

A.A 
SEQ ID


Number
A.A Sequence
Position
Number





SP_001
MFVFLVLLPLVSIQC
 1-15
332





SP_002
VLLPLVSIQCVNLTT
 6-20
333





SP_003
VSIQCVNLTTRTQLP
11-25
334





SP_029
LGVYYHKNNKSCMES
141-155
335





SP_030
HKNNKSCMESEFRVY
146-160
336





SP_031
SCMESEFRVYSSANN
151-165
337





SP_089
LDSKVGGNYNYRYRL
441-455
338





SP_090
GGNYNYRYRLFRKSN
446-460
339





SP_091
YRYRLFRKSNLKPFE
451-465
340





SP_121
GTNTSNQVAVLYQGV
601-615
341





SP_122
NQVAVLYQGVNCTEV
606-620
342





SP_123
LYQGVNCTEVPVAIH
611-625
343
















TABLE 15







Peptide sequences of non-conserved regions 


in SEQUENCE_21D (Eta) (B.1.525)










POOL 





SP





Peptide 

A.A 
SEQ ID


Number
A.A Sequence
Position
Number





SP_009
KVFRSSVLHSTRDLF
41-55
344





SP_010
SVLHSTRDLFLPFFS
46-60
345





SP_011
TRDLFLPFFSNVTWF
51-65
346





SP_012
LPFFSNVTWFHVISG
56-70
347





SP 013
NVTWFHVISGTNGTK
61-75
348





SP_014
HVISGTNGTKRFDNP
66-80
349





SP_027
CEFQFCNDPFLGVYH
131-145
350





SP_028
CNDPFLGVYHKNNKS
136-150
351





SP-029
LGVYHKNNKSWMESE
141-155
352





SP_095
EIYQAGSTPCNGVKG
471-485
353





SP_096
GSTPCNGVKGFNCYF
476-490
354





SP_097
NGVKGFNCYFPLQSY
481-495
355





SP_121
GTNTSNQVAVLYQGV
601-615
356





SP_122
NQVAVLYQGVNCTEV
606-620
357





SP-123
LYQGVNCTEVPVAIH
611-625
358





SP_134
IGAGICASYQTHTNS
666-680
359





SP_135
CASYQTHTNSPRRAR
671-685
360





SP_136
THTNSPRRARSVASQ
676-690
361





SP_176
ALLAGTITSGWTLGA
876-890
362





SP_177
TITSGWTLGAGAALQ
881-895
363





SP_178
WTLGAGAALQIPFAM
886-900
364
















TABLE 16







Peptide sequences of non-conserved regions 


in SEQUENCE_21F (Iota) (B.1.526)










POOL 





SP





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





SP_001
MFVFFVLLPLVSSQC
 1-15
365





SP_017
NPVLPFNDGVYFASI
81-95
366





SP_018
FNDGVYFASIEKSNI
 86-100
367





SP_019
YFASIEKSNIIRGWI
 91-105
368





SP_049
LLALHRSYLTPGGSS
241-255
369





SP_050
RSYLTPGGSSSGWTA
246-260
370





SP_051
PGGSSSGWTAGAAAY
251-265
371





SP_095
EIYQAGSTPCNGVKG
471-485
372





SP_096
GSTPCNGVKGFNCYF
476-490
373





SP_097
NGVKGFNCYFPLQSY
481-495
374





SP_121
GTNTSNQVAVLYQGV
601-615
375





SP_122
NQVAVLYQGVNCTEV
606-620
376





SP_123
LYQGVNCTEVPVAIH
611-625
377





SP_139
SIIAYTMSLGVENSV
691-705
378





SP_140
TMSLGVENSVAYSNN
696-710
379





SP_141
VENSVAYSNNSIAIP
701-715
380
















TABLE 17







Peptide sequences of non-conserved regions 


in SEQUENCE_21G (Lambda) (C.37)










POOL 





SP





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





SP_013
NVTWFHAIHVSGTNV
61-75
381





SP_014
HAIHVSGTNVIKRFD
66-80
382





SP_015
SGTNVIKRFDNPVLP
71-85
383





SP_016
IKRFDNPVLPFNDGV
76-90
384





SP_048
TRFQTLLALHNSSSG
236-250
385





SP_049
LLALHNSSSGWTAGA
241-255
386





SP_050
NSSSGWTAGAAAYYV
246-260
387





SP_089
LDSKVGGNYNYQYRL
441-455
388





SP_090
GGNYNYQYRLFRKSN
446-460
389





SP_091
YQYRLFRKSNLKPFE
451-465
390





SP_096
GSTPCNGVEGFNCYS
476-490
391





SP_097
NGVEGFNCYSPLQSY
481-495
392





SP_098
FNCYSPLQSYGFQPT
486-500
393





SP_121
GTNTSNQVAVLYQGV
601-615
394





SP_122
NQVAVLYQGVNCTEV
606-620
395





SP_123
LYQGVNCTEVPVAIH
611-625
396





SP_170
ARDLICAQKFNGLNV
846-860
397





SP_171
CAQKFNGLNVLPPLL
851-865
398





SP_172
NGLNVLPPLLTDEMI
856-870
399
















TABLE 18







Peptide sequences of non-conserved regions  


in SEQUENCE_21H (B.1.621)










POOL SP





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





SP_017
NPVLPFNDGVYFASI
81-95
400





SP_018
FNDGVYFASIEKSNI
 86-100
401





SP_019
YFASIEKSNIIRGWI
 91-105
402





SP_027
CEFQFCNDPFLGVSN
131-145
403





SP_028
CNDPFLGVSNHKNNK
136-150
404





SP_029
LGVSNHKNNKSWMES
141-155
405





SP_068
CPFGEVFNATKFASV
336-350
406





SP_069
VFNATKFASVYAWNR
341-355
407





SP_070
KFASVYAWNRKRISN
346-360
408





SP_095
EIYQAGSTPCNGVKG
471-485
409





SP_096
GSTPCNGVKGFNCYF
476-490
410





SP_097
NGVKGFNCYFPLQSY
481-495
411





SP_099
PLQSYGFQPTYGVGY
491-505
412





SP_100
GFQPTYGVGYQPYRV
496-510
413





SP_101
YGVGYQPYRVVVLSF
501-515
414





SP_121
GTNTSNQVAVLYQGV
601-615
415





SP_122
NQVAVLYQGVNCTEV
606-620
416





SP_123
LYQGVNCTEVPVAIH
611-625
417





SP_135
CASYQTQTNSHRRAR
671-685
418





SP_136
TQTNSHRRARSVASQ
676-690
419





SP_137
HRRARSVASQSIIAY
681-695
420





SP_188
DSLSSTASALGKLQN
936-950
421





SP_189
TASALGKLQNVVNQN
941-955
422





SP_190
GKLQNVVNQNAQALN
946-960
423
















TABLE 19







Peptide sequences of non-conserved regions 


in SEQUENCE_20A/S:126A (B.1.620)












POOL SP






Peptide 

A.A
SEQ ID



Number
A.A Sequence
Position
Number







SP_004
VNLTTRTQLPSAYTN
16-30
424







SP_005
RTQLPSAYTNSFTRG
21-35
425







SP_006
SAYTNSFTRGVYYPD
26-40
426







SP_012
LPFFSNVTWFHAISG
56-70
427







SP_013
NVTWFHAISGTNGTK
61-75
428







SP_014
HAISGTNGTKRFDNP
66-80
429







SP_024
SLLIVNNATNAVIKV
116-130
430







SP_025
NNATNAVIKVCEFQF
121-135
431







SP_026
AVIKVCEFQFCNDPF
126-140
432







SP_027
CEFQFCNDPFLGVYH
131-145
433







SP_028
CNDPFLGVYHKNNKS
136-150
434







SP_029
LGVYHKNNKSWMESE
141-155
435







SP_047
IGINITRFQTLYRSY
231-245
436







SP_048
TRFQTLYRSYLTPGD
236-250
437







SP_049
LYRSYLTPGDSSSGW
241-255
438







SP_094
RDISTEIYQAGNTPC
466-480
439







SP_095
EIYQAGNTPCNGVKG
471-485
440







SP_096
GNTPCNGVKGFNCYF
476-490
441







SP_097
NGVKGFNCYFPLQSY
481-495
442







SP_121
GTNTSNQVAVLYQGV
601-615
443







SP_122
NQVAVLYQGVNCTEV
606-620
444







SP_123
LYQGVNCTEVPVAIH
611-625
445







SP_135
CASYQTQTNSHRRAR
671-685
446







SP_136
TQTNSHRRARSVASQ
676-690
447







SP_137
HRRARSVASQSIIAY
681-695
448







SP_204
AEIRASANLAAIKMS
1016-1030
449







SP_205
SANLAAIKMSECVLG
1021-1035
450







SP_206
AIKMSECVLGQSKRV
1026-1040
451







SP_222
QRNFYEPQIITTHNT
1106-1120
452







SP_223
EPQIITTHNTFVSGN
1111-1125
453







SP_224
TTHNTFVSGNCDVVI
1116-1130
454










An embodiment is shown in schematic diagram FIG. 5A. The inventors designed peptide pools containing peptides that cover the whole Spike-Wuhan protein (Pool A; 253 peptides of 15 amino acids in length, overlapping adjacent peptides by 10 amino acids, derived from SEQ ID NO: 795) and the non-conserved Spike-Wuhan regions affected by mutations present in the delta variant (Pool B; Table 7). The third peptide pool (Pool C; Table 8) contains peptides from Pool B with the amino acid mutations present in the Spike-Delta.


These peptide pools can be, for example, used in a classical ELISPOT assay and thus used to stimulate PBMC of different vaccinated individuals (FIG. 5B). The number of spots obtained in each experiment is analyzed and utilized to derive in each single individual, the frequency of T cells directed towards the whole Spike (PBMC stimulated with peptide pool A), the frequency of T cells directed toward the non-conserved Spike-Wuhan region (PBMC stimulated with Pool B) and the frequency of T cells inhibited by AA mutations present in these mutated Spike-Delta region (PBMC stimulated with pool C). Overall, the test provides the estimation of the ability of T cells of a given individual to recognize the conserved and non-conserved region of different Spike proteins and the ability of mutations to inhibit the T cell response towards Spike. This experimental system can be done by utilizing different peptide pools covering other mutated SARS-CoV-2 proteins (i.e., NP, M).


Thus, the inventors can obtain a measurement of the alteration that the mutations present in VOCs can exert on total SARS-CoV-2 T cell response.


Example 4
PCR-Based Test to Detect Virus Cellular Immunity

The inventors show in this Example that qPCR can be used to quantify the presence of virus-specific T cells, based on ex vivo stimulation of whole blood samples with a pool of viral peptides covering the spike or other SARS-CoV-2 viral proteins (i.e. nucleoprotein [NP]), followed by direct amplification of IFN-γ or IL-2 (directly produced by SARS-CoV-2 antigen-specific T cells) or CXCL10, a molecule expressed by monocytes in response to T cell activation.


In order to select genes whose induction would correlate with the presence and activation of antigen specific T cells; we first evaluated the transcriptional profile of whole blood after overnight stimulation with SARS-CoV-2 peptide pools by RNA sequencing (FIG. 6A). This initial cohort consisted of 7 naïve and 11 COVID-19 convalescent subjects. Briefly, whole blood was incubated overnight with DMSO or multiple pools of SARS-CoV-2 peptides, including three distinct pools of the spike (S) protein, corresponding to the first 100 peptides covering the first 510 amino acids, and two distinct pools of the structural nucleocapsid protein (NP-1 and NP-2, Tables 2 and 3). The full 253 spike peptides were divided into 7 peptide pools of around 35˜ peptides each. The first 3 pools, comprising the first 100 peptides, cover the S1 chain of the spike protein. RNA was extracted from the cell pellet as described in Example 2, and subjected to Illumina single end sequencing (Koh, C. M. et al., Nature 523: 96-100 (2015))20. We then identified genes activated by viral peptides by performing a differential expression analysis between peptide-stimulated samples and untreated controls, across all subjects (FIG. 6B). Treatment with the S1 pool induced the largest changes in gene expression, with over 600 genes significantly upregulated (FDR<0.05, log 2FoldChange>1) across naïve and SARS-CoV-2 convalescent subjects. However, about half of these genes were shared across the two groups, suggesting that the transcriptional effects induced by this pool of peptides are not highly specific to previous exposure to SARS-CoV-2. NP2 treatment induced the most specific response, with 63 genes uniquely upregulated in convalescent individuals, and only 15 in naïve individuals and 11 shared between groups. Not surprisingly, these upregulated genes belonged to “cellular response to interferon gamma signaling”, “response to cytokine” and “Jak/Stat signaling” pathways. To narrow down a shortlist of candidates for further investigation by qPCR, we selected a panel of 10 genes that were significantly upregulated following either S1 or NP2 stimulation in convalescent subjects, but not as significantly in naïve subjects (FIG. 6C). Upregulation of these genes was further confirmed by RNA-seq in a validation cohort of eight COVID-19 convalescent subjects (FIG. 7).


qPCR


Next, we validated transcriptional induction of shortlisted genes by qPCR (as described in Example 2) in 11 naïve and 8 COVID-19 convalescent subjects. Out of all genes tested (FIG. 8A), we selected CXCL10, IFN-γ and IL2 for further studies. Between the three candidate genes, CXCL10 was not only the most reproducible, but it was the only gene that could accurately distinguish naïve and COVID-19 recovered individuals using multiple qPCR machines, including the QuantStudio 5 (Applied Biosystems), CFX96 (BioRad), CFX384 (BioRad), and bCUBE 2.0 (Hyris) (FIG. 8A-B). We observed that for IFN-γ and IL2, we were able to categorize patients if the assay was performed using the Hyris bCUBE. This instrument is a portable, 2-channel machine that is able to quantify up to 36 wells at a time.


To assess the reliability of the qTACT test, we compared CXCL10 and IFN-γ expression levels to IFN-γ cytokine secretion as quantified by ELLA in a larger cohort comprised of 89 subjects (43 naïve and 46 COVID-19 convalescent. For this cohort, only samples stimulated with DMSO (control) and the spike peptide pool (Table 4) were considered for downstream analysis. CXCL10 was selected as preferred because our previous data had confirmed its reliability and reproducibility when distinguishing between naïve and COVID-19 convalescent subjects. For this larger cohort, we chose to keep IFN-γ, despite its inferiority relative to CXCL10, to include a gene that is expressed by antigen-specific T cells and to directly correlate mRNA expression (qTACT) with IFN-γ protein secretion (ELLA). The cohort was recruited prior to vaccination and followed at day 10 and 20 after the first and second dose of the BNT162b2 vaccine and the data on IFN-γ an IL-2 cytokine secretion have been described elsewhere (Camara C., et al., bioRxiv 2021.03.22.436441; doi:/10.1101/2021.03.22.436441).


Compared to naïve subjects, COVID-19 recovered individuals had a higher median expression of CXCL10 prior to vaccination (2.53 [N=21] vs 0.0087 [N=19] in naïve subjects) (FIG. 9A). A similar trend was observed for IFN-γ levels (median pre-vaccination −0.00185 [N=18] in naïve and 0.0036 [N=19] in COVID-19 recovered subjects) but the difference was not statistically significant (FIG. 9B. Quantification of the spike-specific T cell response by both CXCL10 and IFN-γ qPCR 10 days after the first dose indicates that naïve subjects mount a weaker response compared to COVID-19 recovered individuals (FIG. 9A-B. These results are consistent with data obtained by direct IFN-γ and IL-2 cytokine quantification (Camara C., et al., bioRxiv 2021.03.22.436441; doi: 10.1101/2021.03.22.436441). The technical advantage over the use of ELISA or ELISPOT, is the ease of use of qPCR and, importantly, the internal normalization standard (i.e., ACTIN levels), which is absent in other more laborious methods of quantifying cellular immunity.


A Second Vaccine Dose Increases CXCL10 and IFN-v Expression Levels in Naiive Subjects but not in COVID-19 Recovered Individuals

The effect of a second dose of the vaccine was next studied. Sampling on day 10 and 20 after a second dose confirmed the beneficial effects of the recall vaccine in naïve individuals who increase their CXCL10 and IFN-γ expression to significant levels. On the contrary, the second dose in COVID-19 recovered individuals did not have a boost effect (no significant increase in CXCL10 and IFN-γ levels) (FIG. 9A-B). These findings indicate that while naïve subjects significantly increase their cytokine production induced by SARS-CoV-2 spike protein after the second dose of the vaccine, COVID-19 recovered individuals do not further increase IFN-γ and CXCL10 levels following the standard regimen for COVID-19 vaccination, consistent with previously published work quantifying cytokine production (Camara C., et al., bioRxiv 2021.03.22.436441; doi: 10.1101/2021.03.22.436441; Tauzin, A. et al., bioRxiv. 2021 Mar. 18; 2021.03.18.435972. doi: 10.1101/2021.03.18.435972). Consistent with what we previously reported (Camara C., et al., bioRxiv 2021.03.22.436 441; doi: 10.1101/2021.03.22.43644116), both CXCL10 and IFN-γ mRNA levels induced by the spike peptide pool strongly correlate with IFN-γ cytokine quantification in the same cohort (Spearman r=0.474, and r=0.513, respectively; p<0.0001)(FIG. 10).


qPCR Quantification of CXCL10 as a Proxy for Cellular Immunity


We used the qPCR assay to quantify the levels of CXCL10, as a proxy for cellular immunity, over time. We assessed the level of T cell activation in a small cohort of COVID-19 convalescent subjects at different time points post infection. We observed that, while for some patients CXCL10 levels decreased to background levels after 9 months from SARS-CoV-2 infection, we were able to detect the activation of antigen specific T cells in the majority of subjects even 9-12 months post infection (FIG. 8C).


dqPCR


Blood samples were prepared as described in Example 2. To this end, following overnight incubation with DMSO control, or SARS-CoV-2 nucleoprotein or spike peptide pools (Table 4), we took 50 μl of blood, diluted it (1:3) to avoid PCR inhibition by anticoagulants (i.e., heparin), and loaded 2 μl directly onto a qPCR instrument (dqTACT)(FIG. 6A). For this cohort, we chose to once again include the nucleoprotein (NP-2, Table 3), this time to act as an additional negative control for vaccinated subjects. All of the vaccinated subjects that were selected had never been exposed to the virus, thus, we expected that they would not mount a cellular immune response to anything but the spike protein, upon which the mRNA vaccinations are based.


IFN-γ and CXCL10 were tested as potential readouts for the dqTACT assay (described in Example 2), being optimized for use on the Hyris bCUBE given its high range of detection compared to other tested instruments, the reduced cost, and the ease of assay set up (FIG. 11A). Our results showed that IFN-γ could not reliably stratify naïve and vaccinated individuals (data not shown), while CXCL10 did so robustly (FIG. 11B). Without being bound by theory, the low abundance of IFN-γ is likely due to the small number of antigen-specific T cells in whole blood, which are the direct source of IFN-gamma. In contrast, CXCL10, being an IFN-gamma-stimulated chemokine, is upregulated and expressed by a much higher percentage of cells (i.e. monocytes and neutrophils, which are roughly 5% and 60%, respectively, of all white cells in whole blood) (Ichikawa, A. et al., Am J Respir Crit Care Med 187: 65-77 (2013); Luster, A. D., Nature 315: 672-676 (1985)). Therefore, when taking only a small volume of blood, CXCL10 is not subject to sampling bias as is IFN-γ or other antigen-specific T cell transcripts (0.1% of all white cells in whole blood). In addition, we decided to compare the level of cytokine production in the same cohort. First, we quantified TNFa, CXCL10 (IP-10), IFN-γ and IL-2 by ELLA. All cytokines, except TNFa, successfully stratified naïve from vaccinated subjects (FIG. 11C and FIG. 12A-C), and correlate well with CXCL10 mRNA quantification obtained by the dqTACT assay (FIG. 12D-G).


A lack of rapid, accessible, and accurate diagnostic methods to quantify cellular immunity hinders long-term vaccination strategies and public health responses to global pandemics, such as the one being caused by SARS-CoV-2. Considering that diagnostic centers around the world have ramped up the setup of RT-qPCR based facilities, we developed a qPCR-based dqTACT assay, which is amenable to periodic and repeated testing of patient samples, as it requires only 1 ml of blood and a 24-hour turnaround time. Clinical validation of this assay in response to recent draft guidance from the United States Food and Drug Administration (FDA) and European Medicines Agency (EMA) is ongoing in a Clinical Laboratory Improvement Amendments (CLIA)-certified microbiology laboratory. First, we implemented a Next Generation Sequencing (NGS) approach. The pros of this approach, which could be further implemented by a targeted amplification panel of 15-20 genes, is the possibility of capturing the variability of the response and measure cytokines produced by both T cells and other myeloid cells in the blood. The cons are a longer turnaround time, a higher cost, and the need for skilled technical personnel.


Second, we developed a qPCR-based method (qTACT assay) on a 96 or 384 well platform (BioRad CFX). The advantages of this approach are the accuracy and sensitivity of qPCR probes, the opportunity to combine more than 2 fluorophores to measure the expression of 2-4 genes, and the scalability and potential automation of the process. The cons include a relatively longer processing time (48 hours per 200 samples), the need to purify RNA (by standard RNA-purification kits/columns), higher associated costs, and a certain level of technical skill (although less than that required for NGS).


Third, we optimized a direct qPCR-based method (dqTACT assay) on the HYRIS bCUBE platform. The advantages of this approach are the accuracy of qPCR probes, the increased accuracy of the bCUBE platform over other tested qPCR machines, and the reduced processing time/cost/skill required. Overall, this is an easy to implement protocol that requires minimal training of the operator, thus reducing technical errors. The cons include a relatively lower scalability (18 samples on the bCUBE as opposed to 48/192 samples on the CFX 96/384) and the limitation to a 2 fluorophore/2 genes detection system.


The derived profile of SARS-CoV-2-specific T cell activation by qTACT/dqTACT assays in different cohorts of naïve, infected or vaccinated individuals, will provide information about their level of SARS-CoV-2-specific cellular immunity.


Example 5
HBV Peptide Pools

Eight HBV peptide pools of 15-mers (Tables 20-27) covering the proteome (Core, X, Envelope and Polymerase) of HBV (AB112063 (HBV Gen C)) were generated (FIG. 13A). The Core protein has 212 amino acids, so it requires 41 15-mer peptides overlapping by 10 amino acids to cover the whole protein, resulting in a single peptide pool; X has 154 amino acids, so it requires 29 15-mer peptides overlapping by 10 amino acids to cover the whole protein, resulting in a single peptide pool; Envelope has 389 amino acids, so it requires 76 15-mer peptides overlapping by 10 amino acids to cover the whole protein, resulting in a 2 peptide pools of about 40 peptides each; Polymerase has 843 amino acids, so it requires 167 15-mer peptides overlapping by 10 amino acids to cover the whole protein, resulting in 4 peptide pools of about 40 peptides each.









TABLE 20







Summary of Peptide Pool C (core).










POOL C





Peptide

A.A 
SEQ ID


Number
A.A Sequence
Position
Number





C_1
MQLFHLCLIISCLRP
 1-15
480





C_2
LCLIISCLRPTVQAS
 6-20
481





C_3
SCLRPTVQASKLCLG
11-25
482





C_4
TVQASKLCLGWLWGM
16-30
483





C_5
KLCLGWLWGMDIDPY
21-35
484





C 6
WLWGMDIDPYKEFGA
26-40
485





C_7
DIDPYKEFGASVELL
31-45
486





C_8
KEFGASVELLSFLPS
36-50
487





C_9
SVELLSFLPSDFFPN
41-55
488





C_10
SFLPSDFFPNIRDLL
46-60
489





C_11
DFFPNIRDLLDTASA
51-65
490





C_12
IRDLLDTASALFREA
56-70
491





C_13
DTASALFREALESPE
61-75
492





C_14
LFREALESPEHCTPH
66-80
493





C_15
LESPEHCTPHHTAIR
71-85
494





C_16
HCTPHHTAIRQAILC
76-90
495





C_17
HTAIRQAILCWGELM
81-95
496





C_18
QAILCWGELMNLATW
 86-100
497





C_19
WGELMNLATWVGSNL
 91-105
498





C_20
NLATWVGSNLEDPAS
 96-110
499





C_21
VGSNLEDPASRELVV
101-115
500





C_22
EDPASRELVVGYVNV
106-120
501





C_23
RELVVGYVNVNMGLK
111-125
502





C_24
GYVNVNMGLKIRQLL
116-130
503





C_25
NMGLKIRQLLWFHIS
121-135
504





C_26
IRQLLWFHISCLTFG
126-140
505





C_27
WFHISCLTFGRETVL
131-145
506





C_28
CLTFGRETVLEYLVS
136-150
507





C_29
RETVLEYLVSFGVWI
141-155
508





C_30
EYLVSFGVWIRTPPA
146-160
509





C_31
FGVWIRTPPAYRPPN
151-165
510





C_32
RTPPAYRPPNAPILS
156-170
511





C_33
YRPPNAPILSTLPET
161-175
512





C_34
APILSTLPETTVVRR
166-180
523





C_35
TLPETTVVRRRGRSP
171-185
514





C_36
TVVRRRGRSPRRRTP
176-190
515





C_37
RGRSPRRRTPSPRRR
181-195
516





C_38
RRRTPSPRRRRSQSP
186-200
517





C_39
SPRRRRSQSPRRRRS
191-205
518





C_40
RSQSPRRRRSQSRGS
196-210
519





C_41
RRRRSQSRGSQC
201-215
520
















TABLE 21







Summary of Peptide Pool X.










POOL X





Peptide

A.A
SEQ ID


Number
A.A Sequence
Position
Number





X_1
MAARLCCQLDPARDV
 1-15
521





X_2
CCQLDPARDVLCLRP
 6-20
522





X_3
PARDVLCLRPVGAES
11-25
523





X_4
LCLRPVGAESRGRPV
16-30
524





X_5
VGAESRGRPVSGAFG
21-35
525





X_6
RGRPVSGAFGTLPSP
26-40
526





X_7
SGAFGTLPSPSSSAV
31-45
527





X_8
TLPSPSSSAVPTDHG
36-50
528





X_9
SSSAVPTDHGAHLSL
41-55
529





X_10
PTDHGAHLSLRGLPV
46-60
530





X_11
AHLSLRGLPVCAFSS
51-65
531





X_12
RGLPVCAFSSAGPCA
56-70
532





X_13
CAFSSAGPCALRFTS
61-75
533





X_14
AGPCALRFTSARRME
66-80
534





X_15
LRFTSARRMETTVNA
71-85
535





X_16
ARRMETTVNARQVLP
76-90
536





X_17
TTVNARQVLPKVLHK
81-95
537





X_18
RQVLPKVLHKRTLGL
 86-100
538





X_19
KVLHKRTLGLSAMST
 91-105
539





X_20
RTLGLSAMSTTDLEA
 96-110
540





X_21
SAMSTTDLEAYFKDR
101-115
541





X_22
TDLEAYFKDRVFKDW
106-120
542





X_23
YFKDRVFKDWEELGE
111-125
543





X_24
VFKDWEELGEETRLM
116-130
544





X_25
EELGEETRLMIFVLG
121-135
545





X_26
ETRLMIFVLGGCRHK
126-140
546





X_27
IFVLGGCRHKLVCSP
131-145
547





X_28
GCRHKLVCSPAPCNF
136-150
548





X_29
LVCSPAPCNFFTSA
141-155
549
















TABLE 22







Summary of Peptide Pool E1 (envelope).










POOL E-1





Peptide

A.A
SEQ ID


Number
A.A Sequence
Position
Number





E_1
MGTNLSVPNPLGFFP
 1-15
550





E_2
SVPNPLGFFPDHQLD
 6-20
551





E_3
LGFFPDHQLDPAFGA
11-25
552





E_4
DHQLDPAFGANSNNP
16-30
553





E_5
PAFGANSNNPDWDFN
21-35
554





E_6
NSNNPDWDFNPNKDQ
26-40
555





E_7
DWDFNPNKDQWPAAN
31-45
556





E_8
PNKDQWPAANQVGVG
36-50
557





E_9
WPAANQVGVGSFGPG
41-55
558





E_10
QVGVGSFGPGFTPPH
46-60
559





E_11
SFGPGFTPPHGNLLG
51-65
560





E_12
FTPPHGNLLGWSPQA
56-70
561





E_13
GNLLGWSPQAQGILT
61-75
562





E_14
WSPQAQGILTTVPAA
66-80
563





E_15
QGILTTVPAAPPPAS
71-85
564





E_16
TVPAAPPPASTNRQS
76-90
565





E_17
PPPASTNRQSGRQPT
81-95
566





E_18
TNRQSGRQPTPISLP
 86-100
567





E_19
GRQPTPISLPLRDSH
 91-105
568





E_20
PISLPLRDSHPQAMQ
 96-110
569





E_21
LRDSHPQAMQWNSST
101-115
570





E_22
PQAMQWNSSTFHQAL
106-120
571





E_23
WNSSTFHQALLDPKV
111-125
572





E_24
FHQALLDPKVRGLYL
116-130
573





E_25
LDPKVRGLYLPAGGS
121-135
574





E_26
RGLYLPAGGSSSGTV
126-140
575





E_27
PAGGSSSGTVNPVQT
131-145
576





E_28
SSGTVNPVQTTASPI
136-150
577





E_29
NPVQTTASPISSIFS
141-155
578





E_30
TASPISSIFSRTGDP
146-160
579





E_31
SSIFSRTGDPAPNME
151-165
580





E_32
RTGDPAPNMESTTSG
156-170
581





E_33
APNMESTTSGFLGPL
161-175
582





E_34
STTSGFLGPLLVLQA
166-180
583





E_35
FLGPLLVLQAGFFLL
171-185
584





E_36
LVLQAGFFLLTRILT
176-190
585





E_37
GFFLLTRILTIPQSL
181-195
586





E_38
TRILTIPQSLDSWWT
186-200
587
















TABLE 23







Summary of Peptide Pool E2 (envelope).












POOL E-2






Peptide

A.A
SEQ ID



Number
A.A Sequence
Position
Number







E_39
IPQSLDSWWTSLNFL
191-205
588







E_40
DSWWTSLNFLGGAPT
196-210
589







E_41
SLNFLGGAPTCSGQN
201-215
590







E_42
GGAPTCSGQNLQSPT
206-220
591







E_43
CSGQNLQSPTSNHSP
211-225
592







E_44
LQSPTSNHSPTSCPP
216-230
593







E_45
SNHSPTSCPPICPGY
221-235
594







E_46
TSCPPICPGYRWMCL
226-240
595







E_47
ICPGYRWMCLRRFII
231-245
596







E_48
RWMCLRRFIIFLFIL
236-250
597







E_49
RRFIIFLFILLLCLI
241-255
598







E_50
FLFILLLCLIFLLVL
246-260
599







E_51
LLCLIFLLVLLDYQG
251-265
600







E_52
FLLVLLDYQGMLPVC
256-270
601







E_53
LDYQGMLPVCPLLPG
261-275
602







E_54
MLPVCPLLPGTSTTS
266-280
603







E_55
PLLPGTSTTSMGPCK
271-285
604







E_56
TSTTSMGPCKTCTTP
276-290
605







E_57
MGPCKTCTTPAQGTS
281-295
606







E_58
TCTTPAQGTSMFPSC
286-300
607







E_59
AQGTSMFPSCCCTQP
291-305
608







E_60
MFPSCCCTQPSDGNC
296-310
609







E_61
CCTQPSDGNCTCIPI
301-315
610







E_62
SDGNCTCIPIPSSWA
306-320
611







E_63
TCIPIPSSWAFARFL
311-325
612







E_64
PSSWAFARFLWEWAS
316-330
613







E_65
FARFLWEWASVRFSW
321-335
614







E_66
WEWASVRFSWLSLLV
326-340
615







E_67
VRFSWLSLLVPFVQW
331-345
616







E_68
LSLLVPFVQWFVGLS
336-350
617







E_69
PFVQWFVGLSPTVWL
341-355
618







E_70
FVGLSPTVWLSVIWM
346-360
619







E_71
PTVWLSVIWMMWYWG
351-365
620







E_72
SVIWMMWYWGRSLYN
356-370
621







E_73
MWYWGRSLYNILNPF
361-375
622







E_74
RSLYNILNPFLPLLP
366-380
623







E_75
ILNPFLPLLPIFFCL
371-385
624







E_76
LPLLPIFFCLWVYI
386-390
625

















TABLE 24







Summary of Peptide Pool Pol-1 (Polymerase).










POOL 





Pol-1





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





Pol_1
MPLSYQHFRKLLLLD
 1-15
626





Pol_2
QHFRKLLLLDDEAGP
 6-20
627





Pol_3
LLLLDDEAGPLEEEL
11-25
628





Pol_4
DEAGPLEEELPRLAD
16-30
629





Pol_5
LEEELPRLADEGLNH
21-35
630





Pol_6
PRLADEGLNHRVAED
26-40
631





Pol_7
EGLNHRVAEDLNLGD
31-45
632





Pol_8
RVAEDLNLGDLNVSI
36-50
633





Pol_9
LNLGDLNVSIPWTHK
41-55
634





Pol_10
LNVSIPWTHKVGNFT
46-60
635





Pol_11
PWTHKVGNFTGLYSS
51-65
636





Pol_12
VGNFTGLYSSTVPVF
56-70
637





Pol_13
GLYSSTVPVFNPEWK
61-75
638





Pol_14
TVPVFNPEWKTPSFP
66-80
639





Pol_15
NPEWKTPSFPNIHLK
71-85
640





Pol_16
TPSFPNIHLKEDIIN
76-90
641





Pol_17
NIHLKEDIINRCQQY
81-95
642





Pol_18
EDIINRCQQYVGPLT
 86-100
643





Pol_19
RCQQYVGPLTVNEKR
 91-105
644





Pol_20
VGPLTVNEKRRLKVT
 96-110
645





Pol_21
VNEKRRLKVTMPARF
101-115
646





Pol_22
RLKVTMPARFYPNLT
106-120
647





Pol_23
MPARFYPNLTKYLPL
111-125
648





Pol_24
YPNLTKYLPLDKGIK
116-130
649





Pol_25
KYLPLDKGIKPYYPE
121-135
650





Pol_26
DKGIKPYYPEHIVNH
126-140
651





Pol_27
PYYPEHIVNHYFQTR
131-145
652





Pol_28
HIVNHYFQTRHYLHT
136-150
653





Pol_29
YFQTRHYLHTLWKAG
141-155
654





Pol_30
HYLHTLWKAGILYKR
146-160
655





Pol_31
LWKAGILYKRETTRS
151-165
656





Pol_32
ILYKRETTRSASFCG
156-170
657





Pol_33
ETTRSASFCGSPYSW
161-175
658





Pol_34
ASFCGSPYSWEQELQ
166-180
659





Pol_35
SPYSWEQELQHGTLV
171-185
660





Pol_36
EQELQHGTLVFQTST
176-190
661





Pol_37
HGTLVFQTSTRHGDE
181-195
662





Pol_38
FQTSTRHGDESFGSQ
186-200
663





Pol_39
RHGDESFGSQSSGIL
191-205
664





Pol_40
SFGSQSSGILSRSPV
196-210
665





Pol_41
SSGILSRSPVGPGIR
201-215
666





Pol_42
SRSPVGPGIRSQFKQ
206-220
667
















TABLE 25







Summary of Peptide Pool Pol-2 (Polymerase).










POOL 





Pol-2





Peptide

A.A
SEQ ID


Number
A.A Sequence
Position
Number





Pol_43
GPGIRSQFKQSRLGL
211-225
668





Pol_44
SQFKQSRLGLQPQQG
216-230
669





Pol_45
SRLGLQPQQGSMASG
221-235
670





Pol_46
QPQQGSMASGKPGRS
226-240
671





Pol_47
SMASGKPGRSGIIRA
231-245
672





Pol_48
KPGRSGIIRARVHPT
236-250
673





Pol_49
GIIRARVHPTTRQSF
241-255
674





Pol_50
RVHPTTRQSFGVEPA
246-260
675





Pol_51
TRQSFGVEPAGSGHI
251-265
676





Pol_52
GVEPAGSGHIDNSTS
256-270
677





Pol_53
GSGHIDNSTSSASSC
261-275
678





Pol_54
DNSTSSASSCLHQSA
266-280
679





Pol_55
SASSCLHQSAVRKTA
271-285
680





Pol_56
LHQSAVRKTAYSHLS
276-290
681





Pol_57
VRKTAYSHLSTSKRQ
281-295
682





Pol_58
YSHLSTSKRQSSSGH
286-300
683





Pol_59
TSKRQSSSGHAVELQ
291-305
684





Pol_60
SSSGHAVELQHIPPS
296-310
685





Pol_61
AVELQHIPPSSARSQ
301-315
686





Pol_62
HIPPSSARSQSEGPI
306-320
687





Pol_63
SARSQSEGPIPSCWW
311-325
688





Pol_64
SEGPIPSCWWLQFRN
316-330
689





Pol_65
PSCWWLQFRNSKPCS
321-335
690





Pol_66
LQFRNSKPCSDYCLS
326-340
691





Pol_67
SKPCSDYCLSHIVNL
331-345
692





Pol_68
DYCLSHIVNLLEDWG
336-350
693





Pol_69
HIVNLLEDWGPCTEY
341-355
694





Pol_70
LEDWGPCTEYGEHHI
346-360
695





Pol_71
PCTEYGEHHIRIPRT
351-365
696





Pol_72
GEHHIRIPRTPARVT
356-370
697





Pol_73
RIPRTPARVTGGVFL
361-375
698





Pol_74
PARVTGGVFLVDKNP
366-380
799





Pol_75
GGVFLVDKNPHNTTE
371-385
700





Pol_76
VDKNPHNTTESRLVV
386-390
701





Pol_77
HNTTESRLVVDFSQF
381-395
702





Pol_78
SRLVVDFSQFSRGST
386-400
703





Pol_79
DFSQFSRGSTHVFWP
391-405
704





Pol_80
SRGSTHVFWPKFAVP
396-410
705





Pol_81
HVFWPKFAVPNLQSL
401-415
706





Pol_82
KFAVPNLQSLTNLLS
406-419
707





Pol_83
NLQSLTNLLSSNLSW
411-425
708





Pol_84
TNLLSSNLSWLSLDV
416-430
709
















TABLE 26







Summary of Peptide Pool Pol-3 (Polymerase).










POOL 





Pol-3





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





Pol_85
SNLSWLSLDVSAAFY
421-435
710





Pol_86
LSLDVSAAFYHIPLH
426-440
711





Pol_87
SAAFYHIPLHPAAMP
431-445
712





Pol_88
HIPLHPAAMPHLLVG
436-450
713





Pol_89
PAAMPHLLVGSSGLP
441-455
714





Pol_90
HLLVGSSGLPRYVAR
446-460
715





Pol_91
SSGLPRYVARLSSTS
451-465
716





Pol_92
RYVARLSSTSRNINY
456-470
717





Pol_93
LSSTSRNINYQHGTM
461-475
718





Pol_94
RNINYQHGTMQDLHD
466-480
719





Pol_95
QHGTMQDLHDTCSRN
471-485
720





Pol_96
QDLHDTCSRNLYVSL
476-490
721





Pol_97
TCSRNLYVSLLLLYT
481-495
722





Pol_98
LYVSLLLLYTTFGRK
486-500
723





Pol_99
LLLYTTFGRKLHLYS
491-505
724





Pol_100
TFGRKLHLYSHPIIL
496-510
725





Pol_101
LHLYSHPIILGFRKI
501-515
726





Pol_102
HPIILGFRKIPMGVG
506-520
727





Pol_103
GFRKIPMGVGLSPFL
511-525
728





Pol_104
PMGVGLSPFLLAQFT
516-530
729





Pol_105
LSPFLLAQFTSAICS
521-535
730





Pol_106
LAQFTSAICSVVRRA
526-540
731





Pol_107
SAICSVVRRAFPHCL
531-545
732





Pol_108
VVRRAFPHCLAFSYM
536-550
733





Pol_109
FPHCLAFSYMDDVVL
541-555
734





Pol_110
AFSYMDDVVLGAKSV
546-560
735





Pol_111
DDVVLGAKSVQHLES
551-565
736





Pol_112
GAKSVQHLESLFTAV
556-570
737





Pol_113
QHLESLFTAVTNFLL
561-575
738





Pol_114
LFTAVTNFLLSLGVH
566-580
739





Pol_115
TNFLLSLGVHLNPTK
571-585
740





Pol_116
SLGVHLNPTKTKRWG
576-590
741





Pol_117
LNPTKTKRWGYSLNF
581-595
742





Pol_118
TKRWGYSLNFMGYVI
586-600
743





Pol_119
YSLNFMGYVIGSWGT
591-605
744





Pol_120
MGYVIGSWGTLPQEH
596-610
745





Pol_121
GSWGTLPQEHIVHKI
601-615
746





Pol_122
LPQEHIVHKIKQCFR
606-620
747





Pol_123
IVHKIKQCFRKLPLN
611-625
748





Pol_124
KQCFRKLPLNRPIDW
616-630
749





Pol_125
KLPLNRPIDWKVCQR
621-635
750





Pol_126
RPIDWKVCQRIVGLL
626-640
751
















TABLE 27







Summary of Peptide Pool Pol-4 (Polymerase).










POOL 





Pol-4





Peptide 

A.A
SEQ ID


Number
A.A Sequence
Position
Number





Pol_127
KVCQRIVGLLGFAAP
631-645
752





Pol_128
IVGLLGFAAPFTQCG
636-650
753





Pol_129
GFAAPFTQCGYPALM
641-655
754





Pol_130
FTQCGYPALMPLSAC
646-660
755





Pol_131
YPALMPLSACIQAKR
651-665
756





Pol_132
PLSACIQAKRAFTFS
656-670
757





Pol_133
IQAKRAFTFSPTYRA
661-675
758





Pol_134
AFTFSPTYRAFLCKQ
666-680
759





Pol_135
PTYRAFLCKQYMNLY
671-685
760





Pol_136
FLCKQYMNLYPVARQ
676-690
761





Pol_137
YMNLYPVARQRPGLC
681-695
762





Pol_138
PVARQRPGLCQVFAD
686-700
763





Pol_139
RPGLCQVFADATPTG
691-705
764





Pol_140
QVFADATPTGWGLAI
696-710
765





Pol_141
ATPTGWGLAIGHQRM
701-715
766





Pol_142
WGLAIGHQRMRGTFV
706-720
767





Pol_143
GHQRMRGTFVAPLPI
711-725
768





Pol_144
RGTFVAPLPIHTAEL
716-730
769





Pol_145
APLPIHTAELLAACF
721-735
770





Pol_146
HTAELLAACFARSRS
726-740
771





Pol_147
LAACFARSRSGAKLI
731-745
772





Pol_148
ARSRSGAKLIGTDNS
736-750
773





Pol_149
GAKLIGTDNSVVLSR
741-755
774





Pol_150
GTDNSVVLSRKYTSF
746-760
775





Pol_151
VVLSRKYTSFPWLLG
751-765
776





Pol_152
KYTSFPWLLGCAANW
756-770
777





Pol_153
PWLLGCAANWILRGT
761-775
778





Pol_154
CAANWILRGTSFVYV
766-780
779





Pol_155
ILRGTSFVYVPSALN
771-785
780





Pol_156
SFVYVPSALNPADDP
776-790
781





Pol_157
PSALNPADDPSRGRL
781-795
782





Pol_158
PADDPSRGRLGLYRP
786-800
783





Pol_159
SRGRLGLYRPLLRLP
791-805
784





Pol_160
GLYRPLLRLPFRPTT
796-810
785





Pol_161
LLRLPFRPTTGRTSL
801-815
786





Pol_162
FRPTTGRTSLYAVSP
806-820
787





Pol_163
GRTSLYAVSPSVPSH
811-825
788





Pol_164
YAVSPSVPSHLPDRV
816-830
789





Pol_165
SVPSHLPDRVHFASP
821-835
790





Pol_166
LPDRVHFASPLHVAW
826-840
791





Pol_167
HFASPLHVAWRPP
831-843
792









Human Samples

Whole blood was isolated from either a patient with chronic HBV infection or a healthy individual who was vaccinated for HBV (with recombinant HBV envelope vaccine) and tested within 24 hours after blood draw (FIG. 13B). 400 μl aliquots were separately mixed with 100 μl RPMI containing each of the HBV peptide pools (2 μg/ml final concentration per peptide) or a DMSO control and incubated for a period extending overnight, to allow activation of responsive T cells. A plasma fraction was isolated from each of the incubated samples and the level of cytokines in the sample measured using an Ella™ multi-analyte ELISA machine (ProteinSimple, CA, USA).


It would be understood that the parameters described in the example may be changed and still achieve a valid assay. For example, the ratio of blood to RMPI media can range from 100% blood to 50% blood; 100 μl aliquots or more than 400 μl might be used, but a larger blood sample would be required to test multiple peptide pools. RPMI may be exchanged with other cell culture media. The final concentration of peptides in an assay mixture may be from 1 to 5 μg/ml. The control sample may contain DMSO or may be an unstimulated blood sample. The incubation period may range between about 6-24 h.


HBV reactive T cells in infected individuals can be detected by quantifying the amount of secreted cytokines after the direct addition of the peptide pools into whole blood. At the moment, we have selected the detection of both IFN-γ and IL-2, two cytokines commonly secreted by T cells, as a means of detection.


From the data (FIG. 13B), we observed that T cells specific for the HBV envelope protein were readily detected in the chronic HBV patient and the vaccinated individual. The secretion of IL-2 appears to be more sensitive than IFN-γ to detect the HBV-specific T cells. In conclusion, we demonstrate that the whole blood assay can be readily applied for the detection of T cells specific for other viruses by utilising overlapping peptides specific for the virus of interest.


SUMMARY

The assays presented here are based on the ability of SARS-CoV-2 T cells to respond to different peptides covering different proteins of the virus. With the possibility to use different peptides pools, our approach represents a flexible strategy that can be easily utilized to detect the presence of T cells responding to emerging mutant strains and, thus, immediately gauge the impact that viral mutation might have on cellular immunity. Moreover, the methods exemplified herein are applicable to viruses other than SARS-CoV-2 and its variants.


Hence, a diagnostic method that can be easily adapted to detect the degree of cellular immunity is an urgently needed complement to the currently available tests measuring viral presence or antibody titers.


REFERENCES



  • 1. Braun, J., Loyal, L., Frentsch, M., Wendisch, D., Georg, P., Kurth, F., Hippenstiel, S., Dingeldey, M., Kruse, B., Fauchere, F., Baysal, E., Mangold, M., Henze, L., Lauster, R., Mall, M., Beyer, K., Roehmel, J., Schmitz, J., Miltenyi, S., Mueller, M. A., Witzenrath, M., Suttorp, N., Kern, F., Reimer, U., Wenschuh, H., Drosten, C., Corman, V. M., Giesecke-Thiel, C., Sander, L.-E., Thiel, A., 2020. Presence of SARS-CoV-2 reactive T cells in COVID-19 patients and healthy donors. medRxiv 1-12. doi:10.1101/2020.04.17.20061440.

  • 2. Camara C., et al., Differential effects of the second SARS-CoV-2 mRNA vaccine dose on T cell immunity in naïve and COVID-19 recovered individuals. bioRxiv 2021.03.22.436441; doi: https://doi.org/10.1101/2021.03.22.436441.

  • 3. Dong, T., Peng, Y., Mentzer, A. J., Liu, G., Yao, X., Yin, Z., Dong, D., Dejnirattisai, W., Turtle, L., Rostron, T., Subramaniam, K., Thomson, P., Zhang, P., Dold, C., Ratcliff, J., de Silva, T., Sopp, P., Wellington, D., Rajapaksa, U., Paes, W., Borrow, P., Kessler, B. M., Fry, J. W., Schwabe, N. F., Semple, M. G., Baillie, J. K., Openshaw, P. J., Cornall, R. J., Conlon, C., Screaton, G., Klenerman, P., Mongkolsapaya, J., McMichael, A., Knight, J. C., Ogg, G., Simmonds, P., Lockett, T., Levin, R., Moore, S. C., Salio, M., Napolitani, G., Chen, Y.-L., Dunachie, S., Supasa, P., Liu, C., Lopez-Camacho, C., Slon-Campos, J., Zhao, Y., Stuart, D. I., Paeson, G., Grimes, J., Antson, F., Bayfield, O. W., Hawkins, D. E., Ker, D.-S., Ansari, A., Barnes, E., Frater, J., Kerr, G., Goulder, P., 2020. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent COVID-19 patients. bioRxiv 1-36. doi:10.1101/2020.06.05.134551.

  • 4. Grifoni A, Sidney J, Zhang Y, Scheuermann R H, Peters B, Sette A. A Sequence Homology and Bioinformatic Approach Can Predict Candidate Targets for Immune Responses to SARS-CoV-2. Cell Host Microbe 2020; 27(4): 671-680.e2. doi:10.1016/j.chom.2020.03.002.

  • 5. Grifoni, A., n.d. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. doi:10.1016/j.cell.2020.05.015.

  • 6. Ichikawa, A. et al. CXCL10-CXCR3 enhances the development of neutrophil-mediated fulminant lung injury of viral and nonviral origin. Am J Respir Crit Care Med 187, 65-77 (2013).

  • 7. Kalimuddin S, Tham C Y L, Qui M, de Alwis R, Sim J X Y, Lim J M E, Tan H C, Syenina A, Zhang S L, Le Bert N, Tan A T, Leong Y S, Yee J X, Ong E Z, Ooi E E, Bertoletti A, Low J G. Early T cell and binding antibody responses are associated with COVID-19 RNA vaccine efficacy onset. Med (N Y). 2021 Jun. 11; 2(6):682-688.e4. doi: 10.1016/j.medj.2021.04.003.

  • 8. Le Bert N, Tan A T, Kunasegaran K, et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020; 584(7821): 457-62.

  • 9. Le Bert, N. et al. Highly functional virus-specific cellular immune response in asymptomatic SARS-CoV-2 infection. J Exp Med 218(5):e20202617 (2021).

  • 10. Long, Q.-X., Liu, B.-Z., Deng, H.-J., Wu, G.-C., Deng, K., Chen, Y.-K., Liao, P., Qiu, J.-F., Lin, Y., Cai, X.-F., Wang, D.-Q., Hu, Y., Ren, J.-H., Tang, N., Xu, Y.-Y., Yu, L.-H., Mo, Z., Gong, F., Zhang, X.-L., Tian, W.-G., Hu, L., Zhang, X.-X., Xiang, J.-L., Du, H.-X., Liu, H.-W., Lang, C.-H., Luo, X.-H., Wu, S.-B., Cui, X.-P., Zhou, Z., Zhu, M.-M., Wang, J., Xue, C.-J., Li, X.-F., Wang, L., Li, Z.-J., Wang, K., Niu, C.-C., Yang, Q.-J., Tang, X.-J., Zhang, Y., Liu, X.-M., Li, J.-J., Zhang, D.-C., Zhang, F., Liu, P., Yuan, J., Li, Q., Hu, J.-L., Chen, J., Huang, A.-L., 2020. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med 1-15. doi:10.1038/s41591-020-0897-1.

  • 11. Luster, A. D., Unkeless, J. C. & Ravetch, J. V. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature 315, 672-676 (1985).

  • 12. Tauzin, A. et al., A single BNT162b2 mRNA dose elicits antibodies with Fc-mediated effector functions and boost pre-existing humoral and T cell responses. bioRxiv. 2021 Mar. 18; 2021.03.18.435972. doi: 10.1101/2021.03.18.435972.

  • 13. Zhao, J., Alshukairi, A. N., Baharoon, S. A., Ahmed, W. A., Bokhari, A. A., Nehdi, A. M., Layqah, L. A., Alghamdi, M. G., Gethamy, Al, M. M., Dada, A. M., Khalid, I., Boujelal, M., Johani, Al, S. M., Vogel, L., Subbarao, K., Mangalam, A., Wu, C., Eyck, Ten, P., Perlman, S., Zhao, J., 2017. Recovery from the Middle East respiratory syndrome is associated with antibody and T-cell responses. Science Immunology 2, eaan5393. doi:10.1126/sciimmunol.aan5393.


Claims
  • 1. An in vitro method of discriminating past or currently virus-infected subjects from virus un-infected subjects, comprising: assaying a sample comprising whole blood, or comprising or derived from bronchoalveolar lavage (BAL) fluid, nasal swabs, or nasopharyngeal aspirate from a subject to determine whether it comprises T cells reactive to one or more virus peptide pools, wherein said peptide pools are separately derived from virus antigenic structural and non-structural proteins, wherein;(a) if the sample T cells are reactive to a majority of the peptide pools derived from the virus antigenic proteins, in comparison to unstimulated or DMSO treated cells, the subject is identified as past or currently infected by the virus, or(b) if the sample T cells are reactive to 0, or a minority of the peptide pools derived from the virus antigenic proteins, in comparison to unstimulated or DMSO treated cells,the subject is identified as having been uninfected by the virus.
  • 2. The method of claim 1, wherein the virus is: a) a coronavirus, selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, HKU1, OC43, NL63 and 229E or variant thereof; orb) hepatitis B virus (HBV) or variant thereof.
  • 3. The method of claim 2, wherein: a) the virus antigenic structural and non-structural proteins are membrane (M), nucleoprotein (NP) and/or Spike (S) proteins; orb) the virus antigenic structural and non-structural proteins are Polymerase (Pol), Envelope (E), Core (C) and/or X proteins.
  • 4. The method of claim 3, wherein the virus is a) SARS-CoV-2 or b) HBV; and ai) an M peptide pool comprises or consists of at least one peptide derived from an M protein comprising the amino acid sequence set forth in SEQ ID NO: 793;ii) an NP peptide pool comprises or consists of at least one peptide derived from an NP protein comprising the amino acid sequence set forth in SEQ ID NO: 794; andiii) an S peptide pool comprises or consists of at least one peptide derived from an S protein comprising the amino acid sequence set forth in SEQ ID NO: 795; orbi) a Pol peptide pool comprises or consists of at least one peptide derived from an Pol protein comprising the amino acid sequence set forth in SEQ ID NO: 796;ii) an E peptide pool comprises or consists of at least one peptide derived from an E protein comprising the amino acid sequence set forth in SEQ ID NO: 797;iii) a C peptide pool comprises or consists of at least one peptide derived from a C protein comprising the amino acid sequence set forth in SEQ ID NO: 798; andiv) an X peptide pool comprises or consists of at least one peptide derived from an X protein comprising the amino acid sequence set forth in SEQ ID NO: 799.
  • 5. The method of claim 4, wherein; ai) an M peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 1-43,ii) an NP peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 44-125, andiii) an S peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 126-180; orbi) a Pol peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 626-792,ii) an E peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 550-625;iii) a C peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 480-520; andiv) an X peptide pool comprises or consists of at least one peptide selected from peptides having the amino acid sequences set forth in SEQ ID Nos: 521-549.
  • 6. The method of claim 5, wherein the NP peptide pool is divided into 2 pools, NP1 and NP2; or wherein the Pol peptide pool and/or the E peptide pool is/are divided into a plurality of pools.
  • 7. The method of claim 6, wherein; (a) if the sample T cells are reactive to 3 or 4 of the peptide pools derived from M, NP1, NP2 and S, in comparison to unstimulated or DMSO treated cells, the subject is identified as past or currently infected by SARS-CoV-2,(b) if the sample T cells are reactive to 0, 1 or 2 of the peptide pools derived from M, NP1, NP2 and S, in comparison to unstimulated or DMSO treated cells, the subject is identified as having been uninfected by SARS-CoV-2.
  • 8. The method of claim 1, comprising the steps of: a) mixing the sample with each of said peptide pools to produce: i) assay samples corresponding to M, NP and S; orii) E, Pol, C and X;b) incubating each mixture for a period to allow for T cell activation;c) measuring the level of at least one secreted cytokine in each said mixture and determining whether the level of at least one secreted cytokine is above a threshold control value to indicate a positive T-cell reaction; andd) counting the number of peptide pools that are positive.
  • 9. The method of claim 6, comprising the steps of: a) mixing the sample with each of said peptide pools to produce: i) 4 assay samples corresponding to M, NP1, NP2 and S; orii) 8 assay samples corresponding to C, Pol1, Pol2, Pol3, Pol4, E1, E2 and X;b) incubating each mixture for a period to allow for T cell activation;c) measuring the level of at least one secreted cytokine in each said mixture and determining whether the level of at least one secreted cytokine is above a threshold control value to indicate a positive T-cell reaction; andd) counting the number of peptide pools that are positive.
  • 10. The method of claim 1, wherein if the sample T cells are reactive to 50% or more of the coronavirus peptide pools, in comparison to unstimulated or DMSO-treated cells, the subject is identified as past or currently infected by coronavirus.
  • 11. An in vitro method of determining whether a vaccine or previously virus-infected subject has T cells whose activation may be reduced by a virus variant, such as a variant of concern (VOC), comprising: assaying a sample comprising whole blood, or comprising or derived from bronchoalveolar lavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject to determine whether it comprises T cells reactive to one or more virus peptide pools, wherein said peptide pools are separately derived from (A) the whole virus antigenic protein present in the vaccine or corresponding to an antigenic protein from the virus that infected the subject, (B) non-conserved regions of said virus antigenic protein that are mutated in the virus variant, and (C) virus variant mutated non-conserved regions of the vaccine antigenic protein or corresponding to an antigenic protein from the virus that infected the subject, wherein;the number or proportion of reactive T cells present in each pool is analyzed and utilized to derive in each single individual, the frequency of T cells directed towards the whole virus antigenic protein (PBMC stimulated with peptide pool A), the frequency of T cells directed toward non-conserved regions of said virus antigenic protein that are mutated in the virus variant (PBMC stimulated with Pool B) and the frequency of T cells inhibited by amino acid mutations present in virus variant mutated non-conserved regions (PBMC stimulated with pool C), wherein;(a) if the sample T cells are reactive to peptide pool A, the subject has T cells responsive against the virus antigenic protein, and(b) if the sample T cells are similarly reactive to pool B and pool C, the impact of the amino acid mutations in the variant are negligible on the total T cell response against the said virus antigenic protein;(c) if the sample T cells reacts differently to pool B and pool C, the impact of the amino acid mutations in the variant on the total T cell response against the said virus antigenic protein can be estimated by the proportion of pool C against pool B response,wherein the method provides an estimation of the ability of T cells of the subject to recognize the conserved and non-conserved region of different vaccine antigenic proteins or virus that infected the subject, and of the ability of mutations to reduce the T cell response towards variants.
  • 12. The method of claim 11, wherein: i) the virus is a coronavirus, selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, KHU1, OC43, NL63 and 229E; orii) the virus is HBV or variants thereof; and/oriii) the virus antigenic protein is an M, NP, or S protein; or an E, Pol, C or X protein; and/oriv) peptide pool A and pool B are derived from a wild-type virus.
  • 13.-14. (canceled)
  • 15. The method of claim 12 or, wherein iii) the virus antigenic protein is an M, NP, or S protein; or an E, Pol, C or X protein; and/or iv) peptide pool A and pool B are derived from a wild-type virus; and wherein:ai) the M protein comprises the amino acid sequence set forth in SEQ ID NO: 793;ii) the NP protein comprises the amino acid sequence set forth in SEQ ID NO: 794; andiii) the S protein comprises the amino acid sequence set forth in SEQ ID NO: 795; orbi) the Pol protein comprises the amino acid sequence set forth in SEQ ID NO: 796;ii) the E protein comprises the amino acid sequence set forth in SEQ ID NO: 797;iii) the C peptide protein comprises the amino acid sequence set forth in SEQ ID NO: 798; andiv) the X protein comprises the amino acid sequence set forth in SEQ ID NO: 799.
  • 16. The method of claim 12, wherein iv) peptide pool A and pool B are derived from a wild-type virus; and wherein the wildtype virus is SARS-CoV-2 wildtype and the variant is selected from the group comprising B.1.617.2 (Delta), B.1.1.7 (Alpha V1), B.1.351 (Beta V2), P.1 (Gamma, V3), B.1.617.1 (Kappa), (P2), B.1.427/9 (Epsilon), B.1.525 (Eta), B.1.526 (Iota), C.37 (Lambda), B.1.621 and B.1.620; or the virus is HBV C.
  • 17. The method of claim 11, comprising the steps of: a) mixing the sample with each of said peptide pools A, B, and C to produce assay samples;b) incubating each mixture formed for a period to allow T cell activation;c) measuring the level of at least one secreted cytokine in each said mixture and determining whether the level of at least one secreted cytokine is above a threshold control value to indicate a positive T-cell reaction; andd) determining the number or proportion of reactive T cells present in each pool.
  • 18. The method of any one of claim 8, wherein the secreted cytokine is selected from the group comprising IFN-gamma, IL-2, CXCL9, CXCL10, TNF-alpha, IL-6, IL-10 and IL-1.
  • 19.-20. (canceled)
  • 21. The method of claim 1, wherein: a) whole blood is mixed with each of said peptide pools;b)i) each mixture is incubated for at least 6 h;b)ii) a plasma fraction of the mixture is isolated;c) the level of at least one secreted cytokine in each said plasma fraction is measured and compared to a threshold control value to indicate a positive or negative T cell reaction.
  • 22. A method to quantify the presence of virus-specific T cells in a biological sample comprising or derived from blood, broncholavage (BAL fluid), nasal swabs, or nasopharyngeal aspirate from a subject, comprising; a) Mixing the biological sample with one or more virus peptide pools, wherein said peptide pools are separately derived from virus antigenic structural or non-structural proteins;b) incubating the mixture formed for a period to allow T cell activation;c) Rupture the cells from b);d) Aliquot a sample from c) into PCR reagents, ACTIN (or other internal control) forward and reverse primers, ACTIN (or other internal control) probe, CXCL10 forward and reverse primers and CXCL10 probe for dqPCR; and/ore) extract RNA from a sample from c) and add a portion into PCR reagents, ACTIN (or other internal control) forward and reverse primers, ACTIN (or other internal control) probe, CXCL10 forward and reverse primers and CXCL10 probe for qPCR;f) perform cycles of dqPCR and/or qPCR for d) and e), respectively; andg) quantitate the expression of CXCL10 in the sample and compare with a control,wherein an elevated CXCL10 level indicates the presence of virus-specific T cells in the subject sample.
  • 23. The method of claim 22, wherein the virus is a coronavirus, selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, KHU1, OC43, NL63 and 229E or variant mutants thereof; or HBV or variant mutant thereof: and/or wherein: ai) the M protein comprises the amino acid sequence set forth in SEQ ID NO: 793;ii) the NP protein comprises the amino acid sequence set forth in SEQ ID NO: 794; andiii) the S protein comprises the amino acid sequence set forth in SEQ ID NO: 795; orbi) the Pol protein comprises the amino acid sequence set forth in SEQ ID NO: 796;ii) the E protein comprises the amino acid sequence set forth in SEQ ID NO: 797;iii) the C protein comprises the amino acid sequence set forth in SEQ ID NO: 798; andiv) the X protein comprises the amino acid sequence set forth in SEQ ID NO: 799;and/orwherein the peptide pools comprise one or more M, NP and S peptides listed in Tables 1-4 and 7-19; or one or more Pol, E, C and X peptides listed in Tables 20-27.
  • 24.-25. (canceled)
  • 26. A method of treatment or prophylaxis, comprising administering, respectively, to a subject with T cells reactive to: i) a majority of peptide pools derived from virus antigenic proteins, an effective amount of a virus inhibitor; orii) 0, or a minority of the peptide pools M, NP and S; or E, Pol, C and X, an effective amount of a coronavirus or non-enveloped virus vaccine, respectively.
  • 27. The method of claim 26, wherein the virus is a coronavirus such as a coronavirus selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, HKU1, OC43, NL63 and 229E or variants thereof; or wherein the virus is HBV or variant thereof.
  • 28. The method of claim 27, wherein the virus is SARS-CoV-2, or HBV.
  • 29. The method of claim 28, wherein the peptide pools comprise M, NP1, NP2 and S pools listed in Tables 1-4; or E1, E2, Pol1, Pol2, Pol3, Pol4, C and X pools listed in Tables 20-27.
  • 30. A method of monitoring the efficacy of a virus vaccine, comprising testing whether the recipient of said vaccine has T cells reactive to 0, a minority, 50%, or a majority of peptide pools derived from the virus M, NP and S proteins; or virus E, Pol, C and X proteins.
  • 31. The method of claim 30, wherein the virus is a coronavirus such as a coronavirus selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, KHU1, OC43, NL63 and 229E or variants thereof; or HBV or a variant thereof.
  • 32. The method of claim 31, wherein the virus is SARS-CoV-2 or HBV.
  • 33. The method of claim 32, comprising testing whether the recipient of said vaccine has T cells reactive to 0, 1, 2, 3 or 4 of the peptide pools M, NP1, NP2 and S listed in Tables 1-4; or T cells reactive to 0, 1, 2, 3, 4, 5, 6 or 7 of the peptide pools E1, E2, Pol1, Pol2, Pol3, Pol4, C and X.
  • 34. A kit to discriminate past or currently virus-infected subjects from virus un-infected subjects, the kit comprising a plurality of virus structural or non-structural peptides that stimulate virus-exposed T cells, wherein the virus peptides are in peptide pools derived from virus M, NP and S proteins; or from virus E, Pol, C and X proteins.
  • 35. The kit of claim 34, wherein the virus is a coronavirus such as a coronavirus selected from the group comprising MERS-CoV, SARS-CoV, SARS-CoV-2, KHU1, OC43, NL63 and 229E or variants thereof; or HBV or variant thereof.
  • 36. The kit of claim 35, wherein the virus is SARS-CoV-2 or HBV.
  • 37. The kit of claim 36, wherein: ai) the M protein comprises the amino acid sequence set forth in SEQ ID NO: 793;ii) the NP protein comprises the amino acid sequence set forth in SEQ ID NO: 794; andiii) the S protein comprises the amino acid sequence set forth in SEQ ID NO: 795; orbi) the Pol protein comprises the amino acid sequence set forth in SEQ ID NO: 796;ii) the E protein comprises the amino acid sequence set forth in SEQ ID NO: 797;iii) the C protein comprises the amino acid sequence set forth in SEQ ID NO: 798; andiv) the X protein comprises the amino acid sequence set forth in SEQ ID NO: 799.
  • 38. The kit of claim 37, wherein: the M peptide pool comprises peptides having amino acid sequences set forth in SEQ ID Nos: 1-43;the NP peptide pool comprises peptides having amino acid sequences set forth in SEQ ID Nos: 44-125; andthe S peptide pool comprises peptides selected from peptides having amino acid sequences set forth in SEQ ID Nos: 126-454.
  • 39. The kit of claim 34, further comprising one or more reagents to detect cytokines and/or chemokines secreted from activated T cells.
  • 40. A kit to quantify SARS-CoV-2-specific T cell activation, or HBV-specific T cell activation, in an isolated patient sample, comprising: i) one or more peptide pools selected from the pools in Tables 1-4 and 7-19, or Tables 20-27, respectively;ii) PCR reagents and/or primers and probes to detect CXCL10 and/or IFN-gamma expression by stimulated T cells; and/oriii) ELISPOT reagents.
  • 41. The kit of claim 40, wherein peptide pools from Tables 7-19 are used to analyse the effect of virus variants, including variants of concern (VOC), on T cell activation in vaccinated or previously virus-infected subjects.
  • 42. A set of at least 2 separate pools of peptides suitable to discriminate past or currently SARS-CoV-2-infected subjects or vaccinated subjects from SARS-CoV-2 un-infected subjects, wherein the peptide pools are selected from those listed in Tables 1 to 4 and 7-19, or Tables 20-27.
Priority Claims (1)
Number Date Country Kind
10202010411P Oct 2020 SG national
PCT Information
Filing Document Filing Date Country Kind
PCT/SG2021/050627 10/18/2021 WO